CHIMERIC RECEPTOR THERAPY

Information

  • Patent Application
  • 20220220187
  • Publication Number
    20220220187
  • Date Filed
    January 10, 2022
    2 years ago
  • Date Published
    July 14, 2022
    2 years ago
Abstract
A non-naturally occurring polynucleotide encoding a miRNA that inhibits the expression of an immune checkpoint protein. The polynucleotide may further encode a chimeric receptor, a cytokine, and/or a cell tag. A vector comprising the aforementioned polynucleotide. A modified immune effector cell comprising the aforementioned polynucleotide. Compositions and kits comprising the aforementioned polynucleotide and/or cell. A method for treating a subject suffering from a disease or disorder, comprising administering the aforementioned cell to a subject in need thereof. The use of the aforementioned cell in the manufacture of a medicament for the treatment of a disease or disorder. A method for the detection of a disease or disorder associated with the overexpression of an antigen in a subject. A method for the treatment of a disease or disorder comprising the serial administration of polynucleotides encoding a chimeric antigen receptor or a cell comprising the same.
Description
REFERENCE TO SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jan. 10, 2022, is named 75594-348568_SL.txt and is 579,627 bytes in size.


BACKGROUND OF THE DISCLOSURE

Chimeric antigen receptor (CAR-T) cell and T cell receptor (TCR) therapies have recently undergone rapid development and have been shown to successfully direct killing of tumor cells. Such therapies are, for example, useful in treating autoimmune disorders and cancers. Indeed, several targets for such therapies have been identified to date, including but not limited to CD19, CD33, BCMA, CD44, α-Folate receptor, CAIX, CD30, ROR1, CEA, EGP-2, EGP-40, HER2, HER3, Folate-binding Protein, GD2, GD3, IL-13R-a2, KDR, EDB-F, mesothelin, CD22, EGFR, Folate receptor α, Mucins such as MUC1, MUC4 or MUC16, MAGE-A1, h5T4, PSMA, TAG-72, EGFR, CD20, EGFRvIII, CD123 or VEGF-R2. Among these, CD19, CD33, MUC1, MUC16, and ROR1 have shown particular promise as targets for immunotherapy.


CD19 is an attractive target for immunotherapy due to several factors. It is expressed on a variety of B cell lymphomas and leukemias and on normal B cells, but it is not found on hematopoietic stem cells, plasma cells, and other healthy tissues. In addition, CD19 has a broader expression profile than that of CD20, which is the target of monoclonal antibody therapies such as rituximab, and it is thought to be a better target for antibody-drug conjugates (ADC) compared with CD20, which suffers from inefficient internalization. CD19 has also been shown to be expressed in cases where monoclonal antibody treatment (e.g., rituximab) is ineffective due to CD20 downregulation or other factors. Additionally, because CD19-targeting agents have a mode of action that is distinct from that of anti-CD20 antibodies, they could complement existing monoclonal antibody regimens.


CD33 is an attractive target for immunotherapy due to its high expression in cancer and minimal expression in healthy adult tissues. CD33 is overexpressed on myeloid leukemia and leukemic stem cells. CD33 is overexpressed in acute myeloid leukemia (AML), which is the most common acute leukemia in adults. 85 to 90% of AML patients show expression of CD33 on blast cells. CD33 is also overexpressed in myelodysplastic syndromes (MDS), which are cancerous conditions of the bone marrow generally found in adults in their 70s.


MUC1 is an attractive target for immunotherapy because it is overexpressed in breast cancer, and is absent or expressed at low levels in normal mammary glands. In addition, MUC1 is mostly aberrantly underglycosylated in cancer and the antigens on the cancer surface are different from those on normal cells. Therefore targeting MUC1 for cancer immunotherapy can exploit the differences between cancerous and normal cells.


MUC16 is an attractive target for immunotherapy due to its high expression in cancer and minimal expression in healthy adult tissues. MUC16 is aberrantly expressed in ovarian cancer, breast cancer, pancreatic cancer, endothermal cancer, and lung cancer. For example, MUC16 is overexpressed in over 80% of ovarian tumors, which is the most lethal of the gynecologic malignancies. Meanwhile, limited expression of MUC16 has been found on healthy tissue. The current standard of care for ovarian cancer is surgery, followed by chemotherapy with a combination of platinum agents and taxanes. However, recurrence of the disease occurs in most patients after initial treatment, resulting in a cycle of repeated surgeries and additional rounds of chemotherapy.


Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an attractive target for immunotherapy due to its high expression in cancer and minimal expression in healthy adult tissues. ROR1 is aberrantly expressed in multiple hematological tumors, including chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), acute lymphoblastic leukemia (ALL), and diffuse B-cell lymphoma (DLBCL) and solid tumors, including breast adenocarcinomas encompassing triple negative breast cancer (TNBC), pancreatic cancer, ovarian cancer, and lung adenocarcinoma.


Although many patients have durable responses with CAR-T and TCR therapies, for some patients the anti-tumor effects of such therapies are either short-lived or ineffective. Another immunotherapy that has shown promise is immune checkpoint inhibition, which can prevent the switching off of T cells and promote the activity of these cells. Examples of checkpoint inhibitor targets include but are not limited to PD1, PD-L1, CTLA-4, TIGIT, 4-1BB, PIK3IP1, CD27, CD28, CD40, CD70, CD122, CD137, OX40 (CD134), GITR, ICOS, A2AR, B7-H3 (CD276), B7-H4 (VTCN1), BTLA, IDO, KIR, LAG3, TIM-3, or VISTA. Among these, targeting CTLA4, PD-1, PD-L1, TIM3, TIGIT, LAG3, and/or PIK3IP1 has shown the most promise. One of the most studied checkpoint inhibition pathway is the PD-1/programmed death ligand 1 (PD-L1) pathway, which plays a vital role in how tumor cells evade immune response. Immunotherapy utilizing PD-1/PD-L1 blocking antibodies has been extensively evaluated in the clinic and has been shown to improve tumor regression across multiple malignancies, especially when administered in conjunction with CAR-T cells. However, checkpoint inhibitor blocking antibodies have not performed consistently across cancer types, may have limited access to the tumor microenvironment, require repeated administration, and may lose effectiveness over time. Genome editing is an alternate approach to eliminate PD-1 mediated CAR-T cell exhaustion, and has the advantage of restricting the PD-1 blockade to only the engineered CAR-T cells. However, gene editing adds complexity to the manufacturing process, which increases the turnaround time and cost of the cell therapy.


There is accordingly a continuing need in the art to obtain safer, more effective, less expensive therapies to antigen-associated diseases and conditions, including treatments that combine CAR-T and/or TCR therapy with systemic checkpoint inhibition.


There is also a need to devise ways of diversifying treatment regimens to provide a multi-pronged targeting of antigens in order to address complex in vivo biological issues such as loss of immunological surveillance, genetic alterations in tumor antigen composition and tumor heterogeneity (giving rise to cancer cell phenotypic differences).


SUMMARY OF THE DISCLOSURE

The present invention relates in part to a non-naturally occurring polynucleotide encoding a miRNA that inhibits the expression of an immune checkpoint protein. In certain embodiments, the miRNA targets CTLA4, PD-1, PD-L1, TIM3, TIGIT, LAG3, GITR, or PIK3IP1. In certain embodiments, the miRNA targets PD-1.


In certain embodiments, the polynucleotide comprises a nucleic acid sequence having at least 80% sequence identity with any one of SEQ ID NOs: 72-87 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 72-87. In certain such embodiments, the polynucleotide comprises a nucleic acid sequence having at least 80% sequence identity with any one of SEQ ID NOs: 72, 74, 76, 78, 80, 82, 84, and 86 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 72, 74, 76, 78, 80, 82, 704, 705, 709, and 710. In certain embodiments, the polynucleotide comprises a nucleic acid sequence having at least 80% sequence identity with SEQ ID NO: 179 or 180 or that is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 179 or 180. In certain embodiments, the polynucleotide comprises a nucleic acid sequence having at least 80% sequence identity with SEQ ID NO: 267 or that is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 267.


In certain embodiments, the polynucleotide further comprises: a) a nucleic acid sequence having at least 80% sequence identity with SEQ ID NO: 292 or is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 291; and b) a nucleic acid sequence having at least 80% sequence identity with SEQ ID NO: 292 or is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 292.


In certain embodiments, the polynucleotide further encodes a chimeric receptor. In certain embodiments, the chimeric receptor is a T-cell receptor or a chimeric antigen receptor.


In certain embodiments, the chimeric antigen receptor comprises an antigen-binding domain that binds to an epitope on CD19, CD33, MUC1, MUC16, or ROR1. In certain embodiments, the antigen-binding domain binds to an epitope on ROR1.


In certain embodiments, the antigen-binding domain comprises a variable light chain domain comprising the amino acid sequence of any one of SEQ ID NOs: 347, 351, 355, 359, 363, 367, 371, 375, 379, 383, 387, 391, 395, 399, 403, 407, 411, 415, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, and 463, or a functional fragment or variant thereof. In certain such embodiments, the variable light chain domain comprises the amino acid sequence of SEQ ID NO: 387 or a functional fragment or variant thereof.


In certain embodiments, the antigen-binding domain comprises a variable heavy chain domain comprising the amino acid sequence of any one of SEQ ID NOs: 349, 353, 357, 361, 365, 369, 373, 377, 381, 385, 389, 393, 397, 401, 405, 409, 413, 417, 421, 425, 429, 433, 437, 441, 445, 449, 453, 457, and 461, or a functional fragment or variant thereof. In certain such embodiments, the variable heavy chain domain comprises the amino acid sequence of SEQ ID NO: 349 or a functional fragment or variant thereof.


In certain embodiments, the chimeric antigen receptor comprises a spacer. In certain such embodiments, the spacer comprises a stalk region that is a CD8α hinge domain or a functional fragment or variant thereof. In certain such embodiments, the stalk region comprises the amino acid sequence of SEQ ID NO: 467 or a functional fragment or variant thereof. In certain embodiments, the spacer comprises a stalk extension region. In certain such embodiments, the stalk extension region comprises the amino acid sequence of SEQ ID NO: 473 or a functional fragment or variant thereof.


In certain embodiments, the chimeric antigen receptor further comprises a transmembrane domain. In certain such embodiments, the transmembrane domain comprises a CD8α transmembrane domain or a functional fragment or variant thereof. In certain such embodiments, the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 475 or a functional fragment or variant thereof.


In certain embodiments, the chimeric antigen receptor further comprises an intracellular signaling domain. In certain such embodiments, the intracellular signaling domain comprises a CD3ζ signaling domain or a functional fragment or variant thereof. In certain such embodiments, the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 479 or a functional fragment or variant thereof.


In certain embodiments, the intracellular signaling domain comprises a co-stimulatory domain. In certain such embodiments, the intracellular signaling domain comprises a CD28 signaling domain or a functional fragment or variant thereof. In certain embodiments, the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 481 or a functional fragment or variant thereof.


In certain embodiments, the polynucleotide further encodes a cytokine. In certain embodiments, the cytokine is IL-15 or a functional fragment or variant thereof. In certain such embodiments, the cytokine comprises the amino acid sequence of SEQ ID NO: 519 or a functional fragment or variant thereof.


In certain embodiments, the IL-15, or functional fragment or variant thereof, is membrane bound. In certain such embodiments, the IL-15, or functional fragment or variant thereof, forms part of a fusion protein that also comprises IL-15Rα, or a functional fragment or variant thereof. In certain such embodiments, the fusion protein comprises the amino acid sequence of SEQ ID NO: 523 or a functional fragment or variant thereof. In certain embodiments, the fusion protein comprises the amino acid sequence of SEQ ID NO: 525 or a functional fragment or variant thereof.


In certain embodiments, the polynucleotide further encodes a cell tag. In certain embodiments, the cell tag comprises a truncated HER1, or a functional fragment or variant thereof. In certain embodiments, the truncated HER1 comprises a HER1 Domain III, or a functional fragment or variant thereof, and a truncated HER1 Domain IV, or a functional fragment or variant thereof. In certain such embodiments, the truncated HER1 comprises the amino acid sequence of SEQ ID NO: 565, or a functional fragment or variant thereof, and the amino acid sequence of SEQ ID NO: 567, or a functional fragment or variant thereof.


In certain embodiments, the cell tag further comprises a CD28 transmembrane domain or a functional fragment or variant thereof. In certain such embodiments, the cell tag comprises the amino acid sequence of SEQ ID NO: 571 or a functional fragment or variant thereof.


The present invention also relates to a vector comprising the aforementioned polynucleotide. The vector may be viral or non-viral. In certain embodiments, the vector comprises a Sleeping Beauty transposon.


The present invention further relates to a modified immune effector cell comprising the aforementioned polynucleotide. In certain embodiments, the cell is a T-cell.


The present invention additionally relates to compositions and kits comprising the aforementioned polynucleotide and/or cell.


A further aspect of the present invention is a method for treating a subject suffering from a disease or disorder, comprising administering the aforementioned cell to a subject in need thereof. The invention also relates to the use of the aforementioned cell in the manufacture of a medicament for the treatment of a disease or disorder. The disease or disorder may be one associated with the overexpression of an antigen, for example, CD19, CD33, ROR1, MUC1, or MUC16. In certain embodiments, the disease or disorder is one associated with the overexpression of ROR1.


In certain embodiments, the disease or disorder is cancer. In certain embodiments, the disease or disorder is chronic lymphocytic leukemia, mantle cell lymphoma, acute lymphoblastic leukemia, or diffuse large B-cell lymphoma, breast adenocarcinomas encompassing triple negative breast cancer, pancreatic cancer, ovarian cancer, or lung adenocarcinoma.


Another aspect of the present invention is a method for the detection of a disease or disorder associated with the overexpression of an antigen in a subject, the method comprising: a) contacting a sample from the subject with one or more of the antibodies, or antigen-binding fragments thereof; and b) detecting an increased level of binding of the antibody or fragment thereof to the sample as compared to such binding to a control sample lacking the disease, thereby detecting the disease in the subject.


A yet further aspect of the present invention is a method for the treatment of a disease or disorder, such as cancer and auto-immune disease or disorders, comprising the serial administration of cells, nucleic acids, viral vectors, or non-viral vectors comprising polynucleotides encoding chimeric antigen receptors selected from a collection of chimeric antigen receptors having different structural compositions and binding specificities for an array of antigen targets. In certain such embodiments, the method comprises a first administration of cells expressing one or more chimeric antigen receptors from the collection followed by a second administration of cells expressing one or more chimeric antigen receptors from the collection, wherein a period of time elapses between the first and second administrations.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A is an exemplary depiction of a vector comprising a combination of one or more checkpoint inhibitor miRNAs with a chimeric receptor (SD=Splice Donor; SA=Splice Acceptor).



FIG. 1B is an exemplary depiction of hairpin and loop design of various pri-miRNAs including target miRNA and complementary sequences at various 5′ or 3′ positions. FIG. 1C is exemplary depiction of hairpin and loop design of various pri-miRNAs including its position in a transgene cassette.



FIG. 2 is a graph depicting PD1 relative RNA expression following transfection of various combinations of miRNA constructs in the presence or absence of MUC16-specific CAR. Constructs #1-8 as depicted on the X-axis are as schematically presented in Table 10.



FIGS. 3A, 3B and 3C are graphs depicting normalized absolute transcript counts obtained from gene analysis of >700 genes using a Nanostring human gene panel code set with CD3/CD28 bead-stimulated CD33 CAR-T cells. In FIG. 3A, the Y-axis plots the transcript counts from CAR-T cells containing an intron coding for 2 checkpoint inhibitor miRNAs targeting PD-1 and TIGIT (CD33 CAR-mbIL15-HER1t+ miRNA (PD-1+TIGIT)), and the X-axis plots the transcripts from CAR-T cells only (not containing any checkpoint inhibitory miRNA). The circles denote the genes of interest. In FIG. 3B, the Y-axis plots the transcript counts from PD-1 miRNA containing CAR-T cells (CD33 CAR-mbIL15-HER1t+miRNA (PD-1+PD-1)) and the X-axis plots the transcripts from CAR-T cells only. FIG. 3C plots the non-targeting miRNA control (CD33 CAR-mbIL15-HER1t+miRNA scrambled*) on the Y-axis and CAR-T cells without a miRNA containing intron on the X-axis. The circles denote the genes of interest and used to depict the on-target specificity of the checkpoint inhibitor miRNA designs. All three graphs were derived from one donor. * Scrambled controls are non-targeting miRNAs.



FIG. 4A-C are graphs depicting the normalized absolute transcript counts obtained from gene analysis of >700 genes using a Nanostring human gene panel code set with CD3/CD28 bead stimulated MUC16-specific CAR-T cells. In FIG. 4A, the Y-axis plots the transcript counts from CAR-T cells containing an intron coding for miRNAs targeting two different sequences within PD-1 and a sequence for TIGIT (MUC16CAR-mbIL15-HER1t (collectively also referred to as “MUC16CAR”)+miRNA (PD-1/PD-1/TIGIT)), and the X-axis plots the transcripts from CAR-T cells without a miRNA-containing intron (MUC16CAR-mbIL15-HER1t). The black circles denote the genes of interest. In FIG. 4B, the X-axis is the same, and the Y-axis plots the transcript counts from dual PD-1 targeting miRNA containing CAR-T cells (MUC16CAR-mbIL15-HER1t+miRNA (PD-1/PD-1)). FIG. 4C plots the non-targeting miRNA control (MUC16CAR-mbIL15-HER1t+miRNA (scrambled)) on the Y-axis and CAR-T cells without a miRNA-containing intron on the X-axis. All three graphs are from one donor.



FIG. 5A is a graph depicting the number of GFP+K562 cells expressing MUC16 over time. The line with black circle filled dots at each time point denotes number GFP+ target cells in wells without CAR-T cells. The line with square open points denotes target cell counts in wells with MUC16 CAR-mbIL15-HER1t CAR-T cells without a miRNA-containing intron, ((“with CAR-T cells”)). The line with grey circle filled points denotes the target cell counts in wells with CAR-T cells containing a synthetic intron with dual PD-1 targeting miRNAs (MUC16 CAR-mbIL15-HER1t+miRNA (PD-1/PD-1) (“with CAR-T+miRNA cells”). Data are from one donor, plotted is the mean+SD of triplicate wells. *** P<0.001 based on a 2-way ANOVA with Dunnett's Multiple Comparison post hoc test.



FIG. 5B is a graph depicting the number of GFP+K562/MUC16+/PD-L1+/CD155+ cells over time. The line with square filled points at each time point denotes number GFP+ target cells only in wells. The line with open circle points denotes target cell counts in wells with MUC16-specific CAR-T cells without a miRNA-containing intron (MUC16 CAR-mbIL15-HER1t (with CAR-T cells)). The line with open circle filled points denotes the target cell counts in wells with CAR-T cells containing a synthetic intron with dual PD-1 and a TIGIT targeting miRNAs (MUC16CAR-mbIL15-HER1t+miRNA (PD-1/PD-1/TIGIT)(with CAR-T+miRNA cells)). Data are from one donor, plotted is the mean+SD of triplicate wells.



FIG. 6A-B depicts cytokine expression levels of IFN gamma and GM-CSF in MUC16 CAR-T cells with a combination of one or more checkpoint inhibitor miRNAs following co-culture with tumor target cells (K562/MUC16t). FIG. 6C-D depicts cytokine expression levels of IFN gamma and GM-CSF in MUC16 CAR-T cells with a combination of one or more checkpoint inhibitor miRNAs without co-culture with tumor target cells. Constructs #1-11 as depicted on the X-axis are as schematically presented in Table 11.



FIG. 7 shows the tumor burden in mice treated with MUC16 CAR+mbIL-15+HER1t (shown as “MUC16 CAR”) in combination with various miRNAs.



FIG. 8A demonstrates PD-1 levels in cell populations following gating hCD45/CD3+/HER1t+ expression in the blood of MUC16CAR+mbIL15+HER1t (CAR only) and MUC16CAR+mbIL15+HER1t+miRNA (PD1/PD-1) (CAR+miRNA (PD-1/PD-1)) treated mice.



FIG. 8B shows PD-1 levels as measured by median fluorescent intensity (MFI) in CAR and CAR+miRNA (PD-1/PD-1) treated mice.



FIGS. 9A and 9B demonstrates PD-1 and TIGIT MFI levels in cell populations following gating hCD45/CD3+/HER1t+ expression in the blood of various CAR and CAR+miRNA treated mice. Groups #1-9 as depicted on the X-axis are as schematically presented in Table 12.



FIG. 10A demonstrates that the PD1 silencer module produces guide miRNAs and a corresponding decrease in PD1 mRNA expression in UltraCAR-T cells generated from 5 T cell donors. RT-qPCR results of PD1-targeting guide miRNAs are depicted.



FIG. 10B demonstrates that the PD1 silencer module produced guide miRNAs and a corresponding decrease in PD1 mRNA expression in ultraCAR-T cells generated from 5 T cell donors. RT-qPCR results of PD1 mRNA are depicted.



FIG. 11 demonstrates that the PD1 silencing module preferentially produced PD1-targeting guide miRNAs over non-targeting passenger miRNAs.



FIGS. 12A-E demonstrate that guide miRNAs are the predominant small RNA species originating from the PD1 silencer module.



FIG. 13 shows quantification of mature miRNAs mapping to the PD1 silencer module as a percentage of total small RNAseq reads.



FIGS. 14 A and B shows differential gene expression in ROR1+PD1 silencer cells compared to ROR1 UltraCAR-T control cells.



FIGS. 15A-D show a comparison of predicted miRNA binding strength to transcript log fold change.



FIG. 16 provides an exemplary scheme for a genetic construct of the present disclosure.



FIG. 17 provides a schematic depiction of pathways and elements of the present disclosure.



FIG. 18 provides examples of pathways by which treatment regimens of the present disclosure can proceed.



FIG. 19 provides an indication of additional embodiments of the present disclosure.



FIG. 20 provides an indication of additional embodiments of the present disclosure.





DETAILED DESCRIPTION OF THE DISCLOSURE

The following description and examples illustrate embodiments of the present disclosure in detail.


It is to be understood that the present disclosure is not limited to the particular embodiments described herein and as such can vary. Those of skill in the art will recognize that there are variations and modifications of the present disclosure, which are encompassed within the scope of the present invention.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosure pertains.


The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.


Although various features of the disclosure can be described in the context of a single embodiment, the features can also be provided separately or in any suitable combination. Conversely, although the present disclosure can be described herein in the context of separate embodiments for clarity, the present disclosure can also be implemented in a single embodiment.


The following definitions supplement those in the art and are directed to the current application and are not to be imputed to any related or unrelated case, e.g., to any commonly owned patent or application. The terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.


I. Definitions

In this application, the use of the singular includes the plural unless specifically stated otherwise. As used in the specification, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise.


In this application, the use of “or” means “and/or” unless stated otherwise. The terms “and/or” and “any combination thereof” and their grammatical equivalents as used herein, can be used interchangeably. These terms can convey that any combination is specifically contemplated. Solely for illustrative purposes, the following phrases “A, B, and/or C” or “A, B, C, or any combination thereof” can mean “A individually; B individually; C individually; A and B; B and C; A and C; and A, B, and C.” The term “or” can be used conjunctively or disjunctively, unless the context specifically refers to a disjunctive use.


Furthermore, use of the term “including” as well as other forms, such as “include,” “includes,” and “included,” is not limiting.


Reference in the specification to “some embodiments,” “an embodiment,” “one embodiment” or “other embodiments” means that a particular feature, structure, or characteristic described in connection with the embodiments is included in at least some embodiments, but not necessarily all embodiments, of the present disclosures.


As used in this specification and the claim(s), the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps. It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method or composition of the disclosure, and vice versa. Furthermore, compositions of the present disclosure can be used to achieve methods of the present disclosure.


The term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, up to 10%, up to 5%, or up to 1% of a given value. In another example, the amount “about 10” includes 10 and any amounts from 9 to 11. In yet another example, the term “about” in relation to a reference numerical value can also include a range of values plus or minus 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% from that value. Alternatively, particularly with respect to biological systems or processes, the term “about” can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term “about” meaning within an acceptable error range for the particular value should be assumed.


A “therapeutically-effective amount” or “therapeutically-effective dose” refers to an amount or dose effective, for periods of time necessary, to achieve a desired therapeutic result. The amount can vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the inventive nucleic acid sequences to elicit a desired response in the individual.


“Polynucleotide” or “oligonucleotide” refers to a polymeric form of nucleotides or nucleic acids of any length, either ribonucleotides or deoxyribonucleotides. This term refers only to the primary structure of the molecule. Thus, this term includes double and single stranded DNA, triplex DNA, as well as double and single stranded RNA. It also includes modified, for example, by methylation and/or by capping, and unmodified forms of the polynucleotide. The term is also meant to include molecules that include non-naturally occurring or synthetic nucleotides as well as nucleotide analogs.


Unless otherwise stated, nucleic acid sequences in the text of this specification are given, when read from left to right, in the 5′ to 3′ direction.


The terms “transfection,” “transformation,” “nucleofection,” or “transduction” as used herein refer to the introduction of one or more exogenous polynucleotides into a host cell or organism by using physical, chemical, and/or electrical methods. The nucleic acid sequences and vectors disclosed herein can be introduced into a cell or organism by any such methods, including, for example, by electroporation, calcium phosphate co-precipitation, strontium phosphate DNA co-precipitation, liposome mediated-transfection, DEAE dextran mediated-transfection, polycationic mediated-transfection, tungsten particle-facilitated microparticle bombardment, viral, and/or non-viral mediated transfection. In some cases, the method of introducing nucleic acids into the cell or organism involves the use of viral, retroviral, lentiviral, or transposon, or transposable element-mediated (e.g., Sleeping Beauty) vectors.


“Polypeptide,” “peptide,” and their grammatical equivalents as used herein refer to a polymer of amino acid residues. The polypeptide can optionally include glycosylation or other modifications typical for a given protein in a given cellular environment. Polypeptides and proteins disclosed herein (including functional fragments and functional variants thereof) can comprise synthetic amino acids in place of one or more naturally-occurring amino acids. Such synthetic amino acids are known in the art, and include, for example, aminocyclohexane carboxylic acid, norleucine, α-amino n-decanoic acid, homoserine, S-acetylaminomethyl-cysteine, trans-3- and trans-4-hydroxyproline, 4-aminophenylalanine, 4-nitrophenylalanine, 4-chlorophenylalanine, 4-carboxyphenylalanine, β-phenylserine β-hydroxyphenylalanine, phenylglycine, α-naphthylalanine, cyclohexylalanine, cyclohexylglycine, indoline-2-carboxylic acid, 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid, aminomalonic acid, aminomalonic acid monoamide, N′-benzyl-N′-methyl-lysine, N′,N′-dibenzyl-lysine, 6-hydroxylysine, ornithine, α-aminocyclopentane carboxylic acid, α-aminocyclohexane carboxylic acid, α-aminocycloheptane carboxylic acid, α-(2-amino-2-norbornane)-carboxylic acid, α,γ-diaminobutyric acid, α,β-diaminopropionic acid, homophenylalanine, and α-tert-butylglycine. The present disclosure further contemplates that expression of polypeptides or proteins described herein in an engineered cell can be associated with post-translational modifications of one or more amino acids of the polypeptide or protein. Non-limiting examples of post-translational modifications include phosphorylation, acylation including acetylation and formylation, glycosylation (including N-linked and O-linked), amidation, hydroxylation, alkylation including methylation and ethylation, ubiquitylation, addition of pyrrolidone carboxylic acid, formation of disulfide bridges, sulfation, myristoylation, palmitoylation, isoprenylation, farnesylation, geranylation, glypiation, lipoylation and iodination.


The term “conservative amino acid substitution” or “conservative mutation” refers to the replacement of one amino acid by another amino acid with a common property. A functional way to define common properties between individual amino acids is to analyze the normalized frequencies of amino acid changes between corresponding proteins of homologous organisms (Schulz, G. E. and Schirmer, R. H., Principles of Protein Structure, Springer-Verlag, New York (1979)). According to such analyses, groups of amino acids can be defined where amino acids within a group exchange preferentially with each other, and therefore resemble each other most in their impact on the overall protein structure (Schulz, G. E. and Schirmer, R. H., supra). Examples of conservative mutations include amino acid substitutions of amino acids within the sub-groups below, for example, lysine for arginine and vice versa such that a positive charge can be maintained; glutamic acid for aspartic acid and vice versa such that a negative charge can be maintained; serine for threonine such that a free —OH can be maintained; and glutamine for asparagine such that a free —NH2 can be maintained. Exemplary conservative amino acid substitutions are shown in the following chart:
















Type of Amino Acid
Substitutable Amino Acids









Hydrophilic
Ala, Pro, Gly, Glu, Asp, Gln, Asn, Ser, Thr



Sulphydryl
Cys



Aliphatic
Val, Ile, Leu, Met



Basic
Lys, Arg, His



Aromatic
Phe, Tyr, Trp










An amino acid sequence that differs from a reference amino acid sequence by only conservative amino acid substitutions will be referred to herein as a “conservatively-substituted variant” of the reference sequence.


In some embodiments, the functional variants can comprise the amino acid sequence of the reference protein with at least one non-conservative amino acid substitution. The term “non-conservative mutations” involve amino acid substitutions between different groups, for example, lysine for tryptophan, or phenylalanine for serine, etc. In this case, it is preferable for the non-conservative amino acid substitution to not interfere with, or inhibit the biological activity of, the functional variant. The non-conservative amino acid substitution can enhance the biological activity of the functional variant, such that the biological activity of the functional variant is increased as compared to the homologous parent protein. Amino acid substitutability is discussed in more detail, for example, in L. Y. Yampolsky and A. Stoltzfus, “The Exchangeability of Amino acids in Proteins,” Genetics 2005 August; 170(4):1459-1472.


The terms “identical” and its grammatical equivalents as used herein or “sequence identity” in the context of two nucleic acid sequences or amino acid sequences of polypeptides refer to the residues in the two sequences which are the same when aligned for maximum correspondence over a specified comparison window. A “comparison window,” as used herein, refers to a segment of at least about 20 contiguous positions, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence can be compared to a reference sequence of the same number of contiguous positions after the two sequences are aligned optimally. Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison can be conducted by the local homology algorithm of Smith and Waterman, Adv. Appl. Math., 2:482 (1981); by the alignment algorithm of Needleman and Wunsch, J. Mol. Biol., 48:443 (1970); by the search for similarity method of Pearson and Lipman, Proc. Nat. Acad. Sci U.S.A., 85:2444 (1988); by computerized implementations of these algorithms (including, but not limited to CLUSTAL in the PC/Gene program by Intelligentics, Mountain View Calif., GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis., U.S.A.); the CLUSTAL program is well described by Higgins and Sharp, Gene, 73:237-244 (1988) and Higgins and Sharp, CABIOS, 5:151-153 (1989); Corpet et al., Nucleic Acids Res., 16:10881-10890 (1988); Huang et al., Computer Applications in the Biosciences, 8:155-165 (1992); and Pearson et al., Methods in Molecular Biology, 24:307-331 (1994). Alignment is also often performed by inspection and manual alignment. In one class of embodiments, the polypeptides herein are at least 80%, 85%, 90%, 98% 99% or 100% identical to a reference polypeptide (i.e., the full length thereof), or a fragment thereof, e.g., as measured by BLASTP (or CLUSTAL, or any other available alignment software) using default parameters. Similarly, nucleic acids can also be described with reference to a starting nucleic acid, e.g., they can be 50%, 60%, 70%, 75%, 80%, 85%, 90%, 98%, 99% or 100% identical to a reference nucleic acid (i.e., the full length thereof) or a fragment thereof, e.g., as measured by BLASTN (or CLUSTAL, or any other available alignment software) using default parameters. When one molecule is said to have certain percentage of sequence identity with a larger molecule, it means that when the two molecules are optimally aligned, the percentage of residues in the smaller molecule finds a match residue in the larger molecule in accordance with the order by which the two molecules are optimally aligned.


For purposes of this specification and the claims, it is understood that the phrase “having at least 50% sequence identity with” a reference sequence, or referencing any range therein (e.g., “at least 80% sequence identity with”) encompasses the reference sequence itself. Thus, for example, a claim reciting “a nucleic acid having at least 80% sequence identity with SEQ ID NO: 0” encompasses SEQ ID NO: 0 itself.


The term “substantially identical” and its grammatical equivalents as applied to nucleic acid or amino acid sequences mean that a nucleic acid or amino acid sequence comprises a sequence that has at least 95% sequence identity with a reference sequence using the programs described above, e.g., BLAST, using standard parameters.


“Homology” is generally inferred from sequence identity between two or more nucleic acids or proteins (or sequences thereof). The precise percentage of identity between sequences that is useful in establishing homology varies with the nucleic acid and protein at issue, but as little as 25% sequence identity is routinely used to establish homology. Higher levels of sequence identity, e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% or more can also be used to establish homology. Methods for determining sequence identity percentages (e.g., BLASTP and BLASTN using default parameters) are described herein and are generally available. Nucleic acids and/or nucleic acid sequences are “homologous” when they are derived, naturally or artificially, from a common ancestral nucleic acid or nucleic acid sequence. Proteins and/or protein sequences are “homologous” when their encoding DNAs are derived, naturally or artificially, from a common ancestral nucleic acid or nucleic acid sequence. The homologous molecules can be termed “homologs.” For example, any naturally occurring proteins can be modified by any available mutagenesis method. When expressed, this mutagenized nucleic acid encodes a polypeptide that is homologous to the protein encoded by the original nucleic acid.


Also contemplated and included herein are nucleic acid molecules that hybridize to the disclosed sequences. Hybridization conditions may be mild, moderate, or stringent, as is warranted.


Appropriate stringency conditions which promote DNA hybridization, for example, 6× sodium chloride/sodium citrate (SSC) at about 45° C., followed by a wash of 2×SSC at 50° C., are known or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. “Stringent hybridization conditions” are those that include a salt concentration of 1.0 M NaCl in 50% formamide, at a temperature of 37° C. for 4 to 12 hours, followed by a wash in 0.1×SSC at 60-65° C.


As will be appreciated by the skilled practitioner, slight changes in nucleic acid sequence do not necessarily alter the amino acid sequence of the encoded polypeptide. This disclosure embraces the degeneracy of codon usage as would be understood by one of ordinary skill in the art. For example, as known in the art, different codons will code for the same amino acid as illustrated in the following chart.
















Amino Acid
Codons









Ala/A
GCT, GCC, GCA, GCG



Arg/R
CGT, CGC, CGA, CGG, AGA, AGG



Asn/N
AAT, AAC



Asp/D
GAT, GAC



Cys/C
TGT, UGC



Gln/Q
CAA, CAG



Glu/E
GAA, GAG



Gly/G
GGT, GGC, GGA, GGG



His/H
CAT, CAC



Ile/I
ATT, ATC, ATA



Leu/L
TTA, TTG, CTT, CTC, CTA, CTG



Lys/K
AAA, AAG



Met/M
ATG



Phe/F
TTT, TTC



Pro/P
CCT, CCC, CCA, CCG



Ser/S
TCT, TCC, TCA, TCG, AGT, AGC



Thr/T
ACT, ACC, ACA, ACG



Trp/W
TGG



Tyr/Y
TAT, TAC



Val/V
GTT, GTC, GTA, GTG



START
ATG



STOP
TAG, TGA, TAA










As used herein, the phrase “codon degenerate variant” when used with reference to a nucleic acid sequence means a nucleic acid sequence that differs from the referenced sequence, but that encodes a polypeptide having the same amino acid sequence as that encoded by the referenced sequence.


Additionally, it will be appreciated by persons skilled in the art that partial sequences often work as effectively as full-length versions. The ways in which the nucleotide sequence can be varied or shortened are well known to persons skilled in the art, as are ways of testing the suitability or effectiveness of the altered genes. In certain embodiments, suitability and/or effectiveness of the altered gene may easily be tested by, for example, conventional gas chromatography. All such variations of the genes are therefore included as part of the present disclosure.


The term “isolated” and its grammatical equivalents as used herein refer to the removal of a nucleic acid from its natural environment. It is to be understood, however, that nucleic acids and proteins can be formulated with diluents or adjuvants and still for practical purposes be isolated.


The term “purified” and its grammatical equivalents as used herein refer to a molecule or composition, whether removed from nature (including genomic DNA and mRNA) or synthesized (including cDNA) and/or amplified under laboratory conditions, that has been increased in purity, wherein “purity” is a relative term, not “absolute purity.” For example, nucleic acids typically are mixed with an acceptable carrier or diluent when used for introduction into cells. The term “substantially purified” and its grammatical equivalents as used herein refer to a nucleic acid sequence, polypeptide, protein or other compound that is essentially free, i.e., is more than about 50% free of, more than about 70% free of, more than about 90% free of, the polynucleotides, proteins, polypeptides and other molecules that the nucleic acid, polypeptide, protein or other compound is naturally associated with.


“T cell” or “T lymphocyte” as used herein is a type of lymphocyte that plays a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell surface.


“Transposon,” “transposable element” or “TE” refers to a vector DNA sequence that can change its position within the genome, sometimes creating or reversing mutations and altering the cell's genome size. Transposition often results in duplication of the transposon. Class I transposons are copied in two stages: first, they are transcribed from DNA to RNA, and the RNA produced is then reverse transcribed to DNA. This copied DNA is then inserted at a new position into the genome. The reverse transcription step is catalyzed by a reverse transcriptase, which can be encoded by the transposon itself. The characteristics of retrotransposons are similar to retroviruses, such as HIV. The cut-and-paste transposition mechanism of class II transposons does not involve an RNA intermediate. The transpositions are catalyzed by several transposase enzymes. Some transposases non-specifically bind to any target site in DNA, whereas others bind to specific DNA sequence targets. The transposase makes a staggered cut at the target site resulting in single-strand 5′ or 3′ DNA overhangs (sticky ends). This step cuts out the DNA transposon, which is then ligated into a new target site; this process involves activity of a DNA polymerase that fills in gaps and of a DNA ligase that closes the sugar-phosphate backbone. This results in duplication of the target site. The insertion sites of DNA transposons can be identified by short direct repeats which can be created by the staggered cut in the target DNA and filling in by DNA polymerase, followed by a series of inverted repeats important for the transposon excision by transposase. Cut-and-paste transposons can be duplicated if their transposition takes place during S phase of the cell cycle when a donor site has already been replicated, but a target site has not yet been replicated. Transposition can be classified as either “autonomous” or “non-autonomous” in both Class I and Class II transposons. Autonomous transposons can move by themselves while non-autonomous transposons require the presence of another transposon to move. This is often because non-autonomous transposons lack transposase (for class II) or reverse transcriptase (for class I).


“Transposase” refers an enzyme that binds to the end of a transposon and catalyzes the movement of the transposon to another part of the genome by a cut and paste mechanism or a replicative transposition mechanism. In some embodiments, the transposase's catalytic activity can be utilized to move gene(s) from a vector to the genome.


An “expression vector” or “vector” is any genetic element, e.g., a plasmid, a mini-circle, a nanoplasmid, chromosome, virus, transposon, behaving either as an autonomous unit of polynucleotide replication within a cell. (i.e. capable of replication under its own control) or being rendered capable of replication by insertion into a host cell chromosome, having attached to it another polynucleotide segment, so as to bring about the replication and/or expression of the attached segment. Suitable vectors include, but are not limited to, plasmids, transposons, bacteriophages and cosmids. Vectors can contain polynucleotide sequences that are necessary to effect ligation or insertion of the vector into a desired host cell and to effect the expression of the attached segment. Such sequences differ depending on the host organism; they include promoter sequences to effect transcription, enhancer sequences to increase transcription, ribosomal binding site sequences and transcription and translation termination sequences. Alternatively, expression vectors can be capable of directly expressing nucleic acid sequence products encoded therein without ligation or integration of the vector into host cell DNA sequences. In some embodiments, the vector is an “episomal expression vector” or “episome,” which is able to replicate in a host cell, and persists as an extrachromosomal segment of DNA within the host cell in the presence of appropriate selective pressure (see, e.g., Conese et al., Gene Therapy, 11:1735-1742 (2004)). Representative commercially-available episomal expression vectors include, but are not limited to, episomal plasmids that utilize Epstein Barr Nuclear Antigen 1 (EBNA1) and the Epstein Barr Virus (EBV) origin of replication (oriP). The vectors pREP4, pCEP4, pREP7, and pcDNA3.1 from Invitrogen (Carlsbad, Calif.) and pBK-CMV from Stratagene (La Jolla, Calif.) represent non-limiting examples of an episomal vector that uses T-antigen and the SV40 origin of replication in lieu of EBNA1 and oriP. A vector also can comprise a selectable marker gene. In certain embodiments where nano plasmids are utilized, strains such as R6K that utilizes an antisense RNA selection marker (e.g. sucrose tolerance) can be used.


The term “selectable marker gene” refers to a nucleic acid sequence that allows cells expressing the nucleic acid sequence to be specifically selected for or against, in the presence of a corresponding selective agent. Suitable selectable marker genes are known in the art and described in, e.g., International Patent Application Publications WO 1992/08796 and WO 1994/28143; Wigler et al., Proc. Natl. Acad. Sci. USA, 77: 3567 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA, 78: 1527 (1981); Mulligan & Berg, Proc. Natl. Acad. Sci. USA, 78: 2072 (1981); Colberre-Garapin et al., J. Mol. Biol., 150:1 (1981); Santerre et al., Gene, 30: 147 (1984); Kent et al., Science, 237: 901-903 (1987); Wigler et al., Cell, 11: 223 (1977); Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA, 48: 2026 (1962); Lowy et al., Cell, 22: 817 (1980); and U.S. Pat. Nos. 5,122,464 and 5,770,359.


The term “coding sequence” refers to a segment of a polynucleotide that encodes for protein or polypeptide. The region or sequence is bounded nearer the 5′ end by a start codon and nearer the 3′ end with a stop codon. Coding sequences can also be referred to as open reading frames.


The term “operably linked” refers to refers to the physical and/or functional linkage of a DNA segment to another DNA segment in such a way as to allow the segments to function in their intended manners. A DNA sequence encoding a gene product is operably linked to a regulatory sequence when it is linked to the regulatory sequence, such as, for example, promoters, enhancers and/or silencers, in a manner, that allows modulation of transcription of the DNA sequence, directly or indirectly. For example, a DNA sequence is operably linked to a promoter when it is ligated to the promoter downstream with respect to the transcription initiation site of the promoter and in the correct reading frame with respect to the transcription initiation site so as to allow transcription elongation to proceed through the DNA sequence. An enhancer or silencer is operably linked to a DNA sequence coding for a gene product when it is ligated to the DNA sequence in such a manner as to, respectively, increase or decrease the transcription of the DNA sequence. Enhancers and silencers can be located upstream or downstream of or embedded within the coding regions of the DNA sequence. A DNA for a signal sequence is operably linked to DNA coding for a polypeptide if the signal sequence is expressed as a pre-protein that participates in the secretion of the polypeptide. Linkage of DNA sequences to regulatory sequences is typically accomplished by ligation at suitable restriction sites or via adapters or linkers inserted in the sequence using restriction endonucleases known to one of skill in the art.


The terms “induce,” “induction” and their grammatical equivalents as used herein refer to an increase in nucleic acid sequence transcription, promoter activity and/or expression brought about by a transcriptional regulator, relative to some basal level of transcription.


The term “transcriptional regulator” refers to a biochemical element that acts to prevent or inhibit the transcription of a promoter-driven DNA sequence under certain environmental conditions (e.g., a repressor or nuclear inhibitory protein), or to permit or stimulate the transcription of the promoter-driven DNA sequence under certain environmental conditions (e.g., an inducer or an enhancer).


The term “enhancer,” as used herein, refers to a DNA sequence that increases transcription of, for example, a nucleic acid sequence to which it is operably linked. Enhancers can be located many kilobases away from the coding region of the nucleic acid sequence and can mediate the binding of regulatory factors, patterns of DNA methylation, or changes in DNA structure. A large number of enhancers from a variety of different sources are well known in the art and are available as or within cloned polynucleotides (from, e.g., depositories such as the ATCC as well as other commercial or individual sources). A number of polynucleotides comprising promoters (such as the commonly-used CMV promoter) also comprise enhancer sequences. Enhancers can be located upstream or downstream of coding sequences or within coding sequences. The term “Ig enhancers” refers to enhancer elements derived from enhancer regions mapped within the immunoglobulin (Ig) locus (such enhancers include for example, the heavy chain (mu) 5′ enhancers, light chain (kappa) 5′ enhancers, kappa and mu intronic enhancers, and 3′ enhancers (see generally Paul W. E. (ed), Fundamental Immunology, 3rd Edition, Raven Press, New York (1993), pages 353-363; and U.S. Pat. No. 5,885,827).


The term “promoter” refers to a region of a polynucleotide that initiates transcription of a coding sequence. Promoters are located near the transcription start sites of genes, on the same strand and upstream on the DNA (towards the 5′ region of the sense strand). Some promoters are constitutive as they are active in all circumstances in the cell, while others are regulated becoming active in response to specific stimuli, e.g., an inducible promoter. The term “promoter activity” and its grammatical equivalents as used herein refer to the extent of expression of nucleotide sequence that is operably linked to the promoter whose activity is being measured. Promoter activity can be measured directly by determining the amount of RNA transcript produced, for example by Northern blot analysis or indirectly by determining the amount of product coded for by the linked nucleic acid sequence, such as a reporter nucleic acid sequence linked to the promoter.


“Inducible promoter” refers to a promoter, that is induced into activity by the presence or absence of transcriptional regulators, e.g., biotic or abiotic factors. Inducible promoters are useful because the expression of genes operably linked to them can be turned on or off at certain stages of development of an organism or in a particular tissue. Non-limiting examples of inducible promoters include alcohol-regulated promoters, tetracycline-regulated promoters, steroid-regulated promoters, metal-regulated promoters, pathogenesis-regulated promoters, temperature-regulated promoters and light-regulated promoters. The inducible promoter can be part of a gene switch or genetic switch.


“T cell” or “T lymphocyte” as used herein is a type of lymphocyte that plays a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell surface.


As used herein, the phrase “functional fragment” when used with reference to a polypeptide refers to a fragment of such polypeptide that possesses the primary function of the referenced polypeptide. For example, a functional fragment of a polypeptide that serves as a transmembrane domain is a fragment of that polypeptide that also serves as a transmembrane domain. In certain embodiments, the functional fragment of a polypeptide is shorter than the referenced polypeptide by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. When used with reference to a nucleic acid, the phrase “functional fragment” refers to a fragment of the referenced nucleic acid that encodes a polypeptide having the same primary function as the polypeptide encoded by the referenced nucleic acid.


As used herein, the phrase “functional variant” when used with reference to a polypeptide refers to a polypeptide that differs from the referenced polypeptide but possesses the primary function of the referenced polypeptide. For example, a functional variant of a polypeptide that serves as a transmembrane domain is a fragment of that polypeptide that also serves as a transmembrane domain. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the referenced amino acid sequence and/or is a conservatively-substituted variant of the referenced sequence. When used with reference to a nucleic acid, the phrase “functional variant” refers to a nucleic acid that differs from the referenced nucleic acid but encodes a polypeptide having the same primary function as the polypeptide encoded by the referenced nucleic acid. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the referenced nucleic acid sequence, hybridizes under stringent hybridization conditions with the complement of the referenced nucleic acid sequence, or is a codon degenerate variant of the nucleic acid sequence.


The term “antibody,” also known as immunoglobulin (Ig), as used herein can refer to a monoclonal or polyclonal antibody. The term “monoclonal antibodies,” as used herein, refers to antibodies that are produced by a single clone of B-cells and bind to the same epitope. In contrast, “polyclonal antibodies” refer to a population of antibodies that is produced by different B-cells and bind to different epitopes of the same antigen. The antibodies can be from any animal origin. An antibody can be IgG (including IgG1, IgG2, IgG3, and IgG4), IgA (including IgA1 and IgA2), IgD, IgE, or IgM, and IgY. In some embodiments, the antibody can a single-chain whole antibody. An antibody typically consists of four polypeptides: two identical copies of a heavy (H) chain polypeptide and two identical copies of a light (L) chain polypeptide. Each of the heavy chains contains one N-terminal variable (VH) region and three C-terminal constant (CH1, CH2 and CH3) regions, and each light chain contains one N-terminal variable (VL) region and one C-terminal constant (CL) region. The variable regions of each pair of light and heavy chains form the antigen-binding site of an antibody. The VH and VL regions have a similar general structure, with each region comprising four framework regions, whose sequences are relatively conserved. The framework regions are connected by three complementarity determining regions (CDRs). The three CDRs, known as CDR1, CDR2, and CDR3, form the “hypervariable region” of an antibody, which is responsible for antigen binding. These particular regions have been described by Kabat et al., J. Biol. Chem. 252, 6609-6616 (1977) and Kabat et al., Sequences of protein of immunological interest. (1991), by Chothia et al., J. Mol. Biol. 196:901-917 (1987), and by MacCallum et al., J. Mol. Biol. 262:732-745 (1996), where the definitions include overlapping or subsets of amino acid residues when compared against each other. Preferably, the term “CDR” is a CDR as defined by Kabat, based on sequence comparisons. CDRH1, CDRH2 and CDRH3 denote the heavy chain CDRs, and CDRL1, CDRL2 and CDRL3 denote the light chain CDRs.


The terms “fragment of an antibody,” “antibody fragment,” “fragment of an antibody,” “antigen-binding portion” and their grammatical equivalents are used interchangeably herein to mean one or more fragments or portions of an antibody that retain the ability to specifically bind to an antigen (see, generally, Holliger et al., Nat. Biotech., 23(9):1126-1129 (2005)). The antibody fragment desirably comprises, for example, one or more CDRs, the variable region (or portions thereof), the constant region (or portions thereof), or combinations thereof. Non-limiting examples of antibody fragments include (1) a Fab fragment, which is a monovalent fragment consisting of the VL, VH, CL, and CH1 domains; (2) a F(ab′)2 fragment, which is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the stalk region; (3) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; (4) a single chain Fv (scFv), which is a monovalent molecule consisting of the two domains of the Fv fragment (i.e., VL and VH) joined by a linker that enables the two domains to be synthesized as a single polypeptide chain (see, e.g., Bird et al., Science, 242: 423-426 (1988); Huston et al., Proc. Natl. Acad. Sci. USA, 85: 5879-5883 (1988); and Osbourn et al., Nat. Biotechnol., 16: 778 (1998)) and (5) a diabody, which is a dimer of polypeptide chains, wherein each polypeptide chain comprises a VH connected to a VL by a peptide linker that is too short to allow pairing between the VH and VL on the same polypeptide chain, thereby driving the pairing between the complementary domains on different VH-VL polypeptide chains to generate a dimeric molecule having two functional antigen-binding sites. Antibody fragments are known in the art and are described in more detail in, e.g., U.S. Pat. No. 8,603,950.


The terms “antigen recognition moiety,” “antigen recognition domain,” “antigen-binding domain,” and “antigen binding region” refer to a molecule or portion of a molecule that specifically binds to an antigen. In one embodiment, the antigen recognition moiety is an antibody, antibody like molecule or fragment thereof.


The term “proliferative disease” refers to a unifying concept in which excessive proliferation of cells and/or turnover of cellular matrix contributes significantly to the pathogenesis of the disease, including cancer. In some embodiments, the proliferative disease is cancer.


“Patient” or “subject” refers to a mammalian subject diagnosed with or suspected of having or developing a proliferative disorder such as cancer. In some embodiments, the term “patient” refers to a mammalian subject with a higher than average likelihood of developing a proliferative disorder such as cancer. Exemplary patients can be humans, apes, dogs, pigs, cattle, cats, horses, goats, sheep, rodents and other mammalians that can benefit from the therapies disclosed herein. Exemplary human patients can be male and/or female. “Patient in need thereof” or “subject in need thereof” means a patient diagnosed with or suspected of having a disease or disorder, for instance, but not restricted to cancer.


“Administering” refers to herein as providing one or more compositions described herein to a patient or a subject. By way of example and not limitation, composition administration, e.g., injection, can be performed by intravenous (i.v.) injection, sub-cutaneous (s.c.) injection, intradermal (i.d.) injection, intraperitoneal (i.p.) injection, or intramuscular (i.m.) injection. One or more such routes can be employed. Parenteral administration can be, for example, by bolus injection or by gradual perfusion over time. Alternatively, or concurrently, administration can be by the oral route. Additionally, administration can also be by surgical deposition of a bolus or pellet of cells, or positioning of a medical device.


As used herein, the terms “treatment,” “treating,” and their grammatical equivalents refer to obtaining a desired pharmacologic and/or physiologic effect. In some embodiments, the effect is therapeutic, i.e., the effect partially or completely cures a disease and/or adverse symptom attributable to the disease. In some embodiments, the term “treating” can include “preventing” a disease or a condition.


As used herein, a “treatment interval” refers to a treatment cycle, for example, a course of administration of a therapeutic agent that can be repeated, e.g., on a regular schedule. In some embodiments, a dosage regimen can have one or more periods of no administration of the therapeutic agent in between treatment intervals.


The terms “administered in combination,” “co-administration,” “co-administering,” and “co-providing” as used herein, mean that two (or more) different treatments are delivered to the subject during the course of the subject's affliction with the disorder, e.g., the two or more treatments are delivered after the subject has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons. In some embodiments, the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as “simultaneous” or “concurrent delivery.” In other embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration. For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive. The delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.


In some embodiments, the first treatment and second treatment can be administered simultaneously (e.g., at the same time), in the same or in separate compositions, or sequentially. Sequential administration refers to administration of one treatment before (e.g., immediately before, less than 5, 10, 15, 30, 45, 60 minutes; 1, 2, 3, 4, 6, 8, 10, 12, 16, 20, 24, 48, 72, 96 or more hours; 4, 5, 6, 7, 8, 9 or more days; 1, 2, 3, 4, 5, 6, 7, 8 or more weeks before) administration of an additional, e.g., secondary, treatment. The order of administration of the first and secondary treatment can also be reversed.


The terms “therapeutically effective amount,” therapeutic amount,” “immunologically effective amount,” “anti-tumor effective amount,” “tumor-inhibiting effective amount,” and their grammatical equivalents refer to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result. The therapeutically effective amount can vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of a composition described herein to elicit a desired response in one or more subjects. The precise amount of the compositions of the present disclosure to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient (subject).


Alternatively, the pharmacologic and/or physiologic effect of administration of one or more compositions described herein to a patient or a subject of can be “prophylactic,” i.e., the effect completely or partially prevents a disease or symptom thereof. A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired prophylactic result (e.g., prevention of disease onset).


As used herein, the term “immune checkpoint protein” refers to a molecule that transmits a suppressive signal or has an immunosuppressive function. Examples of such immune checkpoint proteins include, but are not limited to, CTLA-4, PD-1, PD-L1 (programmed cell death-ligand 1), PD-L2 (programmed cell death-ligand 2), LAG-3 (Lymphocyte activation gene 3), TIM3 (T cell immunoglobulin and mucin-3), BTLA (B and T lymphocyte attenuator), B7H3, B7H4, CD160, CD39, CD70, CD73, A2aR (adenosine A2a receptor), KIR (killer inhibitory receptor), VISTA (V-domain Ig-containing suppressor of T cell activation), IDO1 (Indoleamine 2,3-dioxygenase), Arginase I, TIGIT (T cell immunoglobulin and ITIM domain), CD115, and the like (see, Nature Reviews Cancer, 12, p. 252-264, 2012 and Cancer Cell, 27, p. 450-461, 2015).


As used herein, terms used in the identification of biological moieties may include, or may not include, a dash “-” within the term. The presence or absence of a dash does not change the intended meaning or identification of the biological moiety. By way of illustration only, and without limitation to these biological moieties, each of the following paired terms (shown with/without a dash) indicate and identify the same biological entities: CCR-4/CCR4, CD-3/CD3, CD-4/CD4, CD-33/CD33, EGFR-2/EGFR2, FLT-1/FLT1, HER-1/HER1, HER-1t/HER1t, IL-12/IL12, IL-15/IL15, IL-15Ra/IL15Rα MUC-1/MUC1, MUC-16/MUC16, ROR-1/ROR1, ROR-1R/ROR1R, TGF-Beta/TGFBeta, VEGF-1/VEGF1, VEGF-R2/VEGFR2.”


II. Combinations

In certain embodiments, miRNA(s) or polynucleotides encoding the miRNA(s) as described herein can be used in combination with a chimeric receptor or can further comprise a nucleic acid sequence encoding a chimeric receptor, respectively. In some cases, the chimeric receptor is a chimeric antigen receptor (CAR). In some instances, the CAR comprises a pattern-recognition receptor. In other cases, the chimeric receptor comprises an engineered T-cell receptor (TCR). The miRNA(s) can be used in combination with specific CARs, cytokines, and cell tags, for example MUC16-specific CAR with a fusion protein comprising IL15 and IL-15Rα (mbIL15) and truncated HER1 (HER1t), MUC1-specific CAR with mbIL15 and HER1t, or CD33-specific CAR with mbIL15 and HER1t.

  • In certain embodiments, the genetic construct can include a 5′ untranslated region (5′UTR) and/or a 3′ untranslated region (3′UTR). In some embodiments, the sequences encoding the miRNA(s) can be located in the 5′UTR and/or the 3′UTR. In some embodiments, the 5′UTR and/or the 3′UTR do not contain an intron.


In certain embodiments, a nucleic acid sequence encoding a synthetic intron with checkpoint inhibitor miRNAs is inserted into the same genetic construct as that encoding the chimeric receptor (for example, in the portion of the construct corresponding to the 5′UTR of the mRNA encoding the chimeric receptor). An exemplary depiction of the order of polynucleotide sequences can be found in FIG. 1A. In one embodiment, the 5′UTR contains one or more mature miRNAs. In some embodiments, the one or more mature miRNA(s) can be directed to the same immune checkpoint protein target, for example PD-1. In certain other embodiments, the mature miRNAs can be directed to more than one immune checkpoint protein target, for example PD-1 and TIGIT, PD1 and CTLA4, or TIGIT and CTLA4.


III. miRNA(s)

As used herein, the terms “miR,” “mir” and “miRNA” are used to refer to microRNA, a class of small non-coding RNA molecules that are capable of affecting the expression of a gene (the “target gene”) by modulating the translation of messenger RNA transcribed therefrom (either increasing or decreasing the gene's expression) and/or destabilizing such messenger RNA.


The miRNAs can be non-naturally occurring. The terms “non-naturally occurring,” “synthetic,” and “artificial,” as used to describe miRNA(s) herein, are used interchangeably and refer to an miRNA having a sequence that does not occur in nature. In some embodiments, a non-naturally occurring miRNA effectively mimics naturally-occurring miRNA. Non-naturally occurring miRNA can be designed such that the desired hairpins or loops of the corresponding naturally-occurring miRNA are maintained when a naturally-occurring mature miRNA sequence is replaced with a synthetic sequence designed to target a specific transcript. For example, the non-naturally occurring miRNA can have at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, or greater sequence identity with naturally-occurring miRNA and/or can hybridize under stringent hybridization conditions with naturally-occurring miRNA. The term “miRNA,” unless otherwise indicated, refers generically to the mature, pri-, and pre-forms of a particular microRNA and functional fragments and variants thereof.


The term “pri-miRNA” refers to the primary miRNA transcript containing at least one RNA hairpin. Exemplary depictions of pri-miRNA are illustrated in FIG. 1B. The RNA hairpin(s) are cleaved from the pri-miRNA in the cell nucleus to form one or more precursor miRNAs (“pre-miRNAs). This pre-miRNA is exported into the cytoplasm where the stem loop structure is cleaved to produce a double-stranded miRNA comprising a miRNA-5p strand from the former 5′ arm of the hairpin loop and a miRNA-3p strand from the former 3′ arm of the hairpin loop. The Argonaute protein then binds the double-stranded miRNA and one of the strands (either the miRNA-5p sequence or the miRNA-3p sequence) is released. The remaining bound strand become the “guide strand” whereas the released strand is known as the “passenger strand” and preferably degrades. The guide strand then goes on to interact with the messenger RNA derived from the target gene, thus affecting its translation.


Both the miRNA-5p and miRNA-3p strand sequences will be referred to herein as “mature miRNA” sequences. The remaining portions of the pri-miRNA (the portion thereof 5′ to the miRNA-5p sequence, the portion thereof 3′ to the miRNA-3p sequence, and the stem loop sequence in between the miRNA-5p and miRNA-3p sequences) will be collectively referred to as miRNA backbone sequences. The term “5′ backbone sequence” will be used herein to refer to the backbone sequence that, in a pri-miRNA, is 5′ of the miRNA-5p sequence. The term “3′ backbone sequence” will be used herein to refer to the backbone sequence that, in a pri-miRNA, is 3′ of the miRNA-3p sequence. The term “stem loop sequence” refers to the backbone sequence that, in a pri-miRNA, is between the miRNA-5p and miRNA-3p sequences.


The pri-miRNA of the present invention may be produced from naturally-occurring pri-miRNAs by removing the native mature miRNA sequences and replacing them with non-native mature miRNA sequences wherein one of the sequences is capable of serving as a guide miRNA targeting a gene of interest. In the design of a non-naturally occurring pri-miRNA, backbone miRNA nucleic acid sequences can be derived from mouse, rat, or human miRNA sequences. As a non-limiting example, backbone miRNA sequences can be derived from miR150, miR206, miR204, miR17, miR16, miR30a, miR126, miR122, miR213, miR29b1 or miR133a1.


Examples of backbone sequences that may be used in the practice of the present invention include, but are not limited, to those encoded by the DNA sequences listed in Table 1 below. The symbols of “X” and “Y” in Table 1 indicate nucleic acid sequences encoding, respectively, the guide miRNA (which may be either miRNA-5p or miRNA-3p) and the passenger miRNA (which may be either miRNA-5p or miRNA-3p), whereas the symbol of “n” indicates the number of nucleotides in such sequences, for example 16-30, preferably 18-25. In some embodiments, n can be 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34 or 35 nucleotides. In certain embodiments, the backbone miRNA sequences are those that are encoded by sequences that hybridize under stringent hybridization conditions with the complement of any one of the sequences listed in Table 1.









TABLE 1







Nucleic acids encoding miRNA backbone sequences









Nucleic acid sequences encoding


Synthetic
5' Backbone Sequence Xn-Stem Loop


miRNA
Sequence Yn-3' Backbone Sequence





miR204
SEQ ID NO: 1- custom-character  -SEQ ID NO: 2- custom-character  -SEQ ID NO: 3


miR206
SEQ ID NO: 4- custom-character  -SEQ ID NO: 5- custom-character  -SEQ ID NO: 6


miR17
SEQ ID NO: 7- custom-character  -SEQ ID NO: 8- custom-character  -SEQ ID NO: 9


miR150
SEQ ID NO: 10- custom-character  -SEQ ID NO: 11- custom-character  -SEQ ID NO: 12


miR150
SEQ ID NO: 13- custom-character  -SEQ ID NO: 14- custom-character  -SEQ ID NO: 15


miR16
SEQ ID NO: 16- custom-character  -SEQ ID NO: 17- custom-character  -SEQ ID NO: 18


miR30a
SEQ ID NO: 19- custom-character  -SEQ ID



NO: 20- custom-character  -SEQ ID NO: 21


miR126
SEQ ID NO: 22- custom-character  -SEQ ID NO: 23- custom-character  -SEQ ID NO: 24


miR122
SEQ ID NO: 25- custom-character  -SEQ ID NO: 26- custom-character  -SEQ ID NO: 27


miR214
SEQ ID NO: 28- custom-character  -SEQ ID NO: 29- custom-character  -SEQ ID NO: 30


miR214
SEQ ID NO: 31- custom-character  -SEQ ID NO: 32- custom-character  -SEQ ID NO: 33


miR29b1
SEQ ID NO: 34- custom-character  -SEQ ID NO: 35- custom-character  -SEQ ID NO: 36


miR29b1
SEQ ID NO: 37- custom-character  -SEQ ID NO: 38- custom-character  -SEQ ID NO: 39


miR133a1
SEQ ID NO: 40- custom-character  -SEQ ID NO: 41- custom-character  -SEQ ID NO: 42


miR26a
SEQ ID NO: 43- custom-character  -SEQ ID NO: 44- custom-character  -SEQ ID NO: 45


miR412
SEQ ID NO: 46- custom-character  -SEQ ID NO: 47- custom-character  -SEQ ID NO: 48


miR-19
SEQ ID NO: 49- custom-character  -SEQ ID NO: 50- custom-character  -SEQ ID NO: 51


miR-21
SEQ ID NO: 52- custom-character  -SEQ ID NO: 53- custom-character  -SEQ ID NO: 54


miR-142
SEQ ID NO: 55- custom-character  -SEQ ID NO: 56- custom-character  -SEQ ID NO: 57


miR-494
SEQ ID NO: 58- custom-character  -SEQ ID NO: 59- custom-character  -SEQ ID NO: 60


miR-1915
SEQ ID NO: 61- custom-character  -SEQ ID NO: 62- custom-character  -SEQ ID NO: 63









While the miRNA-5p and miRNA-3p sequences hybridize with each other, they are not necessarily exactly complementary. In the design of a non-naturally occurring miRNA, compensatory mutations can be made in the miRNA-5p and/or miRNA-3p sequences so as to maintain the RNA folding and free energy of the native miRNA. In certain embodiments, the sequence encoding the miRNA-3p sequence has at least 50%, 60%, 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the complement to the sequence encoding the miRNA-5p sequence or is capable of hybridizing under stringent hybridization conditions with the sequence encoding the miRNA-5p sequence.


In certain embodiments, the target gene encodes a checkpoint inhibitor protein. Thus, the present invention relates in part to a polynucleotide encoding a non-naturally occurring miRNA that inhibits the expression of an immune checkpoint protein. In certain such embodiments, the target gene encodes CTLA4, PD-1, PD-L1, TIGIT, TIM3, LAG3, GITR, or PIK3IP1. Non-limiting examples of nucleic acid sequences encoding the guide miRNA targeting genes encoding such checkpoint inhibitors are listed in Table 2. Table 2 also lists the sequences encoding the passenger strand. As previous discussed, the guide and passenger strand are not necessarily complementary. It is contemplated that the passenger strand may also serve to target the messenger RNA associated with the target gene. It is also contemplated that sequences that sequences that hybridize under stringent hybridization conditions with the compliments of the sequences listed in Table 2 may also be used. The mature miRNA sequences used may be combined with a specific pri-miRNA backbone. Table 2 also lists backbones that can be combined with the mature guide and passenger miRNAs listed therein.









TABLE 2







Nucleic acid sequences encoding mature miRNA sequences













DNA encoding


miRNA

DNA encoding
passenger


Target
Backbone
guide miRNA
miRNA





CTLA4
miR-204
(SEQ ID NO: 64)
(SEQ ID NO: 65)


CTLA4
miR-26a
(SEQ ID NO: 66)
(SEQ ID NO: 67)


CTLA4
miR-30a
(SEQ ID NO: 68)
(SEQ ID NO: 69)


CTLA4
miR-206
(SEQ ID NO: 70)
(SEQ ID NO: 71)


PD1
miR-204
(SEQ ID NO: 72)
(SEQ ID NO: 73)


PD1
miR-204
(SEQ ID NO: 704)



PD1
miR-204
(SEQ ID NO: 705)



PD1
miR-204

(SEQ ID NO: 706)


PD1
miR-204

(SEQ ID NO: 707)


PD1
miR-204

(SEQ ID NO: 708)


PD1
miR-206
(SEQ ID NO: 74)
(SEQ ID NO: 75)


PD1
miR-206
(SEQ ID NO: 709)



PD1
miR-206
(SEQ ID NO: 710)



PD1
miR-206

(SEQ ID NO: 711)


PD1
miR-206

(SEQ ID NO: 712)


PD1
miR-206

(SEQ ID NO: 713)


PD1
miR-30a
(SEQ ID NO: 76)
(SEQ ID NO: 77)


PD1
miR-412
(SEQ ID NO: 78)
(SEQ ID NO: 79)


PD1
miR122
(SEQ ID NO: 80)
(SEQ ID NO: 81)


PD1
miR-17
(SEQ ID NO: 82)
(SEQ ID NO: 83)


PD1
miR-150
(SEQ ID NO: 74)
(SEQ ID NO: 85)


PD1
miR-486
(SEQ ID NO: 76)
(SEQ ID NO: 87)


TIGIT
miR-17
(SEQ ID NO: 88)
(SEQ ID NO: 89)


TIGIT
miR-150
(SEQ ID NO: 90)
(SEQ ID NO: 91)


TIGIT
miR-204
(SEQ ID NO: 92)
(SEQ ID NO: 93)


TIGIT
miR29b1
(SEQ ID NO: 94)
(SEQ ID NO: 95)


TIGIT
miR214
(SEQ ID NO: 96)
(SEQ ID NO: 97)


TIGIT
miR-206
(SEQ ID NO: 98)
(SEQ ID NO: 99)


TIGIT
miR-204
(SEQ ID NO: 100)
(SEQ ID NO: 101)


TIGIT
miR-22
(SEQ ID NO: 102)
(SEQ ID NO: 103)


TIGIT
miR-16
(SEQ ID NO: 104)
(SEQ ID NO: 105)


TIGIT
miR-21
(SEQ ID NO: 106)
(SEQ ID NO: 107)


TIGIT
miR-494
(SEQ ID NO: 108)
(SEQ ID NO: 109)


TIGIT
miR-142
(SEQ ID NO: 110)
(SEQ ID NO: 111)


TIGIT
miR-19
(SEQ ID NO: 112)
(SEQ ID NO: 113)


TIGIT
miR-1915
(SEQ ID NO: 114)
(SEQ ID NO: 115)


TIGIT
miR-206
(SEQ ID NO: 116)
(SEQ ID NO: 117)


TIGIT
miR-204
(SEQ ID NO: 118)
(SEQ ID NO: 119)


TIGIT
miR-22
(SEQ ID NO: 120)
(SEQ ID NO: 121)


TIGIT
miR-142
(SEQ ID NO: 122)
(SEQ ID NO: 123)


TIGIT
miR-16
(SEQ ID NO: 124)
(SEQ ID NO: 125)


TIGIT
miR-206
(SEQ ID NO: 126)
(SEQ ID NO: 127)


TIGIT
miR-204
(SEQ ID NO: 128)
(SEQ ID NO: 129)


TIGIT
miR-21
(SEQ ID NO: 130)
(SEQ ID NO: 131)


TIGIT
miR-494
(SEQ ID NO: 132)
(SEQ ID NO: 133)


TIGIT
miR-19
(SEQ ID NO: 134)
(SEQ ID NO: 135)


TIGIT
miR-1915
(SEQ ID NO: 136)
(SEQ ID NO: 137)


TIGIT
miR-204
(SEQ ID NO: 138)
(SEQ ID NO: 139)


TIGIT
miR-206
(SEQ ID NO: 140)
(SEQ ID NO: 141)


TIGIT
miR-21
(SEQ ID NO: 142)
(SEQ ID NO: 143)


TIGIT
miR-22
(SEQ ID NO: 144)
(SEQ ID NO: 145)


TIM3
miR204
(SEQ ID NO: 146)
(SEQ ID NO: 147)


TIM3
miR206
(SEQ ID NO: 148)
(SEQ ID NO: 149)


TIM3
miR17
(SEQ ID NO: 150)
(SEQ ID NO: 151)


TIM3
miR126
(SEQ ID NO: 152)
(SEQ ID NO: 153)


TIM3
miR122
(SEQ ID NO: 154)
(SEQ ID NO: 155)


TIM3
miR214
(SEQ ID NO: 156)
(SEQ ID NO: 157)


LAG3
miR-30a
(SEQ ID NO: 158)
(SEQ ID NO: 159)


LAG3
miR122
(SEQ ID NO: 160)
(SEQ ID NO: 161)


GITR
miR206
(SEQ ID NO: 162)
(SEQ ID NO: 163)


GITR
miR29b1
(SEQ ID NO: 164)
(SEQ ID NO: 165)


PIK3IP1
miR206
(SEQ ID NO: 166)
(SEQ ID NO: 167)


PIK3IP1
miR126
(SEQ ID NO: 168)
(SEQ ID NO: 169)


PIK3IP1
miR30a
(SEQ ID NO: 170)
(SEQ ID NO: 171)









In certain embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80% sequence identity with any one of SEQ ID NOs: 64-83, 85, 87-171, and 704-713 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 64-83, 85, 87-171, and 704-713. In certain such embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 704, 705, 709, and 710 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 704, 705, 709, and 710.


In certain embodiments, the miRNA targets PD-1. In certain such embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 72-83, 85, 87, and 704-713 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 72-83, 85, 87, and 704-713. In certain embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 72, 74, 76, 78, 80, 82, 704, 705, 709, and 710 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 72, 74, 76, 78, 80, 82, 704, 705, 709, and 710.


In certain embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 72-75 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 72-75. In certain such embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 72 or 74 or that is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 72 or 74.


In certain embodiments, the present invention relates to a polynucleotide comprising:

    • a) a sequence encoding a 5′ miRNA backbone sequence;
    • b) a sequence encoding a guide miRNA sequence;
    • c) a sequence encoding a stem loop sequence;
    • d) a sequence encoding a passenger miRNA sequence; and
    • e) a sequence encoding a 3′ backbone sequence.


In certain embodiments, the sequence encoding the guide miRNA sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 704, 705, 709, and 710; or is capable of hybridizing under stringent hybridization conditions to the complement of any one of such sequences.


In certain embodiments, the sequence encoding the passenger miRNA sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 65, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123, 125, 127, 129, 131, 133, 135, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 706-708, and 711-713; or is capable of hybridizing under stringent hybridization conditions to the complement of any one of such sequences.


In certain embodiments, the polynucleotide encoding a pri-miRNA encodes:

    • a) a guide miRNA sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 704, 705, 709, and 710, or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of such sequences; and
    • b) a passenger sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to, respectively, any one of SEQ ID NOs: 65, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, 113, 115, 117, 119, 121, 123, 125, 127, 129, 131, 133, 135, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 706-708, and 711-713, or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of such sequences.


In any of the foregoing embodiments, the sequences encoding the 5′ miRNA backbone sequence, the stem loop sequence, and the 3′ miRNA backbone sequence can each comprise:

    • SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively;
    • SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively;
    • SEQ ID NO: 7, SEQ ID NO: 8, and SEQ ID NO: 9, respectively;
    • SEQ ID NO: 10, SEQ ID NO: 11, and SEQ ID NO: 12, respectively;
    • SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15, respectively;
    • SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 18, respectively;
    • SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21, respectively;
    • SEQ ID NO: 22, SEQ ID NO: 23, and SEQ ID NO: 24, respectively;
    • SEQ ID NO: 25, SEQ ID NO: 26, and SEQ ID NO: 27, respectively;
    • SEQ ID NO: 28, SEQ ID NO: 29, and SEQ ID NO: 30, respectively;
    • SEQ ID NO: 31, SEQ ID NO: 32, and SEQ ID NO: 33, respectively;
    • SEQ ID NO: 34, SEQ ID NO: 35, and SEQ ID NO: 36, respectively;
    • SEQ ID NO: 37, SEQ ID NO: 38, and SEQ ID NO: 39, respectively;
    • SEQ ID NO: 40, SEQ ID NO: 41, and SEQ ID NO: 42, respectively;
    • SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively;
    • SEQ ID NO: 46, SEQ ID NO: 47, and SEQ ID NO: 48, respectively;
    • SEQ ID NO: 49, SEQ ID NO: 50, and SEQ ID NO: 51, respectively;
    • SEQ ID NO: 52, SEQ ID NO: 53, and SEQ ID NO: 54, respectively;
    • SEQ ID NO: 55, SEQ ID NO: 56, and SEQ ID NO: 57, respectively;
    • SEQ ID NO: 58, SEQ ID NO: 59, and SEQ ID NO: 60, respectively; or
    • SEQ ID NO: 61, SEQ ID NO: 62, and SEQ ID NO: 63, respectively;


      or sequences having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with such sequences, or that are capable of hybridizing under stringent hybridization conditions to the complements of such sequences.


Nucleic acids encoding exemplary non-naturally occurring pri-miRNA sequences targeting specific checkpoint inhibitors are described in Table 3. In certain embodiments, the non-naturally occurring pri-miRNA sequence may be a sequence that is capable of hybridizing under stringent hybridization conditions with the complement of any one of the sequences listed in Table 3.









TABLE 3







Nucleic acid sequences encoding non-naturally


occurring pri-miRNA sequences









miRNA
Encoded pri-



Target
miRNA(s)
DNA Sequence





CTLA4
miR204
(SEQ ID NO: 178)


PD1
miR204
(SEQ ID NO: 179)


PD1
miR206
(SEQ ID NO: 180)


TIGIT
miR17
(SEQ ID NO: 181)


TIGIT
miR150
(SEQ ID NO: 182)


TIGIT
miR204
(SEQ ID NO: 183)


TIGIT
miR-206
(SEQ ID NO: 184)


TIGIT
miR-204
(SEQ ID NO: 185)


TIGIT
miR-22
(SEQ ID NO: 186)


TIGIT
miR-16
(SEQ ID NO: 187)


TIGIT
miR-16
(SEQ ID NO: 188)


TIGIT
miR-21
(SEQ ID NO: 189)


TIGIT
miR-494
(SEQ ID NO: 190)


TIGIT
miR-142
(SEQ ID NO: 191)


TIGIT
miR-19
(SEQ ID NO: 192)


TIGIT
miR-1915
(SEQ ID NO: 193)


TIGIT
miR-206
(SEQ ID NO: 194)


TIGIT
miR-204
(SEQ ID NO: 195)


TIGIT
miR-22
(SEQ ID NO: 196)


TIGIT
miR-142
(SEQ ID NO: 197)


TIGIT
miR-16
(SEQ ID NO: 198)


TIGIT
miR-206
(SEQ ID NO: 199)


TIGIT
miR-204
(SEQ ID NO: 200)


TIGIT
miR-21
(SEQ ID NO: 201)


TIGIT
miR-494
(SEQ ID NO: 202)


TIGIT
miR-19
(SEQ ID NO: 203)


TIGIT
miR-1915
(SEQ ID NO: 204)


TIGIT
miR-204
(SEQ ID NO: 205)


TIGIT
miR-206
(SEQ ID NO: 206)


TIGIT
miR-21
(SEQ ID NO: 207)


TIGIT
miR-22
(SEQ ID NO: 208)


TIM3
miR204
(SEQ ID NO: 209)


TIM3
miR150
(SEQ ID NO: 210)


TIM3
miR30a
(SEQ ID NO: 211)


TIM3
miR206
(SEQ ID NO: 212)


TIM3
miR16
(SEQ ID NO: 213)


TIM3
miR17
(SEQ ID NO: 214)


TIM3
miR126
(SEQ ID NO: 215)


TIM3
miR122
(SEQ ID NO: 216)


TIM3
miR214
(SEQ ID NO: 217)


TIM3
miR29b1
(SEQ ID NO: 218)


TIM3
miR204
(SEQ ID NO: 219)


TIM3
miR133a1
(SEQ ID NO: 220)


LAG3
miR30a
(SEQ ID NO: 221)


LAG3
miR206
(SEQ ID NO: 222)


LAG3
miR204
(SEQ ID NO: 223)


LAG3
miR150
(SEQ ID NO: 224)


LAG3
miR17
(SEQ ID NO: 225)


LAG3
miR122
(SEQ ID NO: 226)


LAG3
miR126
(SEQ ID NO: 227)


GITR
miR30a
(SEQ ID NO: 228)


GITR
miR206
(SEQ ID NO: 229)


GITR
miR17
(SEQ ID NO: 230)


GITR
miR122
(SEQ ID NO: 231)


GITR
miR150
(SEQ ID NO: 232)


GITR
miR29
(SEQ ID NO: 233)


GITR
miR181a1
(SEQ ID NO: 234)


TIGIT
miR206
(SEQ ID NO: 235)


TIGIT
miR30a
(SEQ ID NO: 236)


TIGIT
miR133a1
(SEQ ID NO: 237)


TIGIT
miR122
(SEQ ID NO: 238)


TIGIT
miR29b1
(SEQ ID NO: 239)


TIGIT
miR214
(SEQ ID NO: 240)


PD1
miR30a
(SEQ ID NO: 241)


PD1
miR412
(SEQ ID NO: 242)


PD1
miR17
(SEQ ID NO: 243)


PD1
miR122
(SEQ ID NO: 244)


PD1
miR150
(SEQ ID NO: 245)


PD1
miR486
(SEQ ID NO: 246)


PD1
miR206
(SEQ ID NO: 247)


PD1
miR122
(SEQ ID NO: 248)


PD1
miR30a
(SEQ ID NO: 249)


CTLA4
miR206
(SEQ ID NO: 250)


CTLA4
miR26a
(SEQ ID NO: 251)


CTLA4
miR30a
(SEQ ID NO: 252)


PIK3IP1
miR206
(SEQ ID NO: 253)


PIK3IP1
miR126
(SEQ ID NO: 254)


PIK3IP1
miR30a
(SEQ ID NO: 255)


TCRa3′UTR
miR150
(SEQ ID NO: 256)


TCRa3′UTR
miR204
(SEQ ID NO: 257)


TCRa3′UTR
miR206
(SEQ ID NO: 258)


TCRa3′UTR
miR26a
(SEQ ID NO: 259)


TCRa3′UTR
miR150
(SEQ ID NO: 260)


TCRa3′UTR
miR16
(SEQ ID NO: 261)


TCRa3′UTR
miR206
(SEQ ID NO: 262)


TCRa3′UTR
miR26a
(SEQ ID NO: 263)









In certain such embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 178-263 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 178-263.


In certain embodiments, the miRNA targets PD-1. In certain such embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 179, 180, and 241-249 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 179, 180, and 241-249. In certain embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NO: 179 or 180 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 179 or 180.


Certain embodiments of the present relate to a polynucleotide that comprises nucleic acid sequences encoding at least two pri-miRNAs. The two or more pri-miRNAs may contain guide miRNA sequences that target the same target gene or the various guide miRNAs may target different genes. In the case of multiple pri-miRNAs, each design may be based on a different native miRNA backbone to reduce the likelihood of misfolding of one miRNA with another. Table 4 provides examples of nucleic acid sequences encoding one or more pri-miRNAs.









TABLE 4







Non-naturally occurring miRNA sequences


comprising two or more pri-miRNAs











miRNA
miRNA




Target
backbone
DNA Sequence







PD1
miR204 +
(SEQ ID NO: 267)




miR206



PD1 +
miR204 +
(SEQ ID NO: 268)



TIGIT
miR150



PD1 +
miR206 +
(SEQ ID NO: 269)



TIGIT
miR150



TIGIT +
miR17 +
(SEQ ID NO: 270)



PD1
miR204



TIGIT +
miR17 +
(SEQ ID NO: 271)



PD1
miR206



TIGIT +
miR204 +
(SEQ ID NO: 272)



PD1
miR206



TIGIT +
miR150 +
(SEQ ID NO: 273)



PD1
miR204



TIGIT +
miR150 +
(SEQ ID NO: 274)



PD1
miR206



TIGIT +
miR17 +
(SEQ ID NO: 275)



PD1 +
miR204 +



PD1
miR206



TIGIT +
miR17 +
(SEQ ID NO: 276)



PD1 +
miR204 +



PD1
miR206 extra




spacing 1



TIGIT +
miR17 +
(SEQ ID NO: 277)



PD1 +
miR204 +



PD1
miR206 extra




spacing 2



PD1 + PD1 +
miR204 +
(SEQ ID NO: 278)



TIGIT
miR206 +




miR17



PD1 + PD1 +
miR204 +
(SEQ ID NO: 279)



TIGIT
miR206 +




miR17 extra




spacing 1



PD1 + PD1 +
miR204 +
(SEQ ID NO: 280)



TIGIT
miR206 +




miR17 extra




spacing 2



PD1 +
miR204 +
(SEQ ID NO: 281)



CTLA4
miR26a



PD1 +
miR204 +
(SEQ ID NO: 282)



PD1
miR206



PD1 +
miR206 +
(SEQ ID NO: 283)



CTLA4
miR26a



PD1 +
miR206 +
(SEQ ID NO: 284)



CTLA4
miR204



TIGIT +
miR17 +
(SEQ ID NO: 285)



CTLA4
miR26a



TIGIT +
miR17 +
(SEQ ID NO: 286)



CTLA4
miR204



TIGIT +
miR17 +
(SEQ ID NO: 287)



PD1
miR204



TIGIT +
miR17 +
(SEQ ID NO: 288)



PD1
206



TIGIT +
miR204 +
(SEQ ID NO: 289)



CTLA4
miR26a



TIGIT +
miR204 +
(SEQ ID NO: 290)



PD1
miR206










In certain such embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 267-290 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 267-290.


In certain such embodiments, the pri-miRNA comprises at least two pre-miRNAs that target PD-1. In certain embodiments, the present invention relates to a polynucleotide comprising a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NO: 267 or that is capable of hybridizing under stringent hybridization conditions to the complement of any one of SEQ ID NOs: 267.


In certain embodiments, the nucleic acid encoding the pri-miRNA(s) is contained in the same genetic construct as that comprising one or more genes encoding protein(s) of interest (e.g., a chimeric antigen receptor, a cytokine, or a cell tag). In certain embodiments, such a genetic construct includes a nucleic acid sequence encoding a 5′ untranslated region (5′UTR) directly upstream of the gene(s) of interest and the sequence encoding the miRNA is included in the 5′UTR. In certain embodiments, such a genetic construct includes a nucleic acid sequence encoding a 3′ untranslated region (3′UTR) directly downstream of the gene(s) of interest and the sequence encoding the miRNA is included in the 3′UTR.


In embodiments wherein the sequence encoding the pri-miRNA is included in the sequence corresponding to the 5′ UTR, the transcribed RNA can include additional sequences such as splice donor, branchpoint and/or acceptor site sequences. The inclusion of splice donor, branchpoint, and acceptor sites is important for splicing of the miRNAs from the transcribed RNA. Without splicing, the highly structured miRNA sequence is likely to impede ribosome scanning to the translation initiation sequence relating to the gene of interest. Examples of sequences encoding such splice donor/acceptor sites include SEQ ID NOs: 291 and 292, sequences having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with such sequences and sequences that are capable of hybridizing with the complement of such sequences under stringent hybridization conditions.


Thus, in certain embodiments, the polynucleotide of the present invention further comprises: a) a nucleic acid sequence having at least 80% sequence identity with SEQ ID NO: 291 or that is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 291; and b) a nucleic acid sequence having at least 80% sequence identity with SEQ ID NO: 292 or that is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 292.


IV. Chimeric Antigen Receptors (CARs)

In any of the foregoing embodiments, a polynucleotide of the present disclosure can further encode a chimeric receptor, such as a chimeric antigen receptor (CAR). Thus, a polynucleotide of the present disclosure can encode miRNA(s) and a CAR as described herein. In any of the embodiments of the present disclosure, a modified immune effector cell can comprise a chimeric receptor as described herein.


A CAR is an engineered receptor that grafts an exogenous specificity onto an immune effector cell. In some instances, a CAR comprises an extracellular domain (ectodomain) that comprises an antigen-binding domain, a transmembrane domain, and an intracellular (endodomain) domain. The intracellular domain comprises an intracellular signaling domain. In certain embodiments, the extracellular domain further comprises a spacer between the antigen-binding domain and the transmembrane domain.


A. Antigen-Binding Domain


An antigen-binding domain can comprise complementary determining regions of a monoclonal antibody and/or antigen binding fragments thereof. A complementarity determining region (CDR) is a short amino acid sequence found in the variable domains of antigen receptor (e.g., immunoglobulin and T-cell receptor) proteins that bind an antigen and therefore provides the receptor with its specificity for that particular antigen. Each polypeptide chain of an antigen receptor can contain three CDRs (CDR1, CDR2, and CDR3).


In certain embodiments, the antigen-binding domain comprises an antibody, or functional fragment or variant thereof, that binds to a target antigen. The functional fragment or variant may comprise the variable domain of the heavy chain of an antibody (VH) and/or the variable domain of the light chain of an antibody (VL), or functional fragments or variants thereof. In certain embodiments, the antigen-binding domain comprises a Fv, Fab, Fab2, Fab′, F(ab′)2, or F(ab′) 3 fragment of an antibody. In certain embodiments, the antigen-binding domain comprises a scFv, sc(Fv)2, a dsFv, a diabody, a minibody, a nanobody, or binding fragments thereof. In certain embodiments, the antigen-binding domain further comprises an Fc fragment of an antibody, for example it may comprise an scFv linked with an Fc fragment.


In some embodiments, the CAR targets an antigen that is elevated in cancer cells, in autoimmune cells, or in cells that are infected by a virus, bacteria or parasite. Pathogens that may be targeted include, without limitation, Plasmodium, trypanosome, Aspergillus, Candida, Hepatitis A, Hepatitis B, Hepatitis C, HSV, HPV, RSV, EBV, CMV, JC virus, BK virus, or Ebola pathogens. Autoimmune diseases can include graft-versus-host disease, rheumatoid arthritis, lupus, celiac disease, Crohn's disease, Sjogren Syndrome, polymyalgia rheumatic, multiple sclerosis, neuromyelitis optica, ankylosing spondylitis, Type 1 diabetes, alopecia areata, vasculitis, temporal arteritis, bullous pemphigoid, psoriasis, pemphigus vulgaris, and autoimmune uveitis.


The pathogen recognized by a CAR may be essentially any kind of pathogen, but in some embodiments, the pathogen is a fungus, bacteria, or virus. Exemplary viral pathogens include those of the families of Adenoviridae, Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Respiratory Syncytial Virus (RSV), JC virus, BK virus, HPV, HSV, HHV family of viruses, Hepatitis family of viruses, Picornaviridae, Herpesviridae, Hepadnaviridae, Flaviviridae, Retroviridae,


Orthomyxoviridae, Paramyxoviridae, Papovaviridae, Polyomavirus, Rhabdoviridae, and Togaviridae. Exemplary pathogenic viruses cause smallpox, influenza, mumps, measles, chickenpox, ebola, and rubella. Exemplary pathogenic fungi include Candida, Aspergillus, Cryptococcus, Histoplasma, Pneumocystis, and Stachybotrys. Exemplary pathogenic bacteria include Streptococcus, Pseudomonas, Shigella, Campylobacter, Staphylococcus, Helicobacter, E. coli, Rickettsia, Bacillus, Bordetella, Chlamydia, Spirochetes, and Salmonella. In some embodiments, the pathogen receptor Dectin-1 may be used to generate a CAR that recognizes the carbohydrate structure on the cell wall of fungi such as Aspergillus. In another embodiment, CARs can be made based on an antibody recognizing viral determinants (e.g., the glycoproteins from CMV and Ebola) to interrupt viral infections and pathology.


In some embodiments, a CAR described herein comprises an antigen-binding domain that binds to an epitope on B7H4, BCMA, BTLA, CAIX, CA125, CCR4, CD3, CD4, CD5, CD7, CD16, CD19, CD20, CD22, CD24, CD25, CD28, CD30, CD33, CD38, CD40, CD44, CD44v6, CD44v7/v8, CD47, CD52, CD56, CD70, CD79b, CD80, CD81, CD86, CD123, CD133, CD137, CD138, CD151, CD171, CD174, CD276, CEA, CEACAM6, CLL-1, c-MET, CS1, CSPG4, CTLA-4, DLL3, EDB-F, EGFR, EGFR2, EGFRvIII, EGP-2, EGP-40, EphA2, FAP, FLT1, FLT4, Folate-binding Protein, Folate Receptor, Folate receptor α, α-Folate receptor, Frizzled, GD2, GD3, GHR, GHRHR, GITR, GPC3, Gp100, gp130, HBV antigens, HER1, HER2, HER3, HER4, HER1/HER3, h5T4, HPV antigens, HVEM, IGF1R, IgKAppa, IL-1-RAP, IL-2R, IL6R, IL-11Rα, IL-13R-a2, KDR, KRASG12V, LewisA, LewisY, L1-CAM, LIFRP, LRP5, LTPR, MAGE-A, MAGE-A1, MAGE-A10, MAGE-A3, MAGEA3/A6, MAGE-A4, MAGE-A6, MART-1, MCAM, mesothelin, PSCA, Mucins such as MUC1, MUC-4 or MUC16, NGFR, NKG2D, Notch-1-4, NY-ESO-1, O-acetylGD2, O-acetylGD3, OX40, P53, PD1, PDE10A, PD-L1, PD-L2, PMSA, PRAME, PSCA, PSMA, PTCH1, RANK, Robol, ROR1, ROR1R, ROR-2, TACI, TAG-72, TCRα, TCRp, TGF, TGFBeta, TGFBeta-II, TGFBR1, TGFBR2, Titin, TLR7, TLR9, TNFR1, TNFR2, TNFRSF4, TRBC1, TWEAK-R, VEGF, VEGF-R2, or WT-1.


In some embodiments, a CAR described herein comprises an antigen-binding domain that binds to an epitope on CD19, CD33, MUC1, MUC16, or ROR1. In some instances, a CAR described herein comprises an antigen-binding domain that binds to an epitope on CD19. In some cases, a CAR described herein comprises an antigen-binding domain that binds to an epitope on CD33. In some instances, a CAR described herein comprises an antigen-binding domain that binds to an epitope on MUC1. In some instances, a CAR described herein comprises an antigen-binding domain that binds to an epitope on MUC16. In some instances, a CAR described herein comprises an antigen-binding domain that binds to an epitope on ROR1. In further embodiments, a CAR described herein comprises an autoantigen or an antigen binding domain that binds to an epitope on HLA-A2, myelin oligodendrocyte glycoprotein (MOG), factor VIII (FVIII), MAdCAM1, SDF1, or collagen type II.


Antigen binding can be assessed by flow cytometry or a cell based assay or any other equivalent assay. Cell based assays may utilize a cell type expressing antigen of interest on the surface to assess antigen-binding. An antigen or a fragment thereof expressed as a soluble protein can be utilized to assess antigen-binding using flow cytometry or similar assay. Improvements in antigen-binding may be indirectly assessed by functional measurement of antigen-binding domain or a chimeric receptor. For example, improved antigen-binding of a chimeric receptor or a CAR, as described herein, can be measured by increased specific cytotoxicity against target cells expressing the antigen.


Cell surface expression level of a polypeptide of the present disclosure can be assessed, for example, using a flow cytometry based assay. Improved expression of an antigen-binding polypeptide can be measured as percentage of analyzed cells expressing said antigen-binding polypeptide or alternatively as average density of said antigen-binding polypeptide on the surface of a cell. Additional suitable methods that can be used for assessing cell surface expression of the antigen-binding polypeptides described herein include western blotting or any other equivalent assay.


1. CD19-specific Antigen-Binding Domain


CD19 is a cell surface glycoprotein of the immunoglobulin superfamily and is found predominately in malignant B-lineage cells. In some instances, CD19 has also been detected in solid tumors such as pancreatic cancer, liver cancer, and prostate cancer.


In some embodiments, the CAR comprises a CD19-specific antigen-binding domain, in which the antigen-binding domain comprises a F(ab′)2, Fab′, Fab, Fv, or scFv. In some embodiments, the antigen-binding domain recognizes an epitope on CD19 that is also recognized by FMC63. In some embodiments the scFv and/or VH/VL domains is/are derived from FMC63. FMC63 generally refers to a mouse monoclonal IgG1 antibody raised against Nalm-1 and -16 cells expressing CD19 of human origin (Ling, N. R., el al. (1987). Leucocyte typing III. 302).


In some embodiments, the antigen-binding domain recognizes an epitope on CD19 that is also recognized by JCAR014, JCAR015, JCAR017, or 19-28z CAR (Juno Therapeutics).


In some embodiments, the antigen-binding domain comprises a scFv antigen-binding domain that recognizes an epitope on CD19 that is also recognized by JCAR014, JCAR015, JCAR017, or 19-28z CAR (Juno Therapeutics).


In some embodiments, the antigen-binding domain comprises an anti-CD19 antibody described in U.S. Patent Application Publication No. 2016/0152723. In some embodiments, the antigen-binding domain comprises an anti-CD19 antibody described in International Patent Application Publication No. WO2015/123642. In some embodiments, the antigen-binding domain comprises an anti-CD19 scFv derived from clone FMC63 (Nicholson et al., Construction and characterization of a functional CD19 specific single chain Fv fragment for immunotherapy of B lineage leukemia and lymphoma., Mol. Immunol. 34:1157-1165 (1997)).


In some embodiments, the antigen-binding domain recognizes an epitope on CD19 that is also recognized by KTE-C19 (Kite Pharma, Inc.).


In some embodiments, the antigen-binding domain comprises a scFv antigen-binding domain, and the antigen-binding domain recognizes an epitope on CD19 that is also recognized by KTE-C19.


In some embodiments, the antigen-binding domain comprises an anti-CD19 antibody described in International Patent Application Publication No. WO2015/187528 or fragment or variant thereof.


In some embodiments, the antigen-binding domain recognizes an epitope on CD19 that is also recognized by CTL019 (Novartis).


In some embodiments, the antigen-binding domain comprises a scFv antigen-binding domain, and the antigen-binding domain recognizes an epitope on CD19 that is also recognized by CTL019.


In some embodiments, the antigen-binding domain recognizes an epitope on CD19 that is also recognized by UCART19 (Cellectis).


In some embodiments, the antigen-binding domain comprises a scFv antigen-binding domain, and the antigen-binding domain recognizes an epitope on CD19 that is also recognized by UCART19.


In some embodiments, the antigen-binding domain recognizes an epitope on CD19 that is also recognized by BPX-401 (Bellicum).


In some embodiments, the antigen-binding domain comprises a scFv antigen-binding domain, and the antigen-binding domain recognizes an epitope on CD19 that is also recognized by BPX-401.


In some cases, the antigen-binding domain recognizes an epitope on CD19 that is also recognized by blinatumomab (Amgen), coltuximabravtansine (ImmunoGen Inc./Sanofi-aventis), MOR208 (Morphosys AG/Xencor Inc.), MEDI-551 (Medimmune), denintuzumabmafodotin (Seattle Genetics), B4 (or DI-B4) (Merck Serono), taplitumomabpaptox (National Cancer Institute), XmAb 5871 (Amgen/Xencor, Inc.), MDX-1342 (Medarex) or AFM11 (Affimed).

  • In some embodiments, the antigen-binding domain comprises a F(ab′) 2, Fab′, Fab, Fv, or scFv, and the antigen-binding domain recognizes an epitope on CD19 that is also recognized by blinatumomab (Amgen), coltuximabravtansine (ImmunoGen Inc./Sanofi-aventis), MOR208 (Morphosys AG/Xencor Inc.), MEDI-551 (Medimmune), denintuzumabmafodotin (Seattle Genetics), B4 (or DI-B4) (Merck Serono), taplitumomabpaptox (National Cancer Institute), XmAb 5871 (Amgen/Xencor, Inc.), MDX-1342 (Medarex) or AFM11 (Affimed).


In some cases, the antigen-binding domain comprises a scFv antigen-binding domain, and the antigen-binding domain recognizes an epitope on CD19 that is also recognized by FMC63, blinatumomab (Amgen), coltuximabravtansine (ImmunoGen Inc./Sanofi-aventis), MOR208 (Morphosys AG/Xencor Inc.), MEDI-551 (Medimmune), denintuzumabmafodotin (Seattle Genetics), B4 (or DI-B4) (Merck Serono), taplitumomabpaptox (National Cancer Institute), XmAb 5871 (Amgen/Xencor, Inc.), MDX-1342 (Medarex) or AFM11 (Affimed).


2. CD33-specific Antigen-Binding Domain


“CD33,” is a 67 kDa single pass transmembrane glycoprotein and is a member of the sialic acid-binding immunoglobulin-like lectins (Siglecs) super-family. CD33 is characterized by a V-set Ig-like domain responsible for sialic acid binding and a C2-set Ig-like domain in its extracellular domain. CD33 is expressed in myeloid lineage cells and has also been detected in lymphoid cells. Alternative splicing of CD33 mRNA leads to a shorter isoform (CD33m) lacking the V-set Ig-like domain as well as the disulfide bond linking the V- and C2-set Ig-like domains. In healthy subjects, CD33 is primarily expressed as a myeloid differentiation antigen found on normal multipotent myeloid precursors, unipotent colony-forming cells, monocytes and maturing granulocytes. CD33 is expressed on more than 80% of myeloid leukemia cells but not on normal hematopoietic stem cells or mature granulocytes. (Andrews, R. et al., The L4F3 antigen is expressed by unipotent and multipotent colony-forming cells but not by their precursors, Blood, 68(5):1030-5 (1986)). CD33 has been reported to be expressed on malignant myeloid cells, activated T cells and activated NK cells and is found on at least a subset of blasts in the vast majority of AML patients (Pollard, J. et al., Correlation of CD33 expression level with disease characteristics and response to gemtuzumab ozogamicin containing chemotherapy in childhood AML, Blood, 119(16):3705-11(2012)). In addition to broad expression on AML blasts, CD33 may be expressed on stem cells underlying AML.


In some embodiments, the antigen-binding domain of a CAR described herein is specific to CD33 (CD33 CAR). The CD33-specific CAR, when expressed on the cell surface, redirects the specificity of T cells to human CD33. In some embodiments, the antigen-binding domain comprises a single chain antibody fragment (scFv) comprising a variable domain light chain (VL) and variable domain heavy chain (VH) of a target antigen specific monoclonal anti-CD33 antibody joined by a flexible linker, such as a glycine-serine linker or a Whitlow linker. In some embodiments, the scFv is M195, m2H12, DRB2, and/or My9-6. In some embodiments, the scFv is humanized, for example, hM195. In some embodiments, the antigen-binding domain may comprise VH and VL that are directionally linked, for example, from N to C terminus, VH-linker-VL or VL-linker-VH. In some embodiments, the antigen-binding domain comprises a F(ab′)2, Fab′, Fab, Fv, or scFv that binds CD33. In some embodiments, the antigen-binding region recognizes an epitope on CD33 that is also recognized by Lintuzumab (Seattle Genetics), BI 836858 (Boehringer Ingelheim).


In some embodiments, a CAR described herein comprises an antigen-binding domain comprising a VL polypeptide.


In certain embodiments, the antigen-binding domain comprises a VL domain comprising the amino acid sequence of SEQ ID NO: 293 (hM195 VL domain), SEQ ID NO: 296 (M2H12 VL domain), SEQ ID NO: 298 (DRB2 VL domain), SEQ ID NO: 300 (My9-6 VL domain) or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than any one of the aforementioned sequences by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of any one of SEQ ID NOs: 293, 296, 298, and 300, and/or is a conservatively-substituted variant of any one of such sequences.


In certain embodiments, the antigen-binding domain comprises a VL domain encoded by SEQ ID NO: 301 (nucleic acid encoding the VL domain for hM195). In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 301; hybridizes under stringent hybridization conditions with the complement of the nucleic acid sequence of SEQ ID NO: 301; or is a codon degenerate variant of SEQ ID NO: 301.


In some embodiments, a CAR described herein comprises an antigen-binding domain comprising a VH polypeptide.


In certain embodiments, the antigen-binding domain comprises a VH domain comprising the amino acid sequence of SEQ ID NO: 294 (hM195 VH domain), SEQ ID NO: 295 (M2H12 VH domain), SEQ ID NO: 297 (DRB2 VH domain), SEQ ID NO: 299 (My9-6 VH domain) or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than any one of the aforementioned sequences by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of any one of SEQ ID NOs: 294, 295, 297, or 299, and/or is a conservatively-substituted variant of the amino acid sequence of any one of SEQ ID NOs: 294, 295, 297, or 299.


In certain embodiments, the antigen-binding domain comprises a VH domain encoded by SEQ ID NO: 302 (nucleic acid encoding the VH domain for hM195), or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 302; hybridizes under stringent hybridization conditions with the complement of the nucleic acid sequence of SEQ ID NO: 302; or is a codon degenerate variant of SEQ ID NO: 302.


In some embodiments, the antigen-binding domain can comprise VH and VL that are directionally linked, for example, from N to C terminus, VH-linker-VL or VL-linker-VH. Any linker as described herein can be used to link the VH and VL domains.


In certain embodiments, the antigen-binding domain comprises scFv. In certain such embodiments, the domain comprises the amino acid sequence of SEQ ID NO: 303, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than any one of the aforementioned sequence by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 303, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 303.


In certain embodiments, the antigen-binding domain is encoded by SEQ ID NO: 304, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 304; hybridizes under stringent hybridization conditions with the complement of the nucleic acid sequence of SEQ ID NO: 304; or is a codon degenerate variant of SEQ ID NO: 304.


3. MUC1-specific Antigen-Binding Domain


In one embodiment, the antigen-binding domain binds to an epitope on MUC1. In some embodiments, the anti-MUC1 antibody fragment is a Fab, Fab2, (Fab′)2, Fv, (Fv)2, scFv, scFv-FC, FC, diabody, triabody, or minibody of the anti-MUC1. In some embodiments, the anti-MUC1 antibody fragment is a single-domain antibody of the anti-MUC1 antibody. In some embodiments, the single-domain antibody is a VNAR or VHH fragment of the anti-MUC1 antibody.


In some embodiments, a CAR described herein comprises an antigen-binding domain comprising a VL polypeptide of MUC1.


In certain embodiments, the antigen-binding domain comprises a VL domain comprising the amino acid sequence of any one of SEQ ID NOs: 310-314, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of any one of SEQ ID NOs: 310-314 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of any one of SEQ ID NOs: 310-314, and/or is a conservatively-substituted variant of the amino acid sequence of any one of SEQ ID NOs: 310-314.


In some embodiments, a CAR described herein comprises an antigen-binding domain comprising a VH polypeptide of MUC1.


In certain embodiments, the antigen-binding domain comprises a VH domain comprising the amino acid sequence of any one of SEQ ID NOs: 305-309, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of any one of SEQ ID NOs: 305-309 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of any one of SEQ ID NOs: 305-309, and/or is a conservatively-substituted variant of the amino acid sequence of any one of SEQ ID NOs: 305-309.


In some embodiments, the antigen binding moiety can comprise VH and VL that are directionally linked, for example, from N to C terminus, VH-linker-VL or VL-linker-VH. Any linker as described herein can be used to link the VH and VL domains.


4. MUC16-Specific Antigen-Binding Domain


In some embodiments, the antigen binding moiety of a CAR described herein is specific to MUC16 (MUC16 CAR). The MUC16-specific CAR, when expressed on the cell surface, redirects the specificity of T cells to human MUC16.


In certain embodiments, the antigen-binding domain comprises a VL domain comprising the amino acid sequence of any one of SEQ ID NOs: 329, 331, 333, 335, 337, 339, 341, 662, 664, 666, 668, 670, 688, 690, 692, 694, 696, 698, and 700, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of any one of the aforementioned sequences by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of any one of SEQ ID NOs: 329, 331, 333, 335, 337, 339, 341, 662, 664, 666, 668, 670, 688, 690, 692, 694, 696, 698, and 700; and/or is a conservatively-substituted variant of the amino acid sequence of any one of SEQ ID NOs: 329, 331, 333, 335, 337, 339, 341, 662, 664, 666, 668, 670, 688, 690, 692, 694, 696, 698, and 700.


In certain embodiments, the antigen-binding domain comprises a VL domain encoded by any one of SEQ ID NOs: 330, 332, 334, 336, 338, 340, 342, 663, 665, 667, 669, 671, 689, 691, 693, 695, 697, 699, and 701, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 330, 332, 334, 336, 338, 340, 342, 663, 665, 667, 669, 671, 689, 691, 693, 695, 697, 699, and 701; or hybridizes under stringent hybridization conditions with the complement of any one of SEQ ID NOs: 330, 332, 334, 336, 338, 340, 342, 663, 665, 667, 669, 671, 689, 691, 693, 695, 697, 699, and 701.


In certain embodiments, the antigen-binding domain comprises a VL domain comprising the amino acid sequence of SEQ ID NO: 692, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 692 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 692; and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 692.


In certain embodiments, the antigen-binding domain comprises a VL domain encoded by SEQ ID NO: 693, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 693; or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 693.


In certain embodiments, the antigen-binding domain comprises a VH domain comprising the amino acid sequence of any one of SEQ ID NOs: 315, 317, 319, 321, 323, 325, 327, 648, 650, 652, 654, 656, 658, 660, 672, 674, 676, 678, 680, 682, 684, and 686, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of any one of the aforementioned sequences by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 315, 317, 319, 321, 323, 325, 327, 648, 650, 652, 654, 656, 658, 660, 672, 674, 676, 678, 680, 682, 684, and 686; and/or is a conservatively-substituted variant of the amino acid sequence of any one of SEQ ID NOs: 315, 317, 319, 321, 323, 325, 327, 648, 650, 652, 654, 656, 658, 660, 672, 674, 676, 678, 680, 682, 684, and 686.


In certain embodiments, the antigen-binding domain comprises a VH domain encoded by any one of SEQ ID NOs: 316, 318, 320, 322, 324, 326, 328, 649, 651, 653, 655, 657, 659, 661, 673, 675, 677, 679, 681, 683, 685, and 687, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 316, 318, 320, 322, 324, 326, 328, 649, 651, 653, 655, 657, 659, 661, 673, 675, 677, 679, 681, 683, 685, and 687; or hybridizes under stringent hybridization conditions with the complement of any one of SEQ ID NOs: 316, 318, 320, 322, 324, 326, 328, 649, 651, 653, 655, 657, 659, 661, 673, 675, 677, 679, 681, 683, 685, and 687.


In certain embodiments, the antigen-binding domain comprises a VH domain comprising the amino acid sequence of SEQ ID NO: 676, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 676 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 676; and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 676.


In certain embodiments, the antigen-binding domain comprises a VH domain encoded by SEQ ID NO: 677, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 677; or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 677.


In some embodiments, the antigen binding moiety can comprise VH and VL that are directionally linked, for example, from N to C terminus, VH-linker-VL or VL-linker-VH. Any linker as described herein can be used to link the VH and VL domains.


In some embodiments, the antigen-binding domain comprises a single chain antibody fragment (scFv) comprising a variable domain light chain (VL) and variable domain heavy chain (VH) of a target antigen specific monoclonal anti-MUC16 antibody joined by a flexible linker, such as a glycine-serine linker or a Whitlow linker. In certain embodiments, the scFv comprises the VH and VL from MUC16-3 and a linker. In certain such embodiments, the scFv comprises the amino acid sequence of SEQ ID NO: 343, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 343 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 343; and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 343.


In certain embodiments, the antigen-binding domain comprises a scFv encoded by SEQ ID NO: 344, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 344; or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 344.


5. ROR1-Specific Antigen-Binding Domain


In some embodiments, a CAR described herein comprises an antigen-binding domain comprising the VLdomain of an anti-ROR1 antibody.


In certain such embodiments, the antigen-binding domain may comprise an amino acid sequence of any one of SEQ ID NOs: 347, 351, 355, 359, 363, 367, 371, 375, 379, 383, 387, 391, 395, 399, 403, 407, 411, 415, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, and 463, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than any one of the aforementioned sequences by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 347, 351, 355, 359, 363, 367, 371, 375, 379, 383, 387, 391, 395, 399, 403, 407, 411, 415, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, and 463, and/or is a conservatively-substituted variant of the amino acid sequence of any one of SEQ ID NOs: 347, 351, 355, 359, 363, 367, 371, 375, 379, 383, 387, 391, 395, 399, 403, 407, 411, 415, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, and 463.


In certain embodiments, the antigen-binding domain comprises a VL domain encoded by any one of SEQ ID NOs: 348, 352, 356, 360, 364, 368, 372, 376, 380, 384, 388, 392, 396, 400, 404, 408, 412, 416, 420, 424, 428, 432, 436, 440, 444, 448, 452, 456, 460, and 464, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 348, 352, 356, 360, 364, 368, 372, 376, 380, 384, 388, 392, 396, 400, 404, 408, 412, 416, 420, 424, 428, 432, 436, 440, 444, 448, 452, 456, 460, and 464; hybridizes under stringent hybridization conditions with the complement of any one of SEQ ID NOs: 348, 352, 356, 360, 364, 368, 372, 376, 380, 384, 388, 392, 396, 400, 404, 408, 412, 416, 420, 424, 428, 432, 436, 440, 444, 448, 452, 456, 460, and 464; or is a codon degenerate variant of any one of SEQ ID NOs: 348, 352, 356, 360, 364, 368, 372, 376, 380, 384, 388, 392, 396, 400, 404, 408, 412, 416, 420, 424, 428, 432, 436, 440, 444, 448, 452, 456, 460, and 464.


In some embodiments, a CAR described herein comprises the VH domain of an anti-ROR1 antibody.


In certain such embodiments, the antigen-binding domain may comprise an amino acid sequence of any one of SEQ ID NOs: 349, 353, 357, 361, 365, 369, 373, 377, 381, 385, 389, 393, 397, 401, 405, 409, 413, 417, 421, 425, 429, 433, 437, 441, 445, 449, 453, 457, and 461, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than any one of the aforementioned sequences by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 349, 353, 357, 361, 365, 369, 373, 377, 381, 385, 389, 393, 397, 401, 405, 409, 413, 417, 421, 425, 429, 433, 437, 441, 445, 449, 453, 457, and 461, and/or is a conservatively-substituted variant of the amino acid sequence of any one of SEQ ID NOs: 349, 353, 357, 361, 365, 369, 373, 377, 381, 385, 389, 393, 397, 401, 405, 409, 413, 417, 421, 425, 429, 433, 437, 441, 445, 449, 453, 457, and 461.


In certain embodiments, the antigen-binding domain comprises a VH domain encoded by any one of SEQ ID NOs: 350, 354, 358, 362, 366, 370, 374, 378, 382, 386, 390, 394, 398, 402, 406, 410, 414, 418, 422, 426, 430, 434, 438, 442, 446, 450, 454, 458, and 462, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 350, 354, 358, 362, 366, 370, 374, 378, 382, 386, 390, 394, 398, 402, 406, 410, 414, 418, 422, 426, 430, 434, 438, 442, 446, 450, 454, 458, and 462; hybridizes under stringent hybridization conditions with the complement of any one of SEQ ID NOs: 350, 354, 358, 362, 366, 370, 374, 378, 382, 386, 390, 394, 398, 402, 406, 410, 414, 418, 422, 426, 430, 434, 438, 442, 446, 450, 454, 458, and 462; or is a codon degenerate variant of any one of SEQ ID NOs: 350, 354, 358, 362, 366, 370, 374, 378, 382, 386, 390, 394, 398, 402, 406, 410, 414, 418, 422, 426, 430, 434, 438, 442, 446, 450, 454, 458, and 462.


In certain embodiments, the antigen-binding domain comprises at least one CDR selected from those comprising the amino acid sequence of any one of the CDRs that bind ROR1 or a functional fragment or variant thereof.


In certain embodiments, the antigen-binding domain comprises the amino acid sequence of any one of SEQ ID NOs: 715-725, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the sequence of any one of the aforementioned sequences by at most 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 715-725, and/or is a conservatively-substituted variant of any one of SEQ ID NOs: 715-725.


In certain embodiments, the antigen-binding domain comprises a VH domain comprising the amino acid sequences of SEQ ID NO: 715, SEQ ID NO: 716, and SEQ ID NO: 717.


In certain embodiments, the antigen-binding domain comprises a VH domain comprising the amino acid sequences of SEQ ID NO: 718, SEQ ID NO: 719, and SEQ ID NO: 720.


In certain embodiments, the antigen-binding domain comprises a VL domain comprising the amino acid sequences of SEQ ID NO: 721, SEQ ID NO: 722, and SEQ ID NO: 723.


In certain embodiments, the antigen-binding domain comprises a VL domain comprising the amino acid sequences of SEQ ID NO: 724, SEQ ID NO: 725, and SEQ ID NO: 723.


In certain embodiments, the antigen-binding domain comprises both the aforementioned VH and VL domains.


In certain embodiments, the antigen-binding domain comprises a variable heavy chain domain comprising the amino acid sequence of SEQ ID NO: 349 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 349 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 349, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 349.


In certain embodiments, the antigen-binding domain comprises a VH domain encoded by SEQ ID NO: 350, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 350; hybridizes under stringent hybridization conditions with the complement SEQ ID NO: 350; or is a codon degenerate variant of SEQ ID NO: 350.


In certain embodiments, the antigen-binding domain comprises a variable light chain domain comprising the amino acid sequence of SEQ ID NO: 387 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 387 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 387, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 387.


In certain embodiments, the antigen-binding domain comprises a VL domain encoded by SEQ ID NO: 388, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 388; hybridizes under stringent hybridization conditions with the complement SEQ ID NO: 388; or is a codon degenerate variant of SEQ ID NO: 388.


In certain embodiments, the antigen-binding domain comprises a variable heavy chain domain comprising the amino acid sequence of SEQ ID NO: 349 or a functional fragment or variant thereof, wherein the variable heavy chain domain comprises, in N-terminal to C-terminal order, the sequences of SEQ ID NO: 715, SEQ ID NO: 716, and SEQ ID NO: 717. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 349 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 349, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 349.


In certain embodiments, the antigen-binding domain comprises a variable heavy chain domain comprising the amino acid sequence of SEQ ID NO: 349 or a functional fragment or variant thereof, wherein the variable heavy chain domain comprises, in N-terminal to C-terminal order, the sequences of SEQ ID NO: 718, SEQ ID NO: 719, and SEQ ID NO: 720. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 349 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 349, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 349.


In certain embodiments, the antigen-binding domain comprises a variable light chain domain comprising the amino acid sequence of SEQ ID NO: 387 or a functional fragment or variant thereof, wherein the variable heavy chain domain comprises, in N-terminal to C-terminal order, the sequences of SEQ ID NO: 721, SEQ ID NO: 722, and SEQ ID NO: 723. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 387 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 387, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 387.


In certain embodiments, the antigen-binding domain comprises a variable light chain domain comprising the amino acid sequence of SEQ ID NO: 387 or a functional fragment or variant thereof, wherein the variable heavy chain domain comprises, in N-terminal to C-terminal order, the sequences of SEQ ID NO: 724, SEQ ID NO: 725, and SEQ ID NO: 723. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 387 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 387, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 387.


In certain embodiments, the antigen-binding domain comprises a variable heavy chain domain comprising the amino acid sequence of SEQ ID NO: 726 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 726 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 726, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 726.


In certain embodiments, the antigen-binding domain comprises a variable light chain domain comprising the amino acid sequence of SEQ ID NO: 727 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 727 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 727, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 727.


In certain embodiments, the antigen-binding domain comprises a variable heavy chain domain comprising the amino acid sequence of SEQ ID NO: 728 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 728 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 728, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 728.


In certain embodiments, the antigen-binding domain comprises a variable light chain domain comprising the amino acid sequence of SEQ ID NO: 729 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 729 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 729, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 729.


In certain embodiments, the antigen-binding domain comprises a linker that links the VH and VL domains. In certain such embodiments, the linker comprises: (a) the amino acid sequence of SEQ ID NO: 424 ((G4S)3) or a conservatively-substituted variant thereof; or (b) an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 424. In certain such embodiments, the linker is encoded by SEQ ID NO: 425, hybridizes under stringent conditions to the complement of SEQ ID NO: 425, or is a codon degenerate version of SEQ ID NO: 425. Any linker as described herein can be used to link the VH and VL domains.


In certain embodiments, the antigen-binding domain comprises an scFv. In certain such embodiments, the domain comprises an amino acid sequence of SEQ ID NO: 465, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 465 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 465, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 465.


In certain embodiments, the antigen-binding domain comprises an scFv encoded by SEQ ID NO: 466, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 466; hybridizes under stringent hybridization conditions with the complement SEQ ID NO: 466; or is a codon degenerate variant of SEQ ID NO: 466.


6. EGFRvIII-Specific Antigen-Binding Domain


In another embodiment, a CAR described herein is a EGFRvIII-specific CAR. “EGFRvIII”, “EGFR variant III”, “EGFR type III mutant”, “EGFR.D2-7” or “de2-7EGFR” is a mutated form of epidermal growth factor receptor (EGFR; ErbB-1; HER1), a transmembrane protein that is a receptor for members of the epidermal growth factor (EGF) family of extracellular protein ligands in human and non-human subjects. EGFRvIII is characterized by a deletion of exons 2-7 of the wild type EGFR gene, which results in an in-frame deletion of 267 amino acids in the extracellular domain of the full length wild type EGFR protein. EGFRvIII also contains a novel glycine residue inserted at the fusion junction compared to wild type EGFR. The truncated receptor EGFRvIII is unable to bind any known EGFR ligand; however, it shows constitutive tyrosine kinase activity. This constitutive activation is important to its pro-oncogenic effect. A kinase-deficient EGFRvIII is unable to confer a similar oncogenic advantage. EGFRvIII is highly expressed in glioblastoma (GBM) and can be detected in some other solid tumor types but not in normal tissues.


In some embodiments, the antigen binding moiety of a CAR described herein is specific to EGFRvIII (EGFRvIII CAR). The EGFRvIII-specific CAR, when expressed on the cell surface, redirects the specificity of T cells to human EGFRvIII. In embodiments, the antigen binding domain comprises a single chain antibody fragment (scFv) comprising a variable domain light chain (VL) and variable domain heavy chain (VH) of a target antigen specific monoclonal anti-EGFRvIII antibody joined by a flexible linker, such as a glycine-serine linker or a Whitlow linker. In some embodiments, the antigen binding moiety may comprise VH and VL that are directionally linked, for example, from N to C terminus, VH-linker-VL or VL-linker-VH.


B. Spacer


In some embodiments, a chimeric antigen receptor of the present disclosure further includes a spacer that is used to link the antigen-binding domain to the transmembrane domain. In some embodiments, the spacer is flexible enough to allow the antigen-binding domain to orient in different directions to facilitate antigen recognition.


In certain embodiments, a chimeric antigen receptor comprising a spacer has improved functional activity compared to an otherwise identical antigen-binding polypeptide lacking the spacer. In certain embodiments, a chimeric antigen receptor comprising a spacer has increased expression on a cell surface compared to an otherwise identical polypeptide lacking the spacer. In an embodiment, a chimeric antigen receptor comprising a spacer is a polypeptide that, were it not for the spacer, would not express on the cell membrane surface and/or would not be able to bind its target due to lack of proximity or steric hindrance.


In certain embodiments, the spacer comprises a stalk region, for example a hinge region from an antibody. In some instances, the stalk region comprises the hinge region from an IgG, for example IgG1. In alternative instances, the stalk region comprises the CH2CH3 region of immunoglobulin and, optionally, portions of CD3. In some cases, the stalk region comprises a CD8α hinge region (SEQ ID NO: 426), an IgG4-Fc 12 amino acid hinge region (SEQ ID NO: 631), or an IgG4 hinge region as described in WO2016/073755. The stalk region can be an extracellular portion of the CAR that links the antigen-binding domain to the cell surface and/or transmembrane region.


In some embodiments, the stalk region can be from about 20 to about 300 amino acids in length. In some cases, the stalk region can be about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60 or greater amino acids in length. In other cases, the stalk region can be about: 100, 125, 150, 175, 200, 225, 250, 275 or 300 amino acids in length. In some cases a stalk region can be less than 20 amino acids in length.


In some embodiments, the stalk region comprises a CD8α hinge domain, a CD28 hinge domain or a CTLA-4 hinge domain, or a functional fragment or variant thereof.


In certain embodiments, the stalk region comprises a CD8α hinge region, or a functional fragment or variant thereof. In certain such embodiments, the spacer comprises the amino acid sequence of SEQ ID NO: 426 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 467 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 467, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 467.


In certain embodiments, the CD8α hinge region, or functional fragment or variant thereof, is encoded by SEQ ID NO: 468, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 468, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 468, or is a codon degenerate variant of SEQ ID NO: 468.


In some embodiments, the stalk region can be capable of dimerizing with a homologous stalk region of a second CAR.


In certain embodiments, in addition to a stalk region, the spacer may comprise one or more stalk extension region(s). In certain embodiments, the stalk extension region is a polypeptide that is homologous to the stalk region. For example, it may comprise at least one amino acid residue substitution as compared with the stalk region. In some embodiments, the stalk region comprises a sequence with at least about 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity to the stalk region to which it is attached, for example a CD8α hinge domain, a CD28 hinge domain, or a CTLA-4 hinge domain.


In some embodiments, the spacer comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 stalk extension regions.


In certain embodiments, the stalk region can be linked to the stalk extension region by way of a linker.


In certain embodiments, the stalk extension region can comprise about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times the length of the stalk region as measured by number of amino acids.


In some embodiments, the stalk region comprises at least one dimerization site. In certain embodiments, the stalk region may comprise one or more dimerization sites to form homo- or hetero-dimerized chimeric polypeptides. In other embodiments, the stalk region or one or more stalk extension regions may contain mutations that eliminate dimerization sites altogether.


In certain embodiments, the stalk extension region has at least one fewer dimerization site as compared to a stalk region. For example, if a stalk region comprises two dimerization sites, a stalk extension region can comprise one or zero dimerization sites. As another example, if a stalk region comprises one dimerization site, a stalk extension region can comprise zero dimerization sites. In some examples, a stalk extension region lacks a dimerization site. In some cases, one or more dimerization site(s) can be membrane proximal. In other cases, one or more dimerization site(s) can be membrane distal.


In certain embodiments, the dimerization site is a cysteine residue capable of forming a disulfide bond. In certain embodiments, the stalk extension region is capable of forming fewer disulfide bond(s) as compared to a stalk region. For example, if a stalk region is capable of forming two disulfide bonds, a stalk extension region may be capable of forming one or no disulfide bonds. As another example, if a stalk region is capable of forming one disulfide bond, a stalk extension region may be capable of forming no such bonds.


Each of the stalk extension regions can be about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, or greater amino acids in length.


In certain embodiments, the stalk extension region is homologous to the CD8α hinge region. In certain such embodiments, the stalk extension region comprises the amino acid sequence of SEQ ID NO: 469 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 469 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 469, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 469.


In certain embodiments, the stalk extension region is encoded by any one of SEQ ID NOs: 470-472, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 470-472, hybridizes under stringent hybridization conditions with the complement of any one of SEQ ID NOs: 470-472, or is a codon degenerate variant of any one of SEQ ID NOs: 470-472.


In certain embodiments, the spacer comprises a stalk region and 1 to 3 stalk extension regions. In certain such embodiments, the spacer comprises a stalk region and 2 stalk extension regions, for example a CD8α hinge region and 2 stalk extension regions wherein each stalk extension region is homologous to the CD8α hinge region.


In some embodiments, each of the stalk region and stalk extension region(s) can be derived from at least one of a CD8α hinge domain, a CD28 hinge domain, a CTLA-4 hinge domain, a LNGFR extracellular domain, IgG1 hinge, IgG4 hinge and CH2-CH3 domain. The stalk and stalk extension region(s) can be separately derived from any combination of CD8α hinge domain, CD28 hinge domain, CTLA-4 hinge domain, LNGFR extracellular domain, IgG1 hinge, IgG4 hinge or CH2-CH3 domain. As an example, the stalk region can be derived from CD8α hinge domain and at least one stalk extension region can be derived from CD28 hinge domain thus creating a hybrid spacer. As another example, the stalk region can be derived from an IgG1 hinge or IgG4 hinge and at least one stalk extension region can be derived from a CH2-CH3 domain of IgG.


In certain such embodiments, the spacer comprises the amino acid sequence of SEQ ID NO: 473 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 473 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 473, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 473.


In certain embodiments, the spacer is encoded by SEQ ID NO: 474, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 474, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 474, or is a codon degenerate variant of SEQ ID NO: 474.


C. Transmembrane Domain


The transmembrane domain can be derived from either a natural or a synthetic source. Where the source is natural, the domain can, for example, be derived from any membrane-bound or transmembrane protein. Suitable transmembrane domains include transmembrane domains from a TCR-alpha chain, a TCR-beta chain, a TCR-γ1 chain, a TCR-δ chain, a TCR-zeta chain, CD28, CD3 epsilon, CD3ζ CD45, CD4, CD5, CD8α, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, ICOS, GITR, CD152 (CTLA-4), or CD154, or a functional fragment or variant thereof. Alternatively, the transmembrane domain can be synthetic, and can comprise hydrophobic residues such as leucine and valine. In some embodiments, a triplet of phenylalanine, tryptophan and valine is found at one or both termini of a synthetic transmembrane domain. In some embodiments, the transmembrane domain comprises a CD8α transmembrane domain, a CD152 (CTLA-4), TCRγ1, TCRδ or a CD3ζ transmembrane domain.


Optionally, a short oligonucleotide or polypeptide linker, in some embodiments between 2 and 10 amino acids in length, may link the transmembrane domain with the intracellular signaling domain of a CAR. In some embodiments, the linker is a glycine-serine linker.


In some embodiments, the transmembrane domain comprises a CD8α transmembrane domain or a CD3ζ transmembrane domain, or a functional fragments or variants thereof.


In certain embodiments, the transmembrane domain comprises a CD8α transmembrane domain, or a functional fragment or variant thereof. In certain such embodiments, the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 475 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 475 by at most 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 475, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 475.


In certain embodiments, the CD8α transmembrane domain, or functional fragment or variant thereof, is encoded by SEQ ID NO: 476 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 476, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 476, or is a codon degenerate variant of SEQ ID NO: 476.


In certain embodiments, the transmembrane domain comprises a CD28 transmembrane domain, or a functional fragment or variant thereof. In certain such embodiments, the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 477 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 477 by at most 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 477, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 477.


In certain embodiments, the CD28 transmembrane domain, or functional fragment or variant thereof, is encoded by SEQ ID NO: 478 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 478, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 478, or is a codon degenerate variant of SEQ ID NO: 438.


D. Intracellular Signaling Domain


The intracellular signaling domain of the CAR may be responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR has been placed. The term “effector function” refers to a specialized function of a cell. Effector function of a T cell, for example, can be cytolytic activity or helper activity including the secretion of cytokines. While usually the entire intracellular signaling domain can be employed, in many cases it is not necessary to use the entire chain. To the extent that a truncated portion of the intracellular signaling domain is used, such truncated portion can be used in place of the intact chain as long as it transduces the effector function signal. In some embodiments, the intracellular domain further comprises a signaling domain for T-cell activation.


In some embodiments, the intracellular cell signaling domain interacts with a T cell, a Natural Killer (NK) cell, a cytotoxic T lymphocyte (CTL), or a regulatory T cell.


The intracellular domain can comprise an amino acid sequence derived from FCER1G, CD19, CD40, KIR3DL1, KIR3DL2, KIR2DL3, KIR2DL4, KIR2DL5, KIR3DL1, KIR3DL2, KIR3DL3, SIRPA, FCRL1, FCRL2, FCRL3, FCRL4, FCRL5, FCRL6, FCGR1A, FCGR2A, FCGR2B, FCGR3A, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, PILRB, NCR1, NCR2, NCR3, NKG2A, NKG2C, NKG2D, DAP12, FCER1G, DAP10, CD84, CD19, KIR3DL1, KIR3DL2, KIR2DL2, KIR2DL3, KIR2DL4, KIR2DL5, KIR3DL2, KIR3DL3, SIRPA, FCRL1, FCRL2, FCRL3, FCRL4, FCRL5, FCRL6, CD4, CD8A, CD8B, LAT, FCGR1A, FCGR2A, FCGR2B, FCGR3A, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, NCR1, NCR2, NCR3, LY9, NKG2C, TCR zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CD3ζ, CD5, CD22, CD79a, CD79b or CD66d, or a functional fragment or variant thereof. In some cases, the signaling domain for T-cell activation comprises a domain derived from CD3ζ. or a functional fragment or variant thereof.


In certain embodiments, the intracellular signaling domain comprises a CD3ζ domain, or a functional fragment or variant thereof. In certain such embodiments, the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 479 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 479 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 479, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 479.


In certain embodiments, the CD3 domain, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 480 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 480, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 480, or is a codon degenerate variant of SEQ ID NO: 480.


The intracellular signaling domain can further comprise one or more co-stimulatory domains. Exemplary co-stimulatory domains include, but are not limited to, CD8, CD27, CD28, 4-1BB (CD137), ICOS, DAP10, DAP12, OX40 (CD134), and CD3-zeta co-stimulatory domains and functional fragments or variants thereof. In some instances, a CAR described herein comprises one or more, or two or more of co-stimulatory domains selected from CD8, CD27, CD28, 4-1BB (CD137), ICOS, DAP10, DAP12, and OX40 (CD134) co-stimulatory domains and functional fragments or variants thereof. In some instances, a CAR described herein comprises one or more, or two or more of co-stimulatory domains selected from CD27, CD28, 4-1BB (CD137), ICOS, and OX40 (CD134) co-stimulatory domains and functional fragments or variants thereof. In some instances, a CAR described herein comprises one or more, or two or more of co-stimulatory domains selected from CD8, CD28, 4-1BB (CD137), DAP10, and DAP12 co-stimulatory domains and functional fragments or variants thereof. In some instances, a CAR described herein comprises one or more, or two or more co-stimulatory domains selected from CD28 and 4-1BB (CD137) co-stimulatory domains and functional fragments or variants thereof. In some instances, a CAR described herein comprises CD28 and 4-1BB (CD137) co-stimulatory domains or their respective functional fragments or variants. In some instances, a CAR described herein comprises CD28 and OX40 (CD134) co-stimulatory domains or their respective functional fragments and variants. In some instances, a CAR described herein comprises CD8 and CD28 co-stimulatory domains or their respective functional fragments and variants. In some instances, a CAR described herein comprises a CD28 co-stimulatory domains or a functional fragment or variant thereof. In some instances, a CAR described herein comprises a 4-1BB (CD137) co-stimulatory domain or a functional fragment or variant thereof. In some instances, a CAR described herein comprises an OX40 (CD134) co-stimulatory domain or a functional fragment or variant thereof. In some instances, a CAR described herein comprises a CD8 co-stimulatory domain or a functional fragment or variant thereof. In some instances, the CAR described herein comprises a DAP10 co-stimulatory domain or a functional fragment or variant thereof. In some instances, the CAR described herein comprises a DAP12 co-stimulatory domain or a functional fragment or variant thereof.


In certain embodiments, the intracellular signaling domain comprises a CD28 co-stimulatory domain, or a functional fragment or variant thereof. In certain such embodiments, the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 481 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 481 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 481, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 481.


In certain embodiments, the CD28 co-stimulatory domain, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 482 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 482, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 482, or is a codon degenerate variant of SEQ ID NO: 442.


In certain embodiments, the intracellular signaling domain comprises a 4-1BB co-stimulatory domain, or a functional fragment or variant thereof. In certain such embodiments, the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 483 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 483 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 483, and/or is a conservatively-substituted variant of SEQ ID NO: 483.


In certain embodiments, the 4-1BB co-stimulatory domain, or functional fragment or variant thereof, is encoded by SEQ ID NO: 484 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 484, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 484, or is a codon degenerate variant of SEQ ID NO: 484.


In certain embodiments, the intracellular signaling domain comprises a DAP10 co-stimulatory domain, or a functional fragment or variant thereof. In certain embodiments, the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 485 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 485 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 485, and/or is a conservatively-substituted variant of SEQ ID NO: 485.


In certain embodiments, the DAP10 co-stimulatory domain, or functional fragment or variant thereof, is encoded by the sequence of SEQ ID NO: 486, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 486, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 486, or is a codon degenerate variant of SEQ ID NO: 486.


In certain embodiments, the intracellular signaling domain comprises a DAP12 co-stimulatory domain, or a functional fragment or variant thereof. In certain embodiments, the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 487 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 487 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 487, and/or is a conservatively-substituted variant of SEQ ID NO: 487.


In certain embodiments, the DAP12 co-stimulatory domain, or functional fragment or variant thereof, is encoded by the sequence of SEQ ID NO: 488, or functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 488, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 488, or is a codon degenerate variant of SEQ ID NO: 488.


In certain embodiments, the intracellular signaling domain comprises both a CD28 co-signaling domain and a 4-1BB co-signaling domain, or respective functional fragments or variants thereof, as described above.


E. Signal Peptide


In an embodiment, a signal peptide directs the nascent CAR protein into the endoplasmic reticulum. This is, for example, if the receptor is to be glycosylated and anchored in the cell membrane. Any eukaryotic signal peptide sequence is envisaged to be functional. Generally, the signal peptide natively attached to the protein or, in the case of a fusion protein, the component closest to the N-terminus is used (e.g., in a scFv with the VL component at closest to the N-terminus, the native signal of the light chain is used). In some embodiments, the signal peptide is native for GM-CSFRa (SEQ ID NO: 489) or IgK (SEQ ID NO: 491), IgE (SEQ ID NO: 493) or a functional fragment or variant thereof. Other signal peptides that can be used include those native to CD8α (SEQ ID NO: 495) and CD28. In some embodiments, the signal peptide is that native to Mouse Ig VH region 3 (SEQ ID NO: 497), β2M signal peptide (SEQ ID NO: 499), Azurocidin (SEQ ID NO: 501), Human Serum Albumin signal peptide (SEQ ID NO: 503), A2M receptor associated protein signal peptide (SEQ ID NO: 505), IGHV3-23 (SEQ ID NO: 507), IGKV1-D33 (HuL1) (SEQ ID NO: 509), IGKV1-D33 (L14F) (HuH7) (SEQ ID NO: 511), or a functional fragment or variant thereof.


In certain embodiments, the CAR is linked to a GM-CSFRa signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the GM-CSFRa signal peptide has the amino acid sequence of SEQ ID NO: 489 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 489 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 489, and/or is a conservatively-substituted variant of SEQ ID NO: 489.


In certain embodiments, the GM-CSFRa signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 490 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 490, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 490, or is a codon degenerate variant of SEQ ID NO: 490.


In certain embodiments, the CAR is linked to an IgK signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the IgK signal peptide has the amino acid sequence of SEQ ID NO: 491 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 491 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 491, and/or is a conservatively-substituted variant of SEQ ID NO: 491.


In certain embodiments, the Igκ signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 492 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 492, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 492, or is a codon degenerate variant of SEQ ID NO: 492.


In certain embodiments, the CAR is linked to an IgE signal peptide native to IgE (“IgE signal peptide”), or a functional fragment or variant thereof. In certain such embodiments, the IgE signal peptide the amino acid sequence of SEQ ID NO: 493 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 493 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 493, and/or is a conservatively-substituted variant of SEQ ID NO: 493.


In certain embodiments, the IgE signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 494 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 494, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 494, or is a codon degenerate variant of SEQ ID NO: 494.


In certain embodiments, the CAR is linked to an CD8α signal peptide native to CD8α, or a functional fragment or variant thereof. In certain such embodiments, the CD8α signal peptide comprises the amino acid sequence of SEQ ID NO: 495 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 495 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 495, and/or is a conservatively-substituted variant of SEQ ID NO: 495.


In certain embodiments, the CD8α signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 496 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 496, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 496 or is a codon degenerate variant of SEQ ID NO: 496.


In certain embodiments, the CAR is linked to a Mouse Ig VH region 3 signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the Mouse Ig VH region 3 signal peptide has the amino acid sequence of SEQ ID NO: 497 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 497 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 497, and/or is a conservatively-substituted variant of SEQ ID NO: 497.


In certain embodiments, the Mouse Ig VH region 3 signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 498 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 498, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 498, or is a codon degenerate variant of SEQ ID NO: 498.


In certain embodiments, the CAR is linked to a β2M signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the β2M signal peptide has the amino acid sequence of SEQ ID NO: 499 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 499 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 499, and/or is a conservatively-substituted variant of SEQ ID NO: 499.


In certain embodiments, the β2M signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 500 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 500, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 500, or is a codon degenerate variant of SEQ ID NO: 500.


In certain embodiments, the CAR is linked to an Azurocidin signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the Azurocidin signal peptide has the amino acid sequence of SEQ ID NO: 501 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 501 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 501, and/or is a conservatively-substituted variant of SEQ ID NO: 501.


In certain embodiments, the Azurocidin signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 502 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 502, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 502, or is a codon degenerate variant of SEQ ID NO: 502.


In certain embodiments, the CAR is linked to a human serum albumin signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the human serum albumin signal peptide has the amino acid sequence of SEQ ID NO: 503 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 503 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 503, and/or is a conservatively-substituted variant of SEQ ID NO: 503.


In certain embodiments, the human serum albumin signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 504 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 504, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 504, or is a codon degenerate variant of SEQ ID NO: 504.


In certain embodiments, the CAR is linked to an A2M receptor associated protein signal peptide, or a functional fragment or variant thereof. In certain such embodiments, A2M receptor associated protein signal peptide has the amino acid sequence of SEQ ID NO: 505 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 505 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 505, and/or is a conservatively-substituted variant of SEQ ID NO: 505.


In certain embodiments, the A2M receptor associated protein signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 506 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 506, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 506, or is a codon degenerate variant of SEQ ID NO: 506.


In certain embodiments, the CAR is linked to an IGHV3-23 signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the IGHV3-23 signal peptide has the amino acid sequence of SEQ ID NO: 507 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 507 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 507, and/or is a conservatively-substituted variant of SEQ ID NO: 507.


In certain embodiments, the IGHV3-23 signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 508 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 508, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 508, or is a codon degenerate variant of SEQ ID NO: 508.


In certain embodiments, the CAR is linked to an IGKV1-D33 signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the IGKV1-D33 signal peptide has the amino acid sequence of SEQ ID NO: 509 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 509 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 509, and/or is a conservatively-substituted variant of SEQ ID NO: 509.


In certain embodiments, the IGKV1-D33 signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 510 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 510, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 510, or is a codon degenerate variant of SEQ ID NO: 510.


In certain embodiments, the CAR is linked to an IGHV3-33 (L14F) signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the IGHV3-33 (L14F) signal peptide has the amino acid sequence of SEQ ID NO: 511 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 511 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 511, and/or is a conservatively-substituted variant of SEQ ID NO: 511.


In certain embodiments, the IGHV3-33 (L14F) signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 512 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 512, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 512, or is a codon degenerate variant of SEQ ID NO: 512.


In certain embodiments, the CAR is linked to an TVB2 (T21A) signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the TVB2 (T21A) signal peptide has the amino acid sequence of SEQ ID NO: 513 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 513 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 513, and/or is a conservatively-substituted variant of SEQ ID NO: 513.


In certain embodiments, the TVB2 (T21A) signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 514 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 514, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 514, or is a codon degenerate variant of SEQ ID NO: 514.


In certain embodiments, the CAR is linked to an CD52 signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the CD52 signal peptide has the amino acid sequence of SEQ ID NO: 515 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 515 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 515, and/or is a conservatively-substituted variant of SEQ ID NO: 515.


In certain embodiments, the CD52 signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 516 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 516, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 516, or is a codon degenerate variant of SEQ ID NO: 516.


In certain embodiments, the CAR is linked to an LNGFR signal peptide, or a functional fragment or variant thereof. In certain such embodiments, the LNGFR signal peptide has the amino acid sequence of SEQ ID NO: 517 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 517 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 517, and/or is a conservatively-substituted variant of SEQ ID NO: 517.


In certain embodiments, the LNGFR signal peptide, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 518 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 518, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 518, or is a codon degenerate variant of SEQ ID NO: 518.


F. Exemplary CAR Constructs


By way of example, but not limitation, the CAR can comprise an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with any one of SEQ ID NOs: 591, 593, 595, 597, 599, 601, 603, 605, 607, 609, 611, 613, 615, 617, 619, 621, 623, 625, 627, and 629 or a conservatively-substituted variant thereof. By way of further example, but not limitation, a polynucleotide encoding the CAR can comprise a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the sequence of any one of SEQ ID NOs: 592, 594, 596, 598, 600, 602, 604, 606, 608, 610, 612, 614, 616, 618, 620, 622, 624, 626, 628, and 630; a sequence that hybridizes under stringent hybridization conditions with the complement of any one of such sequences; or a codon degenerate variant of any one of such sequences.


In certain embodiments, the CAR can comprise an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 623 or a conservatively-substituted variant thereof. In certain such embodiments, the polynucleotide encoding the CAR can comprise a nucleic acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the sequence of SEQ ID NO: 624; a sequence that hybridizes under stringent hybridization conditions with the complement of any one of such sequences; or a codon degenerate variant of any one of such sequences.


CARs and CAR construction as well as compositions are also described, for example, in:

  • Chimeric Antigen Receptor (CAR) T-Cell Therapies for Cancer: A Practical Guide, Edited by: Daniel W. Lee and Nirali N. Shah, 2020 (ISBN 978-0-323-66181-2; DOI https://doi.org/10.1016/C2017-0-04066-1);
  • Second Generation Cell and Gene-based Therapies, Biological Advances, Clinical Outcomes and Strategies for Capitalisation, Editors-in-Chief: Alain A. Vertés, Devyn M. Smith, Nathan J. Dowden, 2020 (ISBN 978-0-12-812034-7; DOI https://doi.org/10.1016/C2016-0-02070-3);
  • Basics of Chimeric Antigen Receptor (CAR) Immunotherapy, Author: Mumtaz Yaseen Balkhi, 2020 (ISBN 978-0-12-819573-4, DOI https://doi.org/10.1016/C2018-0-05356-6);
  • Engineering and Design of Chimeric Antigen Receptors, Authors: Sonia Guedan, Hugo Calderon, Avery D. Posey, Jr., and Marcela V. Maus, Molecular Therapy: Methods & Clinical Development, Vol. 12, March (2019) (https://www.cell.com/molecular-therapy-family/methods/pdf/S2329-0501(18)30133-5.pdf);
  • Chimeric Antigen Receptor T Cell Therapy Pipeline at a Glance: A Retrospective and Systematic Analysis from Clinicaltrials.Gov, Authors: Eider F Moreno Cortes, Caleb K Stein, Paula A Lengerke Diaz, Cesar A Ramirez-Segura, Januario E. Castro, MD, Blood (2019) 134 (Supplement_1): 5629 (https://doi.org/10.1182/blood-2019-132273);
  • WO2020209934 (PCT/US2020/017794)—Novel chimeric antigen receptors and libraries (MIT);
  • WO2020037142 (PCT/US2019/046691)—Compositions and methods for high-throughput activation screening to boost t cell effector function (Yale);
  • WO2015123642 (PCT/US2015/016057)—Chimeric antigen receptors and methods of making (Univ. TX);
  • WO2020014366 (PCT/US2019/041213)—Ror-1 specific chimeric antigen receptors and uses thereof (Precigen);
  • WO2019236577 (PCT/US2019/035384)—Muc16 specific chimeric antigen receptors and uses thereof (Precigen)
  • WO2019079486 (PCT/US2018/056334)—Polypeptide compositions comprising spacers (Precigen)
  • WO2017214333 (PCT/US2017/036440)—Cd33 specific chimeric antigen receptors (Precigen)


V. Cytokine

In some embodiments, the modified immune effector cell of the present invention can comprise a cytokine. The cytokine may, for example, be encoded by the polynucleotide of the present disclosure. For example, the polynucleotide may encode the miRNA(s), CAR and cytokine, or the miRNA(s) and cytokine.


In some cases, the cytokine comprises at least one chemokine, interferon, interleukin, lymphokine, tumor necrosis factor, or variant or combination thereof. In certain embodiments, the cytokine is an interferon, GM-CSF, G-CSF, M-CSF, LT-beta, TNF-alpha, growth factors, hGH, and/or a ligand of human Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, IFN-alpha, IFN-beta, or IFN-gamma.


In certain embodiments, the cytokine is an interleukin. In some cases the interleukin is IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-35, or a functional variant or fragment thereof.


In certain embodiments, the cytokine may be IL-12, or a functional fragment or variant thereof. In some embodiments, the IL-12 is a single chain IL-12 (sclL-12), protease sensitive IL-12, destabilized IL-12, membrane bound IL-12, intercalated IL-12. In some instances, the IL-12 variants are as described in WO2015/095249, WO2016/048903, WO2017/062953.


In certain embodiments, the cytokine may be IL-15, or a functional fragment or variant thereof. In certain embodiments, the IL-15, or functional fragment or variant thereof, is membrane-bound. Such may occur when IL-15, or a functional fragment or variant thereof, is bound to membrane-bound IL-15Rα, or a functional fragment or variant thereof. Thus, certain embodiments of the present invention may involve a fusion protein comprising IL-15 and IL-15Rα, or their respective functional fragments or variants.


In certain embodiments, the IL-15, or functional fragment or variant thereof, comprises the amino acid sequence of SEQ ID NO: 519, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 519 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 519, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 519.


In certain embodiments, the IL-15, or functional fragment or variant thereof, is encoded by a nucleic acid comprising the sequence of SEQ ID NO: 520, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 520, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 520, or is a codon degenerate variant of SEQ ID NO: 520.


In certain embodiments, the IL-15Rα, or functional fragment or variant thereof, comprises the amino acid sequence of SEQ ID NO: 521 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 521 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 521, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 521.


In certain embodiments, the IL-15Rα, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 522, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 522, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 522, or is a codon degenerate variant of SEQ ID NO: 522.


In certain embodiments, the IL-15, or functional fragment or variant thereof, is linked to the IL-15Rα, or functional fragment thereof by way of a linker.


In certain embodiments, the linker comprises the amino acid sequence of SEQ ID NO: 529 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than SEQ ID NO: 529 by at most 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 529, and/or is a conservatively-substituted variant of SEQ ID NO: 529.


In certain embodiments, the linker is encoded by a nucleic acid comprising SEQ ID NO: 530 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 530, hybridizes under stringent hybridization conditions with SEQ ID NO: 530, or is a codon degenerate variant of SEQ ID NO: 530.


In certain embodiments, the fusion protein comprising IL-15 and IL-15Rα comprises the amino acid sequence of SEQ ID NO: 523 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 523 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 523, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 523.


In certain embodiments, the fusion protein comprising IL-15 and IL-15Rα is encoded by a nucleic acid comprising SEQ ID NO: 524 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 524, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 524, or is a codon degenerate variant of SEQ ID NO: 603.


In certain embodiments, the cytokine is linked to a signal peptide. Any signal for use in eukaryotic cells, including those described above for use with the CARs may be linked to the cytokine. In certain embodiments, the cytokine is linked to an IgE signal peptide.


In certain embodiments, the fusion protein comprising IL-15 and IL-15Rα comprises the amino acid sequence of SEQ ID NO: 525 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 525 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 525, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 525.


In certain embodiments, the fusion protein comprising IL-15 and IL-15Rα is encoded by a nucleic acid comprising SEQ ID NO: 526 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 526, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 526, or is a codon degenerate variant of SEQ ID NO: 526.


VI. Cell Tag

In some embodiments, the modified immune effector cell of the present invention can comprise a cell tag. The cell tag may, for example, be encoded by the polynucleotide of the present disclosure. For example, the polynucleotide may encode the miRNA(s), CAR and/or cytokine described herein as well as a cell tag. In some aspects, the cell tag is used as a kill switch, selection marker, a biomarker, or a combination thereof.


In certain embodiments, the cell tag is capable of being bound by a predetermined binding partner. In certain such embodiments, the cell tag is non-immunogenic. In certain such embodiments, the cell tag comprises a polypeptide that is truncated so that it is non-immunogenic.


In certain embodiments, the administration of the predetermined binding partner allows for depletion of infused CAR-T cells. For example, the administration of cetuximab or any antibody that recognizes HER1 allows for the elimination of cells expressing a cell tag comprising truncated non-immunogenic HER1. The truncation of the HER1 sequence eliminates the potential for EGF ligand binding, homo- and hetero-dimerization of EGFR, and/or EGFR-mediated signaling while keeping cetuximab-binding ability intact (Ferguson, K., 2008. A structure-based view of Epidermal Growth Factor Receptor regulation. Annu Rev Biophys, Volume 37, pp. 353-373).


In certain embodiments, the cell tag comprises at least one of a truncated non-immunogenic HER1 polypeptide, a truncated non-immunogenic LNGFR polypeptide, a truncated non-immunogenic CD20 polypeptide, or a truncated non-immunogenic CD52 polypeptide, or a functional fragment or variant thereof.


In certain embodiments, the cell tag comprises a truncated non-immunogenic HER1 polypeptide comprising a HER1 Domain III and a truncated HER1 Domain IV. Such domains and the nucleic acid sequences encoding the same include those described in WO 2018/226897.


In certain embodiments, the HER1 Domain III comprises the amino acid sequence of SEQ ID NO: 604 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 565 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 565, and/or is a conservatively-substituted variant the amino acid sequence of sequence of SEQ ID NO: 565.


In certain embodiments, the HER1 Domain III, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 566 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 566, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 566, or is a codon degenerate variant of SEQ ID NO: 566.


In certain embodiments, the truncated HER1 Domain IV comprises the amino acid sequence of SEQ ID NO: 567 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 567 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 567, and/or is a conservatively-substituted variant the amino acid sequence of sequence of SEQ ID NO: 567.


In certain embodiments, the truncated HER1 Domain IV, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 568 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 568, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 568, or is a codon degenerate variant of SEQ ID NO: 568.


In certain such embodiments, the truncated non-immunogenic HER1 comprises the amino acid sequence of SEQ ID NO: 569 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 569 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 569, and/or is a conservatively-substituted variant the amino acid sequence of sequence of SEQ ID NO: 569.


In certain embodiments, the truncated non-immunogenic HER1, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 570 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 570, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 570, or is a codon degenerate variant of SEQ ID NO: 570.


In certain embodiments, the cell tag comprises a truncated non-immunogenic CD20, or CD20t-1, or a functional fragment or variant thereof. In certain such embodiments, the cell tag comprises the amino acid sequence of SEQ ID NO: 573, SEQ ID NO: 575, or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequences of SEQ ID NO: 573 or SEQ ID NO: 575 by at most 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 573 or SEQ ID NO: 575, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 573 or SEQ ID NO: 575.


In certain embodiments, the truncated non-immunogenic CD20, or CD20t-1, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 574 or SEQ ID NO: 576 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 574 or SEQ ID NO: 576, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 574 or SEQ ID NO: 576, or is a codon degenerate variant of SEQ ID NO: 574 or SEQ ID NO: 576.


In certain embodiments, the cell tag further comprises a transmembrane domain. The transmembrane domain can be derived from either a natural or a synthetic source. Where the source is natural, the domain can, for example, be derived from any membrane-bound or transmembrane protein. Suitable transmembrane domains can include the transmembrane domain(s) of alpha, beta or zeta chain of the T-cell receptor; or a transmembrane domain from CD28, CD3 epsilon, CD3, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 or CD154, or a functional fragment or variant thereof. Alternatively, the transmembrane domain can be synthetic, and can comprise hydrophobic residues such as leucine and valine. In some embodiments, a triplet of phenylalanine, tryptophan and valine is found at one or both termini of a synthetic transmembrane domain.


In some embodiments, the transmembrane domain comprises a CD28 transmembrane domain, or a functional fragment or variant thereof. In certain such embodiments, the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 477 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 477 by at most 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 477, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 477.


In certain embodiments, the CD28 transmembrane domain, or functional fragment or variant thereof, is encoded by a nucleic acid comprising SEQ ID NO: 478, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 478, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 478, or is a codon degenerate variant of SEQ ID NO: 478.


In certain embodiments, the cell tag comprises a truncated HER1, or functional fragment or variant thereof, and a transmembrane domain, or a functional fragment or variant thereof. In some embodiments, the cell comprises the amino acid sequence of SEQ ID NO: 571 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 571 by at most 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 571, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 571.


In certain embodiments, the cell tag is encoded by a nucleic acid comprising SEQ ID NO: 572, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 572, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 572, or is a codon degenerate variant of SEQ ID NO: 572.


In certain embodiments, the cell tag is linked with a signal peptide. The signal peptide can be any signal peptide suitable for use in a eukaryotic cell including those described with respect to CARs herein. In certain embodiments, the signal peptide is a Igic signal peptide comprising the amino acid sequence of SEQ ID NO: 491, or a functional fragment or variant thereof


VII. Genetic Construct

As shown in FIG. 16, an exemplary polynucleotide is provided which can be used as template for the expression of various genes and other regulatory elements in cells. It should be understood that various elements can be included or omitted in the polynucleotide and that different options are shown for various exemplary sites in the polynucleotide.


As shown, the polynucleotide can include an integration signal for attP/attB phage integration of the polynucleotide into a bacterial genome. The polynucleotide can further include a 5′ homology arm or 5′ terminal repeat and a 3′ homology arm or 3′ terminal repeat. The polynucleotide can further include insulators, boundary elements and S/MAR positioned 3′ adjacent to the 5′ homology arm or 5′ terminal repeat and 5′ adjacent to the 3′ homology arm or 3′ terminal repeat. Between the insulators, boundary elements, or S/MAR, the polynucleotide can include, from 5′ to 3′, a promoter which can include a silencer, enhancer, TF binding modules and a core promoter; a 5′ untranslated region which can include stability modules, translation control elements, and intron-embedded elements such as miRNA encoding sequences; one or more genes which can include signal peptides, extracellular domains, transmembrane domains, signaling domains, antibody domains, peptide linkers, inteins and epitope tags; and a 3′ untranslated region that can include stability modules, translation control, 3′ end processing signals and a transcription terminator. It should be understood that the genes to be expressed can be separated by IRES, cleavage peptides, or ribosomal skipping peptides.


The CAR may be encoded in the same genetic construct with the miRNA, the cytokine, and/or the cell tag. An advantage of having two or more of such components expressed using one genetic construct is stoichiometric expression of such components.


A. Linkers


In certain embodiments, the polypeptides of the present invention (e.g., the CAR, the cytokine, and the cell tag) are linked by linker polypeptide(s). The linkers may also be used to link domains of a polypeptide (e.g., the VH and VL domains of a CAR, the truncated HER1 and transmembrane domains of the cell tag, and the IL-15 and IL-15Rα domains).


Linkers suitable in the present invention include flexible linkers, rigid linkers, and in vivo cleavable linkers. In some cases, the linker acts to link functional domains together (as in flexible and rigid linkers) or to release a free functional domain in vivo as in in vivo cleavable linkers.


As noted, in some cases, the linker sequence may include a flexible linker. Flexible linkers can be applied when a joined domain requires a certain degree of movement or interaction. Flexible linkers can be composed of small, non-polar (e.g., Gly) or polar (e.g., Ser or Thr) amino acids. A flexible linker can have sequences consisting primarily of stretches of Gly and Ser residues (“GS” linker). An example of a flexible linker can have the sequence of (Gly-Gly-Gly-Gly-Ser)n. By adjusting the copy number “n”, the length of this exemplary GS linker can be optimized to achieve appropriate separation of functional domains, or to maintain necessary inter-domain interactions. For example, (Gly-Gly-Gly-Gly-Ser)n, wherein n is 4 is a (G4S)4 linker as shown in SEQ ID NO: 608 or a conservatively substituted amino acid sequence thereof. Besides GS linkers, other flexible linkers can be utilized for recombinant fusion proteins. In some cases, flexible linkers can contain additional amino acids such as Thr and Ala to maintain flexibility. In other cases, polar amino acids such as Lys and Glu can be used to improve solubility.


Flexible linkers can be suitable choices when certain movements or interactions are desired for fusion protein domains. In addition, although flexible linkers do not have rigid structures, in some cases they can serve as a passive linker to keep a distance between functional domains. The length of a flexible linker may be adjusted to allow for proper folding or to achieve optimal biological activity of the fusion proteins.


A rigid linker can be utilized to maintain a fixed distance between domains of a polypeptide. Examples of rigid linkers include Alpha helix-forming linkers, Pro-rich sequence, (XP)n, X-Pro backbone, A(EAAAK)nA (n=2−5) (SEQ ID NO: 563) and functional fragments and variants thereof, to name a few. Rigid linkers can exhibit relatively stiff structures by adopting α-helical structures or by containing multiple Pro residues in some cases.


A linker useful in the present invention can be cleavable in some cases. In other cases, the linker is not cleavable. Linkers that are not cleavable can covalently join functional domains together to act as one molecule throughout an in vivo processes or an ex vivo process. A linker can also be cleavable in vivo. A cleavable linker can be introduced to release free functional domains in vivo.


A cleavable linker can be cleaved by the presence of reducing reagents, proteases, to name a few. For example, a reduction of a disulfide bond can be utilized to produce a cleavable linker. In the case of a disulfide linker, a cleavage event through disulfide exchange with a thiol, such as glutathione, could produce a cleavage. In some cases, a cleavable linker can allow for targeted cleavage. For example, an in vivo cleavage of a linker in a recombinant fusion protein can also be carried out by proteases that can be expressed in vivo under pathological conditions (e.g., cancer or inflammation), in specific cells or tissues, or constrained within certain cellular compartments. A cleavable linker can comprise a hydrazone, peptides, a disulfide, or a thioesther. For example, a hydrazone can confer serum stability. In other cases, a hydrazone can allow for cleavage in an acidic compartment. An acidic compartment can have a pH up to 7. A linker can also include a thioether. A thioether can be nonreducible A thioether can be designed for intracellular proteolytic degradation. Examples of cleavable linkers include Furinlink, fmdv, and 2A linkers (e.g., P2A, GSG-P2A, FP2A, T2A, and Furin-T2A), or functional fragments or variants thereof.


A fmdv linker may comprise the amino acid sequence of SEQ ID NO: 539 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 539 by at most 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 539, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 539.


In certain embodiments, the fmdv linker is encoded by a nucleic acid comprising SEQ ID NO: 540, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 540, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 540, or is a codon degenerate variant of SEQ ID NO: 540.


A P2A linker may comprise the amino acid sequence of SEQ ID NO: 547 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 547 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 547, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 547.


In certain embodiments, the P2A linker is encoded by a nucleic acid comprising SEQ ID NO: 548, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 548, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 548, or is a codon degenerate variant of SEQ ID NO: 548.


A GSG-P2A linker may comprise the amino acid sequence of SEQ ID NO: 549 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 549 by at most 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 549, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 549.


In certain embodiments, the GSG-P2A linker is encoded by a nucleic acid comprising SEQ ID NO: 550, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 550, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 550, or is a codon degenerate variant of SEQ ID NO: 550.


A FP2A linker may comprise the amino acid sequence of SEQ ID NO: 555 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 555 by at most 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 555, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 555.


In certain embodiments, the FP2A linker is encoded by a nucleic acid comprising SEQ ID NO: 556, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 556, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 556, or is a codon degenerate variant of SEQ ID NO: 556.


A T2A linker may comprise the amino acid sequence of SEQ ID NO: 541 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 541 by at most 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 541, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 541.


In certain embodiments, the T2A linker is encoded by a nucleic acid comprising SEQ ID NO: 475428, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 542, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 54, or is a codon degenerate variant of SEQ ID NO: 542.


In some embodiments, the linker comprises a furin polypeptide and a 2A polypeptide, wherein the furin polypeptide and the 2A polypeptide are connected by a polypeptide linker comprising at least three hydrophobic amino acids. Such linkers are called “Furin-T2A” linkers. In some cases, the at least three hydrophobic amino acids are selected from the list consisting of glycine (Gly)(G), alanine (Ala)(A), valine (Val)(V), leucine (Leu)(L), isoleucine (Ile)(I), proline (Pro)(P), phenylalanine (Phe)(F), methionine (Met)(M), tryptophan (Trp)(W). In some cases, the polypeptide linker can include one or more GS linker sequences, for instance (GS)n, (SG)n, and (GSG)n, wherein n can be any number from zero to thirty.


A Furin-T2A linker may comprise the amino acid sequence of SEQ ID NO: 543 or 545 or a functional fragment or variant thereof. In certain embodiments, the functional fragment is shorter than the amino acid sequence of SEQ ID NO: 543 or 545 by at most 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues at the N- and/or C-terminus. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the amino acid sequence of SEQ ID NO: 543 or 545, and/or is a conservatively-substituted variant of the amino acid sequence of SEQ ID NO: 543 or 545.


In certain embodiments, the Furin-T2A linker is encoded by a nucleic acid comprising SEQ ID NO: 544 or 546, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 544 or 546, hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 544 or 546, or is a codon degenerate variant of SEQ ID NO: 544 or 546.


A linker can be an engineered linker. For example, a linker can be designed to comprise chemical characteristics such as hydrophobicity. Methods of designing linkers can be computational. In some cases, computational methods can include graphic techniques. Computation methods can be used to search for suitable peptides from libraries of three-dimensional peptide structures derived from databases. For example, a Brookhaven Protein Data Bank (PDB) can be used to span the distance in space between selected amino acids of a linker.


Further exemplary linkers are provided in the Sequence Listing.


In some cases, at least two linker sequences can be included in the same protein. For example, polypeptides of interest within a fusion protein can be separated by at least two linkers. In some cases, polypeptides can be separated by 2, 3, 4, 5, 6, 7, 8, 9, or up to 10 linkers.


The CAR, cell tag, and/or cytokine of the present invention may be expressed as a fusion protein. In such embodiments, such components may be linked together using a self-cleaving peptide, for example a 2A peptide.


In certain embodiments, the self-cleaving peptide is a T2A peptide, or a functional fragment or variant thereof.


In certain embodiments, the self-cleaving peptide is a Furin-T2A peptide, or a functional fragment or variant thereof.


In certain such embodiments, the CAR, the cytokine, and the cell tag are expressed as a fusion protein with the CAR and the cytokine linked by a self-cleaving linker, for example one comprising Furin-T2A, and the cytokine and cell tag linked by a self-cleaving linker, for example one comprising T2A.


B. Promoters


The polynucleotide of the invention can be present in the construct in operable linkage with a promoter. Appropriate promoters can be selected based on the host cell and effect sought. Suitable promoters include constitutive and inducible promoters. The promoters can be tissue specific, such promoters being well known in the art.


Examples of constitutive promoters for use in the present invention include immediate early cytomegalovirus (CMV) promoter; human elongation growth factor 1 alpha 1 (hEF1A1); simian virus 40 (SV40) early promoter; mouse mammary tumor virus (MMTV); human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter; MoMuLV promoter; avian leukemia virus promoter; Epstein-Barr virus immediate early promoter; Rous sarcoma virus promoter; and human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter; and functional fragments and variants thereof.


In certain embodiments, the promoter is a hEF1A1 promoter. A hEF1A1 promoter may comprise the sequence of SEQ ID NO: 577, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 577, or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 577. In certain embodiments, the promoter is a CMV promoter. A CMV promoter may comprise the sequence of SEQ ID NO: 578, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 578, or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 578.


In contrast to constitutive promoters, the use of an inducible promoter provides a molecular switch capable of turning on the expression of the polynucleotide sequence which it is operatively linked when such expression is desired, or turning off the expression when expression is not desired. Examples of inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter. In one aspect, the inducible promoter can be a gene switch ligand inducible promoter. In some cases, an inducible promoter can be a small molecule ligand-inducible two polypeptide ecdysone receptor-based gene switch, such as a RHEOSWITCH® gene switch.


VIII. Vectors and Delivery Systems

In certain embodiments, the polynucleotide of the present invention can be delivered to a target cell by any suitable delivery system, including non-viral and viral delivery systems. In some embodiments, a vector can include a polynucleotide of the present disclosure encoding the miRNA, CAR, cytokine, cell tag, or any combination thereof.


In certain cases, the miRNA(s), CAR, cytokine, and/or cell tag are expressed in separate vectors. In other aspects, the miRNA(s), CAR, cytokine, and/or cell tag are expressed from one single vector. In certain cases, the CAR and the miRNA(s) are expressed in separate vectors. In other aspects, the miRNA(s), CAR and cytokine are expressed from one single vector. In specific cases, the vectors can be lentiviral vectors, retroviral vectors, Sleeping Beauty transposons or vectors containing sequences for serine recombinase mediated integration. In some aspects, the vector is a plasmid, a mini-circle DNA or a nanoplasmid.


In certain embodiments, where the vector is a plasmid, mini-circle DNA or a nanoplasmid, the plasmid, mini-circle DNA or nanoplasmid can further include a bacterial origin of replication. In certain embodiments, the bacterial origin of replication can be from a ColE1 plasmid. In certain embodiments, the bacterial origin of replication comprises the sequence of SEQ ID NO: 579, or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 579, or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 579.


In order to assess the expression of one or more miRNA(s) and a CAR described herein or portions thereof, the expression vector to be introduced into a cell can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors or non-viral vectors. In other aspects, the selectable marker can be carried on a separate piece of DNA and used in a co-transfection procedure. Both selectable markers and reporter genes can be flanked with appropriate regulatory sequences to enable expression in the host cells. Useful selectable markers include, for example, antibiotic-resistance genes, such as neomycin resistance gene (neo) and ampicillin resistance gene and the like. In some embodiments, a truncated epidermal growth factor receptor (HER1t or HER1t-1) tag can be used as a selectable marker gene.


Reporter genes can be used for identifying potentially transfected cells and for evaluating the functionality of regulatory sequences. In general, a reporter gene is a gene that is not present in or expressed by the recipient organism or tissue and that encodes a polypeptide whose expression is manifested by some easily detectable property, e.g., enzymatic activity. Expression of the reporter gene is assayed at a suitable time after the DNA has been introduced into the recipient cells. Suitable reporter genes include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyl transferase, secreted alkaline phosphatase, or the green fluorescent protein gene (e.g., Ui-Tei et al., FEBS Letters 479: 79-82 (2000)). Suitable expression systems are well known and can be prepared using known techniques or obtained commercially. In general, the construct with the minimal 5′ flanking region showing the highest level of expression of reporter gene is identified as the promoter. Such promoter regions can be linked to a reporter gene and used to evaluate agents for the ability to modulate promoter-driven transcription.


In some embodiments, a viral vector described herein can comprise a hEF1A1 promoter to drive expression of transgenes, a bovine growth hormone polyA sequence to enhance transcription, a woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), as well as LTR sequences derived from the pFUGW plasmid.


A. Methods for Introducing Nucleic Acids into Cells


Methods of introducing and expressing genes into a cell are well known. In the context of an expression vector, the vector can be readily introduced into a host cell, e.g., mammalian, bacterial, yeast, or insect cell by any method in the art. For example, the expression vector can be transferred into a host cell by physical, chemical, or biological means.


1. Physical Methods


Physical methods for introducing a polynucleotide into a host cell, for instance an immune effector cell, include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well-known in the art. See, for example, Sambrook et al. (Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York (2001)). In some embodiments, a method for the introduction of a polynucleotide into a host cell is calcium phosphate transfection or polyethylenimine (PEI) Transfection. In some embodiments, a method for introduction of a polynucleotide into a host cell is electroporation.


a. Electroporation (EP) Buffers


Various buffers can be used for electroporation. The buffers disclosed herein were found to have improved properties, including enhanced transfection capabilities, notwithstanding that these buffers comprise fewer components as compared to other known electroporation buffers.


Table 4 provides differing amounts of monobasic and dibasic phosphate used as buffering agents. Table 5 provides buffers 1-20 which contain buffering agents and glucose. Table Z provides buffers 21-37 which contain buffering agents and mannitol. Table 7 provides pH, conductivity, and osmolality for Buffers 1, 2 and 3 compared to a control buffer (Mirus Bio™ Ingenio™ electroporation solution, Catalog No. MIR-50117; Mirus Bio LLC, Madison, Wis., USA) (“Control 1”).









TABLE 4







Differing Amounts of Monobasic and Dibasic


Phosphate Used as a Buffering Agent









0.2M NaH2PO4
0.2M Na2HPO4



(mL)
(mL)
pH












92.0
8.0
5.8


90.0
10.0
5.9


87.7
12.3
6.0


85.5
15.0
6.1


81.5
19.5
6.2


77.5
22.5
6.3


73.5
26.5
6.4


68.5
31.5
6.5


62.5
37.5
6.6


56.5
43.5
6.7


51.0
49.0
6.8


45.0
55.0
6.9


39.0
61.0
7.0


33.0
67.0
7.1


28.0
72.0
7.2


23.0
77.0
7.3


19.0
81.0
7.4


16.0
84.0
7.5


13.0
87.0
7.6


10.5
89.5
7.7


8.5
91.5
7.8
















TABLE 5







Buffers 1 through 20 - Buffering Agents and Glucose
















Na2HPO4/





Sample
Glucose
HEPES
NaH2PO4
KCl
MgCl2
DMSO


No.
(mM)
(mM)
(mM)
(mM)
(mM)
(%)
















1
30
5
105
10
20
0


2
31
0
90
5
15
0


3
30
10
90
5
15
0


4
25
10
120
15
25
0


5
30
25
50
2
10.5
5


6
0
5
160
10
10.5
0


7
0
5
160
2
20
5


8
15
25
160
10
20
5


9
30
5
160
2
1
2.5


10
15
15
105
6
10.5
2.5


11
30
25
50
10
1
0


12
30
5
50
6
20
5


13
30
15
160
10
1
5


14
15
5
50
2
1
0


15
0
5
50
10
1
5


16
0
25
50
10
20
2.5


17
30
25
160
2
20
0


18
0
15
50
2
20
0


19
0
25
160
6
1
0


20
0
25
105
2
1
5
















TABLE 6







Buffers 21 through 37 - Buffering Agents and Mannitol
















Na2HPO4/





Sample
Mannitol
HEPES
NaH2PO4
KCl
MgCl2
DMSO


No.
(mM)
(mM)
(mM)
(mM)
(mM)
(%)
















21
5
25
160
6
1
0


22
150
25
50
2
10.5
5


23
5
15
50
2
20
0


24
150
25
50
10
1
0


25
5
25
105
2
1
5


26
77.5
5
50
2
1
0


27
150
5
160
2
1
2.5


28
150
15
160
10
1
5


29
5
5
50
10
1
5


30
150
25
160
2
20
0


31
150
5
105
10
20
0


32
77.5
15
105
6
10.5
2.5


33
77.5
25
160
10
20
5


34
150
5
50
6
20
5


35
5
25
50
10
20
2.5


36
5
5
160
10
10.5
0


37
5
5
160
2
20
5
















TABLE 7







Composition, pH, Conductivity, and Osmolality


of Buffers 1, 2, and 3 Compared to Control 1


















Na2HPO4/




osm


Sample
Glucose
HEPES
NaH2PO4
KCl
MgCl2

Conductivity
(mOsm/


No.
(mM)
(mM)
(mM)
(mM)
(mM)
pH
(ms/cm)
kgH20)


















1
30
5
105
10
20
7.0
14.3
340


2
31
0
90
5
15
7.1
11.6
280


3
30
10
90
5
15
7.1
12.8
292


Control 1
X
X



7.3
16.9
575









In some embodiments, the buffer comprises a solvent, such as water. In some embodiments, the water may be purified and/or sterilized. For example, the water may be subjected to deionization (e.g., capacitive deionization or electrodeionization), reverse osmosis, carbon filtering, microfiltration, ultrafiltration, and/or ultraviolet sterilization. In some embodiments, the water is deionized. In some embodiments, the water is of a quality designated as “water for injection”; also known as “sterile water for injection.” Water for injection is generally made by distillation or reverse osmosis. Water for injection is a sterile, nonpyrogenic, solute-free preparation of water, chemically designated “H2O,” and having a pH of between about 5.0 and about 7.0, preferably about 5.5.


In some embodiments, the solvent comprises between 0.1% and 99.9% by volume of the total buffer volume. For example, the solvent may comprise at least about 0.1%, 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 99.1% by volume of the total buffer volume.


In some embodiments, the buffer comprises a solute, for example a sugar or an organic compound derived from sugar, for example a sugar alcohol. In embodiments wherein the buffer comprises a sugar, the sugar may comprise a monosaccharide, a disaccharide, and/or a polysaccharide. In some embodiments, the sugar comprises a monosaccharide, for example glucose, fructose, and/or galactose. In some embodiments, the sugar comprises a disaccharide, for example sucrose, lactose, and maltose. In some embodiments, the sugar comprises a polysaccharide, for example cellulose or starch. In embodiments wherein the buffer comprises a sugar alcohol, the sugar alcohol may comprise mannitol, sorbitol, xylitol, lactitol, isomalt, maltitol, and/or hydrogenated starch hydrolysates (HSH).


In some embodiments, the sugar is present in an amount less than about 50 millimolar (mM). For example, the sugar may be present in an amount less than about 45 mM, 40 mM, 35 mM, 30 mM, 25 mM, 20 mM, 15 mM, 10 mM, or 5 mM. In some embodiments, the sugar is present in an amount that ranges between about 10 mM and about 50 mM, about 10 mM and about 40 mM, about 10 mM and about 20 mM, or about 25 mM and about 35 mM. In some embodiments, the sugar is present in an amount of about 25 mM, 26 mM, 27 mM, 28 mM, 29 mM, 30 mM, 31 mM, 32 mM, 33 mM, 34 mM, or 35 mM.


In some embodiments, the sugar is glucose. In these embodiments, the glucose may be present in an amount less than about 50 millimolar (mM). For example, the glucose may be present in an amount less than about 45 mM, 40 mM, 35 mM, 30 mM, 25 mM, 20 mM, 15 mM, 10 mM, or 5 mM. In some embodiments, the glucose is present in an amount that ranges between about 10 mM and about 50 mM, about 10 mM and about 40 mM, about 10 mM and about 20 mM, or about 25 mM and about 35 mM. In some embodiments, the glucose is present in an amount of about 25 mM, 26 mM, 27 mM, 28 mM, 29 mM, 30 mM, 31 mM, 32 mM, 33 mM, 34 mM, or 35 mM. In certain embodiments, the glucose is present in an amount of about 30 mM or 31 mM.


In some embodiments, the sugar is mannitol. In these embodiments, the mannitol may be present in an amount less than about 50 millimolar (mM). For example, the mannitol may be present in an amount less than about 45 mM, 40 mM, 35 mM, 30 mM, 25 mM, 20 mM, 15 mM, 10 mM, or 5 mM. In some embodiments, the mannitol is present in an amount that ranges between about 10 mM and about 50 mM, about 10 mM and about 40 mM, about 10 mM and about 20 mM, or about 25 mM and about 35 mM. In some embodiments, the mannitol is present in an amount of about 25 mM, 26 mM, 27 mM, 28 mM, 29 mM, 30 mM, 31 mM, 32 mM, 33 mM, 34 mM, or 35 mM.


In some embodiments, the EP buffer comprises one or more chloride salts, for example potassium chloride (KCl) and/or magnesium chloride (MgCl2). In some embodiments, the buffer further comprises one or more buffering agents, for example, Na2HPO4, NaH2PO4, or Na2HPO4/NaH2PO4. In some embodiments, the buffer further comprises one or more of HEPES and/or DMSO. In other embodiments, the buffer specifically excludes one or more buffering agents commonly found in commercial electroporation (EP) buffers. For example, in some embodiments, the buffer excludes one or both of DMSO and/or HEPES.


In some embodiments, the buffer comprises water (H2O), glucose, KCl, MgCl2, and Na2HPO4/NaH2PO4. In some embodiments, the buffer comprises water (H2O), glucose, KCl, MgCl2, Na2HPO4/NaH2PO4, and HEPES. In other embodiments, the buffer comprises water (H2O), glucose, KCl, MgCl2, Na2HPO4/NaH2PO4, HEPES, and DMSO.


In some embodiments, the buffer consists essentially of water (H2O), glucose, KCl, MgCl2, and Na2HPO4/NaH2PO4. In some embodiments, the buffer consists essentially of water (H2O), glucose, KCl, MgCl2, Na2HPO4/NaH2PO4, and HEPES. In other embodiments, the buffer consists essentially of water (H2O), glucose, KCl, MgCl2, Na2HPO4/NaH2PO4, HEPES, and DMSO.


In some embodiments, the buffer consists of water (H2O), glucose, KCl, MgCl2, and Na2HPO4/NaH2PO4. In some embodiments, the buffer consists of water (H2O), glucose, KCl, MgCl2, Na2HPO4/NaH2PO4, and HEPES. In other embodiments, the buffer consists of water (H2O), glucose, KCl, MgCl2, Na2HPO4/NaH2PO4, HEPES, and DMSO.


In some embodiments, the buffering agent has a pH ranging from about 6.0 to 8.0, 6.5 to 8.0, 7.0 to 8.0, 7.5 to 8.0, 6.0 to 7.5, 6.0 to 7.0, 6.0 to 6.5, 6.5 to 7.5, or 6.5 to 7.0. In some embodiments, the buffering agent has a pH of about 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0.


In some embodiments, the buffer comprising the one or more buffering agents has a pH ranging from about 6.0 to 8.0, 6.5 to 8.0, 7.0 to 8.0, 7.5 to 8.0, 6.0 to 7.5, 6.0 to 7.0, 6.0 to 6.5, 6.5 to 7.5, or 6.5 to 7.0. In some embodiments, the buffer has a pH of about 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0.


In some embodiments, the buffer comprises one or both of Na2HPO4 and/or NaH2PO4. In embodiments wherein the buffer comprises both buffering agents, the ratio of the two (i.e., Na2HPO4/NaH2PO4) may be about 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 9:1, 8:1, 7:1, 6:1 5:1, 4:1, 3:1, 2:1, or 2:3. In some embodiments, the ratio of Na2HPO4/NaH2PO4 has a pH of 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, or 7.9.


In some embodiments, a mixture of Na2HPO4 and NaH2PO4 (also referred to “Na2HPO4/NaH2PO4” or “sodium phosphate”) may be present in the buffer in an amount ranging from about 50 mM and 160 mM, 60 mM to 150 mM, 70 mM to 140 mM, 75 mM to 130 mM, 80 mM to 125 mM, 90 mM to 125 mM, 90 mM to 120 mM, 90 mM to 115 mM, or 90 mM to 105 mM. In some embodiments, the Na2HPO4/NaH2PO4 is present in an amount of at least 50 mM, 60 mM, 70 mM, 80 mM, 90 mM, or 100 mM. In some embodiments, the Na2HPO4/NaH2PO4 is present in an amount of 80 mM, 81 mM, 82 mM, 83 mM, 84 mM, 85 mM, 86 mM, 87 mM, 88 mM, 89 mM, 90 mM, 91 mM, 92 mM, 93 mM, 94 mM, 95 mM, 96 mM, 97 mM, 98 mM, 99 mM, 100 mM, 101 mM, 102 mM, 103 mM, 104 mM, 105 mM, 106 mM, 107 mM, 108 mM, 109 mM, 110 mM, 111 mM, 112 mM, 113 mM, 114 mM, 115 mM, 116 mM, 117 mM, 118 mM, 119 mM, or 120 mM. In certain embodiments, the Na2HPO4/NaH2PO4 is present in an amount of 90 mM or 105 mM.


In embodiments wherein the buffer comprises KCl, the KCl may be present in an amount less than about 30 mM. For example, the KCl may be present in an amount less than about 25 mM, 20 mM, 15 mM, 10 mM, or 5 mM. In some embodiments, the KCl is present in an amount that ranges between about 1 mM and about 30 mM, about 2 mM and about 25 mM, about 3 mM and about 20 mM, about 4 mM and about 15 mM, about 5 mM and about 10 mM, or about 5 mM to about 15 mM. In some embodiments, the KCl is present in an amount of about 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, or 15 mM. In certain embodiments, the KCl is present in an amount of about 5 mM or 10 mM. In some embodiments, the KCl has a pH of about 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, or 7.9.


In embodiments wherein the buffer comprises MgCl2, the MgCl2 may be present in an amount less than about 50 mM. For example, the MgCl2 may be present in an amount less than about 45 mM, 35 mM, 30 mM 25 mM, 20 mM, 15 mM, 10 mM, or 5 mM. In some embodiments, the MgCl2 is present in an amount that ranges between about 5 mM and about 50 mM, about 6 mM and about 45 mM, about 7 mM and about 40 mM, about 8 mM and about 35 mM, about 9 mM and about 30 mM, about 10 mM and about 25 mM, or about 15 mM and about 25 mM. In some embodiments, the MgCl2 is present in an amount of about 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, 15 mM, 16 mM, 17 mM, 18 mM, 19 mM, 20 mM, 21 mM, 22 mM, 23 mM, 24 mM, 25 mM, 26 mM, 27 mM, 28 mM, 29 mM, or 30 mM. In certain embodiments, the MgCl2 is present in an amount of about 15 mM or 20 mM. In some embodiments, the MgCl2 has a pH of about 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, or 7.9.


In embodiments wherein the buffer comprises HEPES, the HEPES may be present in an amount less than about 30 mM. For example, the HEPES may be present in an amount less than about 25 mM, 20 mM, 15 mM, 10 mM, 5 mM, 4 mM, 3 mM, 2 mM, 1 mM, 0.5 mM, or 0.1 mM. In some embodiments, the HEPES is present in an amount that ranges between about 1 mM and about 30 mM, about 2 mM and about 25 mM, about 3 mM and about 20 mM, about 4 mM and about 15 mM, about 5 mM and about 10 mM. In some embodiments, the HEPES is present in an amount of about 0.1 mM, 0.5 mM, 1 mM, 2 mM, 3 mM, 4 mM, 5 mM, 6 mM, 7 mM, 8 mM, 9 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, or 15 mM. In certain embodiments, the HEPES is present in an amount of 0 mM, 5 mM, or 10 mM. In some embodiments, the HEPES has a pH of about 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, or 7.9.


In embodiments wherein the buffer comprises DMSO, the DMSO may be present in an amount equal to or less than 5%, 4%, 3%, 2%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% by volume of the total buffer volume. In some embodiments, DMSO is present from about 0% to about 2.5% by volume of the total buffer volume. In some embodiments, DMSO is present in an amount ranging from about 0.1% to 5%, 1% to 5%, 2% to 5%, 3% to 5%, or 4% to 5% by volume of the total buffer volume. In some embodiments, the DMSO has a pH of about 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, or 7.9. In other embodiments, DMSO is not included in the buffer at all.


In certain embodiments, the buffer comprises a sugar in an amount equal to or less than 50 mM; HEPES in an amount equal to or less than 25 mM; Na2HPO4/NaH2PO4 in an amount equal to or less than 160 mM; KCl in an amount equal to or less than 10 mM; MgCl2 in an amount equal to or less than 20 mM; and DMSO in an amount equal to or less than 5% by volume of the total buffer volume. In some of these embodiments, the sugar may comprise a monosaccharide and/or a sugar alcohol. In some of these embodiments, the sugar is mannitol and/or glucose. In some of these embodiments, the sugar is glucose. In some embodiments, the buffer does not comprise DMSO.


In certain embodiments, the buffer comprises a sugar in an amount of at least about 15 mM; HEPES in an amount equal to or less than 25 mM; Na2HPO4/NaH2PO4 in an amount of at least about 90 mM; KCl in an amount of at least about 2 mM; MgCl2 in an amount of at least 15 mM; and DMSO in an amount equal to or less than 5% by volume of the total buffer volume. In some of these embodiments, the sugar may comprise a monosaccharide and/or a sugar alcohol. In some of these embodiments, the sugar is mannitol and/or glucose. In some of these embodiments, the sugar is glucose. In some embodiments, the buffer does not comprise DMSO.


In certain embodiments, the buffer comprises a sugar in an amount ranging from about 15 mM to about 35 mM; KCl in an amount ranging from about 5 mM to about 10 mM; MgCl2 in an amount ranging from about 10.5 mM to about 20 mM; Na2HPO4/NaH2PO4 in an amount ranging from about 90 mM to about 105 mM; HEPES in an amount equal to or less than 25 mM; and DMSO in an amount equal to or less than 5% by volume of the total buffer volume. In some of these embodiments, the sugar may comprise a monosaccharide and/or a sugar alcohol. In some of these embodiments, the sugar is mannitol and/or glucose. In some of these embodiments, the sugar is glucose. In some embodiments, the buffer does not comprise DMSO.


In certain embodiments, the buffer comprises glucose in an amount of about 15 mM; KCl in an amount of about 6 mM; MgCl2 in an amount of about 10.5 mM Na2HPO4/NaH2PO4 in an amount of about 105 mM; HEPES in an amount ranging from about 15 mM; and DMSO in an amount of about 2.5% by volume of total buffer volume.


In certain embodiments, the buffer comprises glucose in an amount of about 30 mM; KCl in an amount of about 10 mM; MgCl2 in an amount of about 20 mM; Na2HPO4/NaH2PO4 in an amount of about 105 mM; and HEPES in an amount of about 5 mM. In some embodiments, DMSO is specifically excluded from the buffer.


In certain embodiments, the buffer comprises glucose in an amount of about 31 mM; KCl in an amount of about 5 mM; and MgCl2 in an amount of about 15 mM; and Na2HPO4/NaH2PO4 in an amount of about 90 mM. In some embodiments, one or more of HEPES and DMSO is/are specifically excluded from the buffer.


In certain embodiments, the buffer comprises glucose in an amount of about 30 mM; KCl in an amount of about 5 mM; and MgCl2 in an amount of about 15 mM; Na2HPO4/NaH2PO4 in an amount of about 90 mM; and HEPES in an amount of about 10 mM. In some embodiments, DMSO is specifically excluded from the buffer.


In certain embodiments, the buffer comprises glucose in an amount of about 25 mM; KCl in an amount of about 15 mM; and MgCl2 in an amount of about 25 mM; Na2HPO4/NaH2PO4 in an amount of about 120 mM; and HEPES in an amount of about 10 mM. In some embodiments, DMSO is specifically excluded from the buffer.


In certain embodiments, the pH of the buffer may be adjusted. In some embodiments, the buffer is adjusted to a pH of between 6.5 and 8. In some embodiments, the buffer is adjusted to a pH between about 7.0 and 7.6. In some embodiments, the buffer is adjusted to a pH between about 6.9 and 7.2, or between about 7.0 and 7.1. In some embodiments, the buffer is adjusted to a pH of about 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, or 8.0. In certain embodiments, the buffer is adjusted to a pH of about 7.0 or 7.1.


In certain embodiments, the conductivity of the buffer is between about 7.0 ms/cm to about 16.0 ms/cm, about 9.0 ms/cm to about 16.0 ms/cm, about 11.0 ms/cm to about 16.0 ms/cm, or about 13.0 ms/cm to about 16.0 ms/cm. In some embodiments, the conductivity of the buffer is between about 7.0 ms/cm to about 15.0 ms/cm, about 9.0 ms/cm to about 15.0 ms/cm, about 11.0 ms/cm to about 15.0 ms/cm, or about 13.0 ms/cm to about 15.0 ms/cm. In some embodiments, the conductivity of the buffer is about 7.0 ms/cm, about 7.1 ms/cm, about 7.2 ms/cm, about 7.3 ms/cm, about 7.4 ms/cm, about 7.5 ms/cm, about 7.6 ms/cm, about 7.7 ms/cm, about 7.8 ms/cm, about 7.9 ms/cm, about 8.0 ms/cm, about 8.1 ms/cm, about 8.2 ms/cm, about 8.3 ms/cm, about 8.4 ms/cm, about 8.5 ms/cm, about 8.6 ms/cm, about 8.7 ms/cm, about 8.8 ms/cm, about 8.9 ms/cm, about 9.0 ms/cm, about 9.1 ms/cm, about 9.2 ms/cm, about 9.3 ms/cm, about 9.4 ms/cm, about 9.5 ms/cm, about 9.6 ms/cm, about 9.7 ms/cm, about 9.8 ms/cm, about 9.9 ms/cm, about 10.0 ms/cm, about 10.1 ms/cm, about 10.2 ms/cm, about 10.3 ms/cm, about 10.4 ms/cm, about 10.5 ms/cm, about 10.6 ms/cm, about 10.7 ms/cm, about 10.8 ms/cm, about 10.9 ms/cm, about 11.0 ms/cm, about 11.1 ms/cm, about 11.2 ms/cm, about 11.3 ms/cm, about 11.4 ms/cm, about 11.5 ms/cm, about 11.6 ms/cm, about 11.7 ms/cm, about 11.8 ms/cm, about 11.9 ms/cm, about 12.0 ms/cm, about 12.1 ms/cm, about 12.2 ms/cm, about 12.3 ms/cm, about 12.4 ms/cm, about 12.5 ms/cm, about 12.6 ms/cm, about 12.7 ms/cm, about 12.8 ms/cm, about 12.9 ms/cm, about 13.0 ms/cm, about 13.1 ms/cm, about 13.2 ms/cm, about 13.3 ms/cm, about 13.4 ms/cm, about 13.5 ms/cm, about 13.6 ms/cm, about 13.7 ms/cm, about 13.8 ms/cm, about 13.9 ms/cm, about 14.0 ms/cm, about 14.1 ms/cm, about 14.2 ms/cm, about 14.3 ms/cm, about 14.4 ms/cm, about 14.5 ms/cm, about 14.6 ms/cm, about 14.7 ms/cm, about 14.8 ms/cm, about 14.9 ms/cm, about 15.0 ms/cm, about 15.1 ms/cm, about 15.2 ms/cm, about 15.3 ms/cm, about 15.4 ms/cm, about 15.5 ms/cm, about 15.6 ms/cm, about 15.7 ms/cm, about 15.8 ms/cm, about 15.9 ms/cm, or about 16.0 ms/cm. In certain embodiments, the conductivity of the buffer is about 11.6, 12.8, or 14.3.


In some embodiments, the osmolality of the buffer is lower than the osmolality of the cells being transfected (i.e., also known as “intracellular osmolality”). In some embodiments, the osmolality of the buffer ranges from about 250 mOsm/kg H2O to about 1255 mOsm/kg H2O, about 250 mOsm/kg H2O to about 1100 mOsm/kg H2O, about 250 mOsm/kg H2O to about 900 mOsm/kg H2O, about 250 mOsm/kg H2O to about 700 mOsm/kg H2O, about 250 mOsm/kg H2O to about 500 mOsm/kg H2O, about 250 mOsm/kg H2O to about 400 mOsm/kg H2O, or about 250 mOsm/kg H2O to about 360 mOsm/kg H2O. In some embodiments, the osmolality is about 360 mOsm/kg H2O to about 1255 mOsm/kg H2O, about 360 mOsm/kg H2O to about 1100 mOsm/kg H2O, about 360 mOsm/kg H2O to about 900 mOsm/kg H2O, about 360 mOsm/kg H2O to about 700 mOsm/kg H2O, about 360 mOsm/kg H2O to about 500 mOsm/kg H2O, about 360 mOsm/kg H2O to about 400 mOsm/kg H2O. In some embodiments, the osmolality is about 250 mOsm/kg H2O, 255 mOsm/kg H2O, 260 mOsm/kg H2O, 270 mOsm/kg H2O, 275 mOsm/kg H2O, about 280 mOsm/kg H2O, about 285 mOsm/kg H2O, about 290 mOsm/kg H2O, about 300 mOsm/kg H2O, about 305 mOsm/kg H2O, about 310 mOsm/kg H2O, about 315 mOsm/kg H2O, about 320 mOsm/kg H2O, about 325 mOsm/kg H2O, about 330 mOsm/kg H2O, about 335 mOsm/kg H2O, about 340 mOsm/kg H2O, about 345 mOsm/kg H2O, about 350 mOsm/kg H2O, about 355 mOsm/kg H2O, about 360 mOsm/kg H2O, about 365 mOsm/kg H2O, about 370 mOsm/kg H2O, about 375 mOsm/kg H2O, about 380 mOsm/kg H2O, about 385 mOsm/kg H2O, about 390 mOsm/kg H2O, about 395 mOsm/kg H2O, or about 400 mOsm/kg H2O. In certain embodiments, the osmolality is about 280 mOsm/kg H2O, about 292 mOsm/kg H2O, about 340 mOsm/kg H2O, or about 362 mOsm/kg H2O.


In some embodiments, the buffer is selected from one or more of the exemplary buffers set forth in Tables 5 and 6. In certain embodiments, the buffer is selected from Buffer 1, Buffer 2, or Buffer 3.


In some embodiments, the buffer of the invention is used in conjunction with an UltraPorator™ electroporation apparatus and cartridge (or, cassette); see, PCT/US20/59984 (filed Nov. 11, 2020) and U.S. patent application Ser. No. 17/095,028 (filed Nov. 11, 2020). This apparatus is designed to enable rapid manufacturing for a range of gene and cell therapies. UltraPorator™ is a high-throughput, semi-closed electroporation system for electroporation of large quantities of cells in a single operation. The UltraPorator™ system is an advancement over current electroporation devices by significantly reducing the processing time and contamination risk. For example, UltraPorator may be utilized as a scale-up and commercialization solution for decentralized chimeric antigen receptor (CAR) T-cell manufacturing, such as in the UltraCAR-T™ manufacturing of T-cells reprogrammed to target cancer antigens in vivo.


Buffers of the invention are surprisingly effective in producing high cell transfection efficiencies when electroporation is performed using the buffers in the UltraPorator™ electroporation apparatus and/or cartridge (or, cassette); see, PCT/US20/59984 (filed Nov. 11, 2020) and U.S. patent application Ser. No. 17/095,028.


b. Methods Utilizing the EP Buffer and Recombinant Cells Produced Using Those Methods


In another aspect of the invention, a method is provided that utilizes the buffer according to the invention to introduce biologically active material (e.g., DNA or RNA) into cells via electric current (i.e., electroporation). The method comprises forming a suspension by combining cells obtained from a human along with an exogenous biological material into the buffer of the invention, and then applying an electric current in the form of a voltage pulse to the suspension, thereby facilitating the introduction of the biological material into the cells.


In certain embodiments, the voltage pulse may have a field strength of up to 1 to 10 kV*cm-1 and a duration of 5 to 250 μs and a current density of at least 2 A*cm-2. In certain embodiments, the voltage pulse permits the biologically active material (e.g., DNA) to be transfected directly into the cell nucleus of animal and human cells. In certain embodiments, a current flow following the voltage pulse without interruption, having a current density of 2 to 14 A*cm-2, preferably up to 5 A*cm-2, and a duration of 1 to 100 ms, may also be applied.


Using the method according to the invention, the transfection of biologically active material into cells, including into the nucleus of animal cells, may be optimized. In this case, the biologically active material (e.g., nucleic acids, polypeptides, or the like) can be introduced into quiescent or dividing animal cells with a high efficiency.


In some embodiments, the cells are exposed to the buffer for less than 10 minutes. For example, the cells may be exposed to the buffer for less than 9 minutes, less than 8 minutes, less than 7 minutes, less than 6 minutes, less than 5 minutes, less than 4 minutes, less than 3 minutes, less than 2 minutes, or less than 1 minute.


In some embodiments, the method is used to introduce biologically active material into primary human blood cells, pluripotent precursor cells of human blood, as well as primary human fibroblasts and endothelial cells. In some embodiments, the cells are human blood cells, for example immune cells. In certain embodiments, the immune cells are neutrophils, eosinophils, basophils, mast cells, monocytes, macrophages, dendritic cells, natural killer cells, and lymphocytes (B cells and T cells), or some combination thereof. In some embodiments, the lymphocytes are T-cells. In certain embodiments, the cells are obtained from a patient.


In some embodiments, the biological material includes a nucleic acid, peptide, polypeptide, protein, enzyme, RNP, or some combination thereof. In some embodiments, the biological material is heterologous to the cells. In some embodiments, the biological material is partially or fully synthetic.


In some embodiments, the nucleic acid is selected from DNA or RNA. In some embodiments, the DNA may comprise cDNA. In some embodiments, the RNA may comprise mRNA, tRNA, mRNA, lncRNA, sRNA, or a combination thereof. In some embodiments, the nucleic acid is a recombinant nucleic acid. In some embodiments, the peptide comprises a polypeptide, protein, enzyme, antibody, antibody fragment, or combination thereof. In some embodiments, the peptide is recombinant.


Methods utilizing the buffer of the invention result in desirably high transfection yields, especially as compared to methods utilizing other electroporation buffers. In some embodiments, the transfection yield with a buffer of the invention is at least about 1.1 times that of the transfection yield with a control (prior art) buffer. For example, the transfection yield with a buffer of the invention may be about 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 2.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, or 5.0 times higher than that of a control (prior art) buffer. In some embodiments, the transfection yield with a buffer of the invention may be greater than 5 times than that of a control (prior art) buffer, such as 6, 7, 8, 9, or 10 times higher. In certain embodiments, the transfection yield with a buffer of the invention is 1.35, 1.41, 1.46, 1.97, 1.98, 2.05, 2.12, 2.40, or 2.44 times higher than that of a control (prior art) buffer.


Methods utilizing the buffer of the invention result in desirably high transfected cell recovery yields, especially as compared to methods utilizing other electroporation buffers. In some embodiments, the transfected cell recovery yield with a buffer of the invention is at least about 1.1 times that of the transfected cell recovery yield with a control (prior art) buffer. For example, the transfected cell recovery yield with a buffer of the invention may be about 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 2.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, or 5.0 higher than that of a control (prior art) buffer. In some embodiments, the transfected cell recovery yield with a buffer of the invention may be greater than 5 times than that of a control (prior art) buffer. In certain embodiments, the transfected cell recovery yield with a buffer of the invention is 1.53, 1.66, 1.72, 1.80, 2.06, 2.17, 2.23, 2.34, or 2.61 times higher than that of a control (prior art) buffer.


In another aspect of the invention, recombinant cells are provided. In some embodiments, recombinant immune cells are produced using the method of the invention. In certain embodiments, the recombinant immune cell is a modified T-cell. In some embodiments, the modified T-cell is a chimeric antigen receptor (CAR) T-cell. In some embodiments, the CAR-T cell is administered to a patient for therapeutic purposes.


c. Electroporation Apparatuses and Their Methods of Use


An exemplary electroporation apparatus comprises: one or more chambers configured to store the buffer and cells during an electroporation process; one or more pairs of electrodes configured to generate electric fields within the one or more chambers during the electroporation process, each electric field corresponding to one chamber; and a flow channel configured to transport the cells during a cell collection process after the electroporation process.


In some embodiments, the apparatus comprises one chamber, two chambers, three chambers, four chambers, five chambers, six chambers, seven chambers, eight chambers, nine chambers, ten chambers, or ten or more chambers. In certain embodiments, the apparatus utilizes continuous flow or a microfluidic system.


In some embodiments, the electroporation apparatus further comprises a pump for pumping a liquid medium from the flow channel into at least one of the chambers during a collection process, wherein the liquid medium is obtained at the inlet port. In some embodiments, the pump comprises a valve or valves connecting the one or more chambers to the flow channel. In some embodiments, the valve or valves are opened one at a time. In some embodiments, the valve or valves permit only one-directional flow of fluid. In some embodiments, each valve corresponds to one chamber. In some embodiments, each valve corresponding to the chamber valves is a pinch-valve or pinch-type valve. In some embodiments, each of the valves operates using a spring motion, a lever motion, or a piston motion.


In some embodiments, the one or more chambers comprises a given chamber; each electrode of the pair of electrodes is located on opposite sides of the given chamber; and each electrode of the pair of electrodes comprises both an interior portion inside the given chamber and an exterior portion external to the given chamber.


In some embodiments, the electroporation apparatus further comprises: an inlet port; an outlet port; and one or more flanking flow channels connecting the inlet port and the outlet port to the flow channel.


In some embodiments, the electroporation apparatus further comprises: a pump for pumping a liquid medium from the flow channel into at least one of the chambers during a collection process, wherein the liquid medium is obtained at the inlet port.


In some embodiments, the electroporation apparatus further comprises: a surface comprising a one or more openings leading to the one or more chambers; and an airflow channel below the openings and connecting airflow between the chambers.


In some embodiments, the electroporation apparatus further comprises: a vent or air filter connecting the airflow channel to an exterior of the electroporation apparatus.


In some embodiments, the electroporation apparatus further comprises: a seal configured to cover the one or more openings. In some embodiments, each chamber in the electroporation apparatus comprises a shape which narrows toward the respective valve(s). In some embodiments, the electroporation apparatus further comprises a pair of electrodes wherein each electrode of the electrode pair is located on opposite sides of each chamber. The distance between the two electrodes in an electrode pair is referred to as the “gap distance” or “separation distance.” This distance spans the width of the chamber.


In some embodiments, each of the one or more chambers comprises a gap distance of about 0.1 mm to about 20 mm, about 0.5 mm to about 10 mm, about 1 mm to about 7 mm, or about 1 mm to about 4 mm. In some embodiments, the gap distance is about 0.5 mm, 1.0 mm, 1.5 mm, 2.0 mm, 2.5 mm, 3.0 mm, 3.5 mm, 4.0 mm, 4.5 mm, 5.0 mm, 5.5 mm, 6.0 mm, 6.5 mm, 7.0 mm, 7.5 mm, or 8.0 mm. In some embodiments, a gap distance of about 2.5 mm, 2.6 mm, 2.7 mm, 2.8 mm, 2.9 mm, 3.0 mm, 3.1 mm, 3.2 mm, 3.3 mm, 3.4 mm, 3.5 mm, 3.6 mm, 3.7 mm, 3.8 mm, 3.9 mm, or 4.0 mm. In some embodiments, the gap distance is less than about 4 mm, less than about 3.5 mm, less than about 3.0 mm, less than about 2.5 mm, less than about 2.0 mm, less than about 1.5 mm, or less than about 1.0 mm. In some embodiments, a gap distance of less than about 4.0 mm improves the electroporation performance of the buffer provided herein.


In some embodiments, each electrode of the pair of electrodes of the electroporation apparatus comprises: an interior portion inside the given chamber; and an exterior portion external to the given chamber, wherein each pair of electrodes is configured to connect to an electric circuit. In some embodiments, the interior portion inside the given chamber has an elliptical face and comprises a gold coating.


In some embodiments, each chamber of the electroporation apparatus is configured to store a volume of at least about 50 μL, at least about 100 μL, at least about 150 at least about μL, at least about 200 μL, at least about 250 μL, at least about 300 μL, at least about 350 μL, at least about 400 μL, at least about 450 μL, at least about 150 μL, at least about 500 μL, at least about 550 μL, at least about 600 μL, at least about 650 μL, at least about 700 μL, at least about 750 μL, at least about 800 μL, at least about 850 μL, at least about 900 μL, at least about 950 μL, or at least about 1000 μL (1.0 mL).


In some embodiments, the chambers of the electroporation apparatus, in combination, are configured to store at least about 500 μL, at least about 1.0 mL, at least about 1.2 mL, at least about 1.4 mL, at least about 1.6 mL, at least about 1.8 mL, at least about 2.0 mL, at least about 2.2 mL, at least about 2.4 mL, at least about 2.6 mL, at least about 2.8 mL, at least about 3.0 mL, at least about 3.2 mL, at least about 3.4 mL, at least about 3.6 mL, at least about 3.8 mL, at least about 4.0 mL, at least about 4.2 mL, at least about 4.4 mL, at least about 4.6 mL, at least about 4.8 mL, at least about 5.0 mL, at least about 5.2 mL, at least about 5.4 mL, at least about 5.6 mL, at least about 5.8 mL, at least about 6.0 mL, at least about 6.2 mL, at least about 6.4 mL, at least about 6.6 mL, at least about 6.8 mL, or at least about 7.0 mL of cells in liquid suspension for electroporation.


In some embodiments, the cells involved in the electroporation process comprises a population selected from a group consisting of: at least 1×108 cells, at least 2×108 cells, at least 3×108 cells, at least 4×108 cells, at least 5×108 cells, at least 6×108 cells, at least 7×108 cells, at least 8×108 cells, at least 9×108 cells, at least 1×109 cells, at least 2×109 cells, at least 3×109 cells, at least 4×109 cells, at least 5×109 cells, at least 6×109 cells, at least 7×109 cells, at least 8×109 cells, at least 9×109 cells, at least 1×1010 cells, at least 2×1010 cells, at least 3×1010 cells, at least 4×1010 cells, at least 5×1010 cells, at least 6×1010 cells, at least 7×1010 cells, at least 8×1010 cells, at least 9×101° cells, at least 1×1011 cells, at least 2×1011 cells, at least 3×1011 cells, at least 4×1011 cells, at least 5×1011 cells, at least 6×1011 cells, at least 7×1011 cells, at least 8×1011 cells, at least 9×1011 cells, at least 1×1012 cells, at least 2×1012 cells, at least 3×1012 cells, at least 4×1012 cells, at least 5×1012 cells, at least 6×1012 cells, at least 7×1012 cells, at least 8×1012 cells, and at least 9×1012.


In some embodiments, the apparatus of the invention comprises an UltraPorator™ electroporation apparatus and cartridge (see, PCT/US20/59984 and U.S. patent application Ser. No. 17/095,028). As noted above, the UltraPorator™ electroporation apparatus is designed to enable rapid manufacturing for a range of gene and cell therapies. The apparatus may be utilized as a scale-up and commercialization solution for decentralized CAR T-cell manufacturing, such as in the UltraCAR-T™ manufacturing of T-cells reprogrammed to target cancer antigens in vivo.


In some embodiments, the apparatus of the invention is used in a method of electroporation, the method comprising: executing an electroporation process by generating an electric field within a chamber using a pair of electrodes, wherein the chamber is configured to store the buffer and cells during the electroporation process; and executing a cell collection process by: opening a valve connected to the chamber; and transporting the buffer and cells to an outlet port using a flow channel connected to the valve, wherein the chamber, the electrode pair, the valve, the outlet port, and the flow channel are each located within an electroporation apparatus.


In some embodiments, the step of executing a cell collection process further comprises: pumping, through use of a pump, a liquid medium from the flow channel into the chamber, wherein the liquid medium is obtained at an inlet port, and wherein the inlet port and the outlet port are connected to the flow channel by a flanking flow channel within the electroporation apparatus.


In some embodiments, the cell collection process further comprises: draining the chamber into the flow channel, wherein pressure within the chamber is maintained via a vent or air filter connected to an air flow channel running between the chamber and another chamber.


In some embodiments, the method of electroporation further comprises: depositing the cells into an opening leading to the chamber holding the buffer; applying a seal to the opening; and connecting the electrode pair to at least one circuit by, for example, inserting the electroporation apparatus into a docking station.


In some embodiments, the method utilizes one or more of the exemplary buffers set forth in Tables 5 and 6. In certain embodiments, the method utilizes Buffer 1, Buffer 2, or Buffer 3.


In some embodiments, the method is performed in an UltraPorator™ electroporation apparatus (see, PCT/US20/59984 and U.S. patent application Ser. No. 17/095,028). In certain embodiments, the method is performed in an UltraPorator™ electroporation apparatus and utilizes one or more of the exemplary buffers set forth in Tables 5 and 6. In certain embodiments, the method is performed in an UltraPorator™ electroporation apparatus and utilizes Buffer 1, Buffer 2, or Buffer 3 (as set forth in Table 5).


d. Electroporation Systems


In another aspect of the invention, a system for electroporation is provided. In some embodiments, the system for electroporation comprises an electroporation apparatus, as described herein, and an electroporation buffer, as described herein. In some embodiments, the electroporation system comprises and UltraPorator™ electroporation apparatus and cartridge (see, PCT/US20/59984 and U.S. patent application Ser. No. 17/095,028). As noted above, the UltraPorator™ electroporation apparatus is designed to enable rapid manufacturing for a range of gene and cell therapies. The device may be utilized as a scale-up and commercialization solution for decentralized CAR T-cell manufacturing, such as in the UltraCAR-T™ manufacturing of T-cells reprogrammed to target cancer antigens in vivo.


In some embodiments, the system for electroporation further comprises a buffer is from one or more of the exemplary buffers set forth in Tables 5 and 6. In certain embodiments, the system for electroporation comprises a buffer selected from Buffer 1, Buffer 2, or Buffer 3. It has been found that systems comprising an UltraPorator™ device and one of Buffers 1, 2, or 3 result in surprisingly high cell transfection efficiencies, as compared to systems comprising an UltraPorator™ device and a control buffer.


e. Kits for Electroporation


In another aspect of the invention, a kit for electroporation is provided. The kit may include any of the buffers as described herein. In some embodiments, the kit comprises one or more of the exemplary buffers set forth in Tables 5 and 6. In certain embodiments, the kit comprises a buffer selected from Buffer 1, Buffer 2, or Buffer 3.


In some embodiments, the kit includes one or more containers filled with a buffer according to the invention and other suitable reagents and/or devices. For example, the kit may additionally comprise a vector comprising a nucleic acid of interest. In some embodiments, the kit may include a dropper, pipette, and/or cuvette. In some embodiments, the buffer may be packaged in aliquoted containers or as a stock solution.


In some embodiments, the kit further comprises packaging to safely transport the buffer and any additional reagents and/or devices. In some embodiments, the kit includes information about the contents of the buffer and any additional reagents. Further, the kit may comprise written materials, for example a user manual or answers to frequently asked questions.


2. Chemical Methods


Chemical methods for introducing a polynucleotide into a host cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (e.g., an artificial membrane vesicle).


3. Viral Vectors and Delivery Methods


In the case where a viral delivery system is utilized, an exemplary delivery vehicle is a liposome. Lipid formulations can be used for the introduction of the nucleic acids into a host cell (in vitro, ex vivo, or in vivo). In another aspect, the nucleic acid can be associated with a lipid. The nucleic acid associated with a lipid can be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide, entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid. Lipid, lipid/DNA or lipid/expression vector associated compositions are not limited to any particular structure in solution. For example, they can be present in a bilayer structure, as micelles, or with a “collapsed” structure. They can also simply be interspersed in a solution, possibly forming aggregates that are not uniform in size or shape. Lipids are fatty substances which can be naturally occurring or synthetic lipids. For example, lipids include the fatty droplets that naturally occur in the cytoplasm as well as the class of compounds which contain long-chain aliphatic hydrocarbons and their derivatives, such as fatty acids, alcohols, amines, amino alcohols, and aldehydes.


Lipids suitable for use can be obtained from commercial sources. For example, dimyristyl phosphatidylcholine (“DMPC”) can be obtained from Sigma, St. Louis, Mo.; dicetyl phosphate (“DCP”) can be obtained from K & K Laboratories (Plainview, N.Y.); cholesterol (“Chol”) can be obtained from Calbiochem-Behring; dimyristyl phosphatidylglycerol (“DMPG”) and other lipids can be obtained from Avanti Polar Lipids, Inc. (Birmingham, Ala.). Stock solutions of lipids in chloroform or chloroform/methanol can be stored at about −20° C. Chloroform is used as the only solvent since it is more readily evaporated than methanol. “Liposome” is a generic term encompassing a variety of single and multilamellar lipid vehicles formed by the generation of enclosed lipid bilayers or aggregates. Liposomes can be characterized as having vesicular structures with a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution. The lipid components undergo self-rearrangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers (Ghosh et al., Glycobiology 5: 505-10 (1991)). However, compositions that have different structures in solution than the normal vesicular structure are also encompassed. For example, the lipids can assume a micellar structure or merely exist as nonuniform aggregates of lipid molecules. Also contemplated are lipofectamine-nucleic acid complexes.


Also provided herein are viral-based delivery systems, in which a nucleic acid of the present invention is inserted. Representative viral expression vectors include, but are not limited to, the adenovirus-based vectors (e.g., the adenovirus-based Per.C6 system available from Crucell, Inc. (Leiden, The Netherlands)), adeno-associated virus based vectors, lentivirus-based vectors (e.g., the lentiviral-based pLPI from Life Technologies (Carlsbad, Calif.)), retroviral vectors (e.g., the pFB-ERV plus pCFB-EGSH), and herpes virus-based vectors. In an embodiment, the viral vector is a lentivirus vector. Vectors derived from retroviruses such as the lentivirus are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene and its propagation in daughter cells. Lentiviral vectors have the added advantage over vectors derived from onco-retroviruses such as murine leukemia viruses in that they can transduce non-proliferating cells, such as hepatocytes. They also have the added advantage of low immunogenicity. In general, and in embodiments, a suitable vector contains an origin of replication functional in at least one organism, a promoter sequence, convenient restriction endonuclease sites, and one or more selectable markers, (e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).


In some embodiments, a lentiviral vector is provided comprising a backbone and a nucleic acid sequence encoding one or more miRNA(s) and a CAR. Optionally, the vector further comprises a nucleic acid encoding a cytokine and a cell tag such as truncated HER1, CD20t-1 or a full length CD20.


In some embodiments, the nucleic acid encoding one or more miRNA(s) and a CAR is cloned into a vector comprising lentiviral backbone components. Exemplary backbone components include, but are not limited to, pFUGW, and pSMPUW. The pFUGW lentiviral vector backbone is a self-inactivating (SIN) lentiviral vector backbone and has unnecessary HIV-1 viral sequences removed resulting in reduced potential for the development of neoplasia, harmful mutations, and regeneration of infectious particles. In some embodiments, the vector encoding one or more miRNA(s) and a CAR also encodes a cytokine in a single construct. In some embodiments, the one or more miRNA(s) and a CAR and cytokine are encoded on two separate lentiviral vectors. In some embodiments, the cytokine is expressed with a cell tag. In some embodiments, one or more miRNA(s) and a CAR can be co-expressed with the cytokine and the cell tag from a single lentiviral vector. In further embodiments, one or more miRNA(s) and a CAR can be under the control of an inducible promoter. In another embodiment, the cytokine can be under the control of an inducible promoter. In one aspect, the inducible promoter can be a gene switch ligand inducible promoter. In some cases, an inducible promoter can be a small molecule ligand-inducible two polypeptide ecdysone receptor-based gene switch, such as RHEOSWITCH® gene switch.


Other suitable vectors include integrating expression vectors, which can randomly integrate into the host cell's DNA, or can include a recombination site to enable the specific recombination between the expression vector and the host cell's chromosome. Such integrating expression vectors can utilize the endogenous expression control sequences of the host cell's chromosomes to effect expression of the desired protein. Examples of vectors that integrate in a site specific manner include, for example, components of the flp-in system from Invitrogen (Carlsbad, Calif.) (e.g., pcDNA™5/FRT), or the cre-lox system, such as can be found in the pExchange-6 Core Vectors from Stratagene (La Jolla, Calif.). Examples of vectors that randomly integrate into host cell chromosomes include, for example, pcDNA3.1 (when introduced in the absence of T-antigen) from Invitrogen (Carlsbad, Calif.), and pCI or pFN10A (ACT) FLEXI™ from Promega (Madison, Wis.). Additional promoter elements, e.g., enhancers, regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription.


In some embodiments, the cell tag gene is cloned into a lentiviral plasmid backbone in frame with the CAR gene. In other embodiments, the cell tag is cloned into a separate lentiviral vector.


4. Non-Viral Vectors and Delivery Systems


a. Sleeping Beauty Transposon System


A polynucleotide encoding one or more miRNA(s) alone or a polynucleotide encoding one or more miRNA(s) and a chimeric receptor, cytokine, and/or cell tag as described herein can be introduced into immune effector cells using non-viral based delivery systems, such as the “Sleeping Beauty (SB) Transposon System,” which refers to a synthetic DNA transposon system for introducing DNA sequences into the chromosomes of vertebrates. An exemplary Sleeping Beauty transposon system is described for example, in U.S. Pat. Nos. 6,489,458 and 8,227,432. As used herein, the Sleeping Beauty transposon system can comprise Sleeping Beauty transposase polypeptides as well as derivatives, functional fragments, and variants thereof, and Sleeping Beauty transposon polynucleotides, derivatives, and functional variants and fragments thereof. In certain embodiments, the Sleeping Beauty transposase is encoded by an mRNA. In some embodiments, the mRNA encodes for a SB10, SB11, SB100x or SB110 transposase. In some embodiments, the mRNA comprises a cap and a poly-A tail.


DNA transposons translocate from one DNA site to another in a simple, cut-and-paste manner. Transposition is a precise process in which a defined DNA segment is excised from one DNA molecule and moved to another site in the same or different DNA molecule or genome. As with other Tc 1/mariner-type transposases, SB transposase inserts a transposon into a TA dinucleotide base pair in a recipient DNA sequence. The insertion site can be elsewhere in the same DNA molecule, or in another DNA molecule (or chromosome). In mammalian genomes, including humans, there are approximately 200 million TA sites. The TA insertion site is duplicated in the process of transposon integration. This duplication of the TA sequence is a hallmark of transposition and used to ascertain the mechanism in some experiments. The transposase can be encoded either within the transposon or the transposase can be supplied by another source, in which case the transposon becomes a non-autonomous element. Non-autonomous transposons are most useful as genetic tools because after insertion they cannot independently continue to excise and re-insert. Sleeping Beauty transposons envisaged to be used as non-viral vectors for introduction of genes into genomes of vertebrate animals and for gene therapy.


Briefly, the Sleeping Beauty system (Hackett et al., Mol Ther 18:674-83, (2010)) was adapted to genetically modify the immune effector cells (Cooper et al., Blood 105:1622-31, (2005)). In one embodiment, this involves two steps: (i) the electro-transfer of DNA plasmids expressing a Sleeping Beauty transposon (Jin et al., Gene Ther 18:849-56, (2011); Kebriaei et al., Hum Gene Ther 23:444-50, (2012)) and Sleeping Beauty transposase and (ii) the propagation and expansion of T cells stably expressing integrants on designer artificial antigen-presenting cells (AaPC) derived from the K562 cell line (also known as AaPCs (Activating and Propagating Cells). In another, embodiment, the second step (ii) is eliminated and the genetically modified T cells are cryopreserved or immediately infused into a patient.


In one embodiment, the Sleeping Beauty transposon systems are described for example in Hudecek et al., Critical Reviews in Biochemistry and Molecular Biology, 52:4, 355-380 (2017), Singh et al., Cancer Res (8):68 (2008). Apr. 15, 2008 and Maiti et al., J Immunother. 36(2): 112-123 (2013).


In some embodiments, one or more miRNA(s), a CAR, a cytokine, and a cell tag can be encoded by a single transposon DNA plasmid vector. In some embodiments, the one or more miRNA(s), CAR, cytokine, and cell tag can be encoded by different transposon DNA plasmid vectors. In further embodiments, one or more miRNA(s) and a CAR can be under the control of an inducible promoter. In another embodiment, the cytokine can be under the control of an inducible promoter. In one aspect, the inducible promoter can be a gene switch ligand inducible promoter. In some cases, an inducible promoter can be a small molecule ligand-inducible two polypeptide ecdysone receptor-based gene switch, such as RHEOSWITCH® gene switch further described below. In certain embodiments, the CAR, cytokine, and cell tag can be configured in one, two or more transposons.


In some embodiments, the MUC16, CD33, or ROR1-specific CARs and other genetic elements are delivered to a cell using the SB11 transposon system, the SB100X transposon system, the SB110 transposon system, the piggyBac transposon system (see, e.g., U.S. Pat. Nos. 6,489,458; 8,227,432, 9,228,180, Wilson et al, “PiggyBac Transposon-mediated Gene Transfer in Human Cells,” Molecular Therapy 15:139-145 (2007) and WO 2016/145146) and/or the piggyBat transposon system (see, e.g., Mitra et al., “Functional characterization of piggyBat from the bat Myotis lucifugus unveils an active mammalian DNA transposon,” Proc. Natl. Acad. Sci USA 110:234-239 (2013). Additional transposases and transposon systems are provided in U.S. Pat. Nos. 7,148,203; 8,227,432; U.S. Patent Publn. No. 2011/0117072; Mates et al., Nat Genet, 41(6):753-61 (2009). doi: 10.1038/ng.343. Epub 2009 May 3, Gene Ther., 18(9):849-56 (2011). doi: 10.1038/gt.2011.40. Epub 2011 Mar. 31 and in Ivics et al., Cell. 91(4):501-10, (1997).


In certain embodiments, the vector is a Sleeping Beauty plasmid that comprises a left transposon repeat region and a right transposon repeat region. In certain embodiments, the left transposon repeat region comprises a nucleic acid comprising SEQ ID NO: 580 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 580, or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 580. In certain embodiments, the right transposon repeat region comprises a nucleic acid comprising SEQ ID NO: 581 or a functional fragment or variant thereof. In certain embodiments, the functional variant has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with SEQ ID NO: 581, or hybridizes under stringent hybridization conditions with the complement of SEQ ID NO: 581.


b. Recombinase-Based Delivery Systems


In other embodiments, nucleic acids encoding one or more miRNA(s), a CAR, a cytokine, and/or a cell tag can be integrated into the immune effector cell's DNA through a recombinase and integrating expression vectors. Such vectors can randomly integrate into the host cell's DNA, or can include a recombination site to enable the specific recombination between the expression vector and the host cell's chromosome. Such integrating expression vectors can utilize the endogenous expression control sequences of the host cell's chromosomes to effect expression of the desired protein. In some embodiments, targeted integration is promoted by the presence of sequences on the donor polynucleotide that are homologous to sequences flanking the integration site. For example, targeted integration using the donor polynucleotides described herein can be achieved following conventional transfection techniques, e.g. techniques used to create gene knockouts or knockins by homologous recombination. In other embodiments, targeted integration is promoted both by the presence of sequences on the donor polynucleotide that are homologous to sequences flanking the integration site, and by contacting the cells with donor polynucleotide in the presence of a site-specific recombinase. By a site-specific recombinase, or simply a recombinase, it is meant a polypeptide that catalyzes conservative site-specific recombination between its compatible recombination sites. As used herein, a site-specific recombinase includes native polypeptides as well as derivatives, variants and/or fragments that retain activity, and native polynucleotides, derivatives, variants, and/or fragments that encode a recombinase that retains activity.


The recombinases can be introduced into a target cell before, concurrently with, or after the introduction of a targeting vector. The recombinase can be directly introduced into a cell as a protein, for example, using liposomes, coated particles, or microinjection. Alternately, a polynucleotide, either DNA or messenger RNA, encoding the recombinase can be introduced into the cell using a suitable expression vector. The targeting vector components described above are useful in the construction of expression cassettes containing sequences encoding a recombinase of interest. However, expression of the recombinase can be regulated in other ways, for example, by placing the expression of the recombinase under the control of a regulatable promoter (i.e., a promoter whose expression can be selectively induced or repressed).


A recombinase can be from the Integrase or Resolvase families. The Integrase family of recombinases has over one hundred members and includes, for example, FLP, Cre, and lambda integrase. The Integrase family, also referred to as the tyrosine family or the lambda integrase family, uses the catalytic tyrosine's hydroxyl group for a nucleophilic attack on the phosphodiester bond of the DNA. Typically, members of the tyrosine family initially nick the DNA, which later forms a double strand break. Examples of tyrosine family integrases include Cre, FLP, SSV1, and lambda (λ) integrase. In the resolvase family, also known as the serine recombinase family, a conserved serine residue forms a covalent link to the DNA target site (Grindley, et al., (2006) Ann Rev Biochem 16:16).


In one embodiment, the recombinase is an isolated polynucleotide sequence comprising a nucleic acid sequence that encodes a recombinase selecting from the group consisting of a SPβc2 recombinase, a SF370.1 recombinase, a Bxb1 recombinase, an A118 recombinase and a ϕRv1 recombinase. Examples of serine recombinases are described in detail in U.S. Pat. No. 9,034,652.


Recombinases for use in the practice of the present invention can be produced recombinantly or purified as previously described. Polypeptides having the desired recombinase activity can be purified to a desired degree of purity by methods known in the art of protein ammonium sulfate precipitation, purification, including, but not limited to, size fractionation, affinity chromatography, HPLC, ion exchange chromatography, heparin agarose affinity chromatography (e.g., Thorpe & Smith, Proc. Nat. Acad. Sci. 95:5505-5510, 1998.)


In one embodiment, the recombinases can be introduced into the eukaryotic cells that contain the recombination attachment sites at which recombination is desired by any suitable method. Methods of introducing functional proteins, e.g., by microinjection or other methods, into cells are well known in the art. Introduction of purified recombinase protein ensures a transient presence of the protein and its function, which is often a preferred embodiment. Alternatively, a gene encoding the recombinase can be included in an expression vector used to transform the cell, in which the recombinase-encoding polynucleotide is operably linked to a promoter which mediates expression of the polynucleotide in the eukaryotic cell. The recombinase polypeptide can also be introduced into the eukaryotic cell by messenger RNA that encodes the recombinase polypeptide. It is generally preferred that the recombinase be present for only such time as is necessary for insertion of the nucleic acid fragments into the genome being modified. Thus, the lack of permanence associated with most expression vectors is not expected to be detrimental. One can introduce the recombinase gene into the cell before, after, or simultaneously with, the introduction of the exogenous polynucleotide of interest. In one embodiment, the recombinase gene is present within the vector that carries the polynucleotide that is to be inserted; the recombinase gene can even be included within the polynucleotide.


In one embodiment, a method for site-specific recombination comprises providing a first recombination site and a second recombination site; contacting the first and second recombination sites with a prokaryotic recombinase polypeptide, resulting in recombination between the recombination sites, wherein the recombinase polypeptide can mediate recombination between the first and second recombination sites, the first recombination site is attP or attB, the second recombination site is attB or attP, and the recombinase is selected from the group consisting of a Listeria monocytogenes phage recombinase, a Streptococcus pyogenes phage recombinase, a Bacillus subtilis phage recombinase, a Mycobacterium tuberculosis phage recombinase and a Mycobacterium smegmatis phage recombinase, provided that when the first recombination attachment site is attB, the second recombination attachment site is attP, and when the first recombination attachment site is attP, the second recombination attachment site is attB


Further embodiments provide for the introduction of a site-specific recombinase into a cell whose genome is to be modified. One embodiment relates to a method for obtaining site-specific recombination in an eukaryotic cell comprises providing a eukaryotic cell that comprises a first recombination attachment site and a second recombination attachment site; contacting the first and second recombination attachment sites with a prokaryotic recombinase polypeptide, resulting in recombination between the recombination attachment sites, wherein the recombinase polypeptide can mediate recombination between the first and second recombination attachment sites, the first recombination attachment site is a phage genomic recombination attachment site (attP) or a bacterial genomic recombination attachment site (attB), the second recombination attachment site is attB or attP, and the recombinase is selected from the group consisting of a Listeria monocytogenes phage recombinase, a Streptococcus pyogenes phage recombinase, a Bacillus subtilis phage recombinase, a Mycobacterium tuberculosis phage recombinase and a Mycobacterium smegmatis phage recombinase, provided that when the first recombination attachment site is attB, the second recombination attachment site is attP, and when the first recombination attachment site is attP, the second recombination attachment site is attB. In an embodiment the recombinase is selected from the group consisting of an A118 recombinase, a SF370.1 recombinase, a SPβc2 recombinase, a ϕRv1 recombinase, and a Bxb1 recombinase. In one embodiment the recombination results in integration.


c. Other Non-Viral Delivery Systems


In other embodiments, nucleic acids encoding one or more miRNA(s), a CAR, a cytokine, and/or a cell tag, can be integrated into the immune effector cell's DNA through gene editing systems that utilize CRISPR, TALEN or Zinc-Finger nucleases.


Regardless of the method used to introduce exogenous nucleic acids into a host cell, in order to confirm the presence of the recombinant DNA sequence in the host cell, a variety of assays can be performed. Such assays include, for example, “molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; “biochemical” assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify peptides or proteins or nucleic acids falling within the scope of the invention.


IX. Engineered T-Cell Receptor (TCR)

In some embodiments, the chimeric receptor comprises an engineered T-cell receptor. The T cell receptor (TCR) is composed of two chains (aβ or γδ) that pair on the surface of the T cell to form a heterodimeric receptor. In some instances, the αβ TCR is expressed on most T cells in the body and is known to be involved in the recognition of specific MHC-restricted antigens. Each α and β chain are composed of two domains: a constant domain (C) that anchors the protein to the cell membrane and is associated with invariant subunits of the CD3 signaling apparatus; and a variable domain (V) that confers antigen recognition through six loops, referred to as complementarity determining regions (CDRs). In some instances, each of the V domains comprises three CDRs; e.g., CDR1, CDR2 and CDR3 with CDR3 as the hypervariable region. These CDRs interact with a complex formed between an antigenic peptide bound to a protein encoded by the major histocompatibility complex (pepMHC) (e.g., HLA-A, HLA-B, HLA-C, HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1, HLA-DRA, or HLA-DRB1 complex). In some instances, the constant domain further comprises a joining region that connects the constant domain to the variable domain. In some cases, the beta chain further comprises a short diversity region which makes up part of the joining region.


Both the α and β chains are highly variable, although the T-cell receptor α chain contains a constant (preserved region), i.e., the Vα 24-Ja18 junction (amino acid sequence GSTLGR or a conservatively substituted amino acid sequence thereof).


In some cases, such TCR are reactive to specific tumor antigen, e.g. NY-ESO, Mage A3, Titin, MART-1, HPV, HBV, MAGE-A4, MAGE-A10, MAGE A3/A6, gp100, MAGE-A 1, or PRAME. In other cases, such TCR are reactive to specific neoantigens expressed within a patient's tumor (i.e. patient-specific, somatic, non-synonymous mutations expressed by tumors). In some cases, engineered TCRs can be affinity-enhanced.


In some embodiments, a TCR is described using the International Immunogenetics (IMGT) TCR nomenclature, and links to the IMGT public database of TCR sequences. For example, there can be several types of alpha chain variable (Vα) regions and several types of beta chain variable (Vβ) regions distinguished by their framework, CDR1, CDR2, and CDR3 sequences. As such, a Vα type can be referred to in IMGT nomenclature by a unique TRAV number. For example, “TRAV21” defines a TCR Vα region having unique framework and CDR1 and CDR2 sequences, and a CDR3 sequence, which is partly defined by an amino acid sequence which is preserved from TCR to TCR but which also includes an amino acid sequence which varies from TCR to TCR Similarly, “TRBV5-1” defines a TCR Vβ region having unique framework and CDR1 and CDR2 sequences, but with only a partly defined CDR3 sequence.


In some cases, the beta chain diversity region is referred to in IMGT nomenclature by the abbreviation TRBD.


In some instances, the unique sequences defined by the IMGT nomenclature are widely known and accessible to those working in the TCR field. For example, they can be found in the IMGT public database and in “T cell Receptor Factsbook”, (2001) LeFranc and LeFranc, Academic Press, ISBN 0-12-441352-8.


In some embodiments, an αβ heterodimeric TCR is, for example, transfected as full length chains having both cytoplasmic and transmembrane domains. In some cases, the TCRs contain an introduced disulfide bond between residues of the respective constant domains, as described, for example, in WO 2006/000830.


In some instances, TCRs described herein are in single chain format, for example see WO 2004/033685. Single chain formats include αβ TCR polypeptides of the Vα-L-Vβ, Vβ-L-Vα, Vα-Cα-L-Vβ, Vα-L-Vβ-Cβ, Vα-Cα-L-Vβ-Cβ types, wherein Vα and Vβ are TCR α and β variable regions respectively, Cα and Cβ are TCR α and β constant regions respectively, and L is a linker sequence. In certain embodiments single chain TCRs of the invention may have an introduced disulfide bond between residues of the respective constant domains, as described in WO 2004/033685.


In some embodiments, the TCR can be an αβ TCR. In some embodiments, the TCR can be a γδ TCR. It should be understood that a TCR of the present disclosure can bind to any of the antigen targets described herein.


In some embodiments, the TCR may be any one of the Vδ1, Vδ2, and Vδ1negVδ2neg TCR subsets. In some embodiments, the engineered cell may express any combination of a Vδ1, Vδ2, Vδ3, Vδ5, Vδ7, or Vδ8 TCR chain with a Vγ2, Vγ3, Vγ7, Vγ8, Vγ9, Vγ10, or Vγ11 TCR chain. In some embodiments, the engineered cell may have essentially identical genetic material. In one aspect, the engineered cell may not contain a chimeric antigen receptor.


The TCR described herein may be associated with a detectable label, a therapeutic agent or a PK modifying moiety. Exemplary detectable labels for diagnostic purposes include, but are not limited to, fluorescent labels, radiolabels, enzymes, nucleic acid probes and contrast reagents.


In some cases, each chain of TCR disclosed herein, for example αβ or γδ, comprises a modified spacer region connecting the constant region of a TCR chain to the transmembrane region. In some cases, a spacer region of each chain of TCR disclosed herein comprises 1) a stalk region and 1 or more stalk extension region(s) adjacent to said stalk region. The stalk and stalk extension regions may, for example, be those as previously described for use in chimeric antigen receptors. In some embodiments, each chain of TCR disclosed herein incorporates a spacer that comprises a stalk region (s) and up to 20 stalk extension regions.


In some instances, the stalk region comprises the extracellular hinge region from TCRα or TCRβ chain or the stalk region comprises a sequence with at least 80% homology to the extracellular hinge region from TCRα or TCRβ chain. In alternative instances, the stalk region comprises any portion of extracellular region of TCRα or TCRβ constant region with at least 80% homology to the extracellular region of TCRα or TCRβ constant region respectively. For example, the stalk region can comprise a sequence with at least 80%, 85%, 90%, 95%, or greater than 95% homology to the any portion of extracellular region of TCRα or TCRβ constant region.


TCR chain heterodimers are formed by inter-chain disulfide bonds in extracellular hinge region of a and (3 chains. In some embodiments, the stalk region comprises a dimerization site. A dimerization site can comprise a disulfide bond formation site. A dimerization site can comprise cysteine residue(s). A stalk region can be capable of forming a disulfide bond. Such a disulfide bond can be formed at a disulfide bond forming site or a dimerization site. In some examples, the dimerization occurs between a and (3 chains of TCR.


In some embodiments, a stalk extension region is used to link the stalk region to the transmembrane region TCR α and (3 chains. In certain embodiments, a stalk extension region is used to link the stalk region to constant region of TCR α and β chains. In certain embodiments, the stalk region and the stalk extension region(s) can be connected via a linker.


In some instances, the stalk extension domain comprises a sequence that is partially homologous to the stalk region. In some instances, each of the stalk extension region comprises a sequence that is homologous to the stalk region, except that the stalk extension region lacks the dimerization site of the stalk region. In some cases, each of the stalk extension region comprises a sequence identical to the stalk region. In other cases, each of the stalk extension regions comprise a sequence identical to the stalk region with at least one amino acid residue substitution relative to the stalk region. In some cases, each of the stalk extension region is not capable of forming a disulfide bond or is not capable of dimerization with a homologous stalk extension region.


In other embodiments, one stalk extension region can be connected to another stalk extension region via a linker. Examples of such linkers can include glycine-serine rich linkers.


In some embodiments, the addition of stalk extension region(s) prevents mispairing of transgenic TCR α and β chains with native TCR α and β chains expressed by T cells that are genetically modified.


In some embodiments, a modified immune effector cell of the present disclosure can comprise a TCR of the present disclosure and a cytokine of the present disclosure. In certain embodiments, the modified immune effector cell can comprise a TCR and a fusion protein comprising IL-15 and IL-15Rα, or a fusion protein comprising functional fragments or variants of such domains.


X. Immune Checkpoint Inhibitors

In some embodiments, an engineered cell of the present disclosure can include an immune checkpoint inhibitor. In certain embodiments, a polynucleotide of the present disclosure can further encode an immune checkpoint inhibitor. In certain embodiments, an engineered cell of the present disclosure can comprise such a polynucleotide.


In certain embodiments, the immune checkpoint inhibitor can be an antibody or a functional fragment or variant thereof. In certain embodiments, the immune checkpoint inhibitor can inhibit the activity of an immune checkpoint protein such as PD1, PD-L1, CTLA-4, TIGIT, 4-1BB, PIK3IP1, CD27, CD28, CD40, CD70, CD122, CD137, OX40 (CD134), GITR, ICOS, A2AR, B7-H3 (CD276), B7-H4 (VTCN1), BTLA, IDO, KIR, LAGS, TIM-3, or VISTA.


In some embodiments, the immune checkpoint inhibitor can be an anti-CTLA-4 antibody. The anti-CTLA-4 antibody (e.g., ipilimumab) has shown durable anti-tumor activities and prolonged survival in participants with advanced melanoma, resulting in its Food and Drug Administration (FDA) approval in 2011. See Hodi et al., Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. (2010) Aug. 19; 363(8):711-23. In some embodiments, the one or more checkpoint inhibitors can be an anti-PD-L1 antibody. In some embodiments, the anti-PD-L1 antibody can be a full-length atezolizumab (anti-PD-L1), avelumab (anti-PD-L1), durvalumab (anti-PD-L1), or a fragment or a variant thereof. In some embodiments, the one or more checkpoint inhibitors can be any one or more of CD27 inhibitor, CD28 inhibitor, CD40 inhibitor, CD122 inhibitor, CD137 inhibitor, OX40 (also known as CD134) inhibitor, GITR inhibitor, ICOS inhibitor, or any combination thereof. In some embodiments, the one or more checkpoint inhibitors can be any one or more of A2AR inhibitor, B7-H3 (also known as CD276) inhibitor, B7-H4 (also known as VTCN1) inhibitor, BTLA inhibitor, IDO inhibitor, KIR inhibitor, LAG3 inhibitor, TIM-3 inhibitor, VISTA inhibitor, or any combination thereof.


In some embodiments, the immune checkpoint inhibitor is an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-CD28 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, an anti-TIM3 antibody, an anti-GITR antibody, an anti-4-1BB antibody, or an anti-OX-40 antibody. In some embodiments, the additional therapeutic agent is an anti-TIGIT antibody. In some embodiments, the immune checkpoint inhibitor is an anti-PD-L1 antibody selected from the group consisting of: BMS935559 (MDX-1105), atexolizumab (MPDL3280A), durvalumab (MEDI4736), and avelumab (MSB0010718C). In some embodiments, the immune checkpoint inhibitor is an anti-CTLA-4 antibody selected from the group consisting of: ipilimumab (YERVOY) and tremelimumab. In some embodiments, the additional therapeutic agent is an anti-LAG-3 antibody selected from the group consisting of: BMS-986016 and LAG525. In some embodiments, the immune checkpoint inhibitor is an anti-OX-40 antibody selected from the group consisting of: MEDI6469, MEDI0562, and MOXR0916. In some embodiments, the additional therapeutic agent is an anti-4-1BB antibody selected from the group consisting of: PF-05082566.


In some embodiments, the engineered cell can include an immune checkpoint inhibitor comprising a PD1-binding moiety. In some embodiments, the PD1-binding moiety (referred to herein as an “anti-PD-1”) is selected from an antibody identified in Table 8 (below), or a functional fragment or variant thereof.









TABLE 8







PD-1 Antibodies










Name
Also Known as
Company
Reference(s)





cemiplimab
Libtayo, cemiplimab.
Regeneron, Sanofi
WO 2015/112800



REGN2810


pembrolizumab
Keytruda, MK-3475,
Merck (MSD),
WO 2008/156712,



SCH 900475,
Schering-Plough
U.S. Pat. No. 8,354,509.



lambrolizumab

U.S. Pat. No. 8,952,136,





U.S. Pat. No. 8,900,587


nivolumab
Opdivo, ONO-4538,
BMS, Medarex, Ono
U.S. Pat. No. 8,728,474.



MDX-1106, BMS-

U.S. Pat. No. 8,779,105,



936558, 5C4

U.S. Pat. No. 8.008,449,





U.S. Pat. No. 9,067,999,





U.S. Pat. No. 9,073,994


toripalimab
JS001, JS-001, TAB001,
Junmeng Biosciences,
Si-Yang Liu et al., J.



triprizumab
Shanghai Junshi,

Hematol. Oncol. 10: 136





TopAlliance Bio
(2017)


sintilimab
Tyvyt, IBI308
Adimab, Innovent,
WO 2017/024465, WO




Lilly
2017/025016, WO





2017/132825, WO





2017/133540


LY3434172

Lilly, Zymeworks
ClinicalTrials.gov





Identifier: NCT03936959


JTX-4014

Jounce Therapeutics
U.S. 2018/0118829;




Inc.
ClinicalTrials.gov





Identifier: NCT03790488;





Papdopolous et al., Cancer






Immunol Immunother






70(3): 763-772 (2021))


609 A
609A
3S Bio; Sunshine
ClinicalTrials.gov




Guojian Pharma
Identifier: NCT03998345


Sym021

Symphogen A/S
Gjetting et al., mAbs 11(4):





666-680 (2019


LZM009

Livzon Pharmaceutical
ClinicalTrials.gov




Group
Identifier: NCT03286296


budigalimab
ABBV-181, PR-
Abbvie
Powderly et al., Annals of



1648817


Oncology 29(8) (2018);






ClinicalTrials.gov





Identifier: NCT03000257


IB
IBI-318
Innovent, Lilly
ClinicalTrials.gov





Identifier: NCT03875157


SCT-I10A

Sinocelltech Ltd.
ClinicalTrials.gov





Identifier: NCT03821363


SG001

CSPC ZhongQi
ClinicalTrials.gov




Pharmaceutical
Identifier: NCT03852823




Technology Co., Ltd.


AMP-224
GSK-2661380
Astra Zeneca, Glaxo
Floudas et al., Clin.




Smith Kline

Colorectal Cancer 18(4)






(2019)


AMG 404
AMG404
Amgen
ClinicalTrials.gov





Identifier: NCT03853109


AK112

Akesobio Australia Pty
ClinicalTrials.gov




Ltd
Identifier: NCT04047290


CS1003

CStone Pharma
Li et al., Acta






Pharmacologica Sinica 42:






142-148 (2021)


MEDI0680
AMP-514
Astra Zeneca,
WO 2012/145493, WO




Amplimmune,
2014/194293




Medimmune


RO7121661

Roche
ClinicalTrials.gov





Identifier: NCT03708328


F520

Shandong New Time
ClinicalTrials.gov




Pharmaceutical Co.
Identifier: NCT03657381


sasanlimab
PF-06801591, RN-888
Pfizer
Cho et al., Annals of






Oncology 30(5) (2019)



BI 754091
BI754091
Boehringer Ingelheim
Kang et al., J. Clin.






Oncology 38(15) (2020)



cetrelimab
JNJ-63723283
Janssen Biotech
Rutkowski et al., J. Clin.






Oncology 37: 8 (2019)



HerinCAR-PD-1

Ningbo Cancer
ClinicalTrials.gov




Hospital
Identifier: NCT02873390


HX008

Taizhou Hanzhong Bio
Zhang et al, mAbs 12(1)





(2020); ClinicalTrials.gov





Identifier: NCT03704246


zimberelimab
WBP3055, GLS-010,
Arcus, Guangzhou
US 2019/0270815, Si-



AB122
Gloria Bio, Harbin
Yang Liu et al., J.




Gloria Pharma, WuXi

Hematol. Oncol. 10: 136





Biologies
(2017)


retifanlimab
MGA012,
Incyte, MacroGenics
WO 2017/19846



INCMGA00012


balstilimab
AGEN2034, AGEN-
Agenus, Ludwig Inst.,
WO 2017/040790



2034
Sloan-Kettering


pidilizumab
CT-011, hBat-1,
CureTech, Medivation,
Rosenblatt et al., J



MDV9300
Teva

Immunother. 34(5): 409-






418 (2011)


teripalimab

Henan Cancer Hospital
ClinicalTrials.gov





Identifier: NCT03985670


CBT-501
GB226, GB 226,
CBT Pharmaceuticals,
ClinicalTrials.gov



Genolimzumab,
Genor
Identifier:



Genormab

NCT03053466.


BAT1306

Bio-Thera Solutions
Wu et al., J. Clin. Oncol.





37(4)(2019)


tislelizumab
BGB-A317
BeiGene, Celgene
WO 2015/35606,





US 2015/0079109


AK105

Akeso, HanX Bio
ClinicalTrials.gov





Identifier: NCT03866967


spartalizumab
PDR001, BAP049
Dana-Farber, Novartis
WO 2015/112900; Lin et





al., Ann. of Oncology, 29: 8





(2018)


prolgolimab
BCD-100
Biocad
Kaplon et





al., mAbs 10(2): 183-203





(2018)


serplulimab
HLX10
Henlix Biotech
ClinicalTrials.gov





Identifier: NCT04297995


dostarlimab
ANB011, TSR-042,
AnaptysBio, Tesaro
WO 2014/179664



ABT1, WBP-285


camrelizumab
SHR-1210
Incyte, Jiangsu
WO 2015/085847; Si-Yang




Hengrui, Shanghai
Liu et al., J. Hematol.




Hengrui
Oncol. 10: 136(2017)


IBI319
IBI-319
Innovent Biologies
ClinicalTrials.gov




(Suzhou) Co. Ltd.,
Identifier: NCT04708210




Lilly


KY1043

Kymab
Van Krinks, Kymab poster





no. P625, “KY1043, a





novel PD-L1 IL-2





immunocytokine directed





towards CD25, delivers





potent anti-tumour activity





in vitro and in vivo”


STI-1110

Sorrento Therapeutics
WO 2014/194302


CA05100948
Antibody 948
UCB Biopharma
U.S. Pat. No. 8.993,731


Nb97
MY2935, MY2626
Fudan University,
Xian et al., Biochem. &




Novamab

Biophys. Res. Comm's






519(3), 267-273 (2019)


ENUM 388D4

Enumeral
Scheuplein et al.,






Immunology, Abstract






4871 (2016)


hAb-10D3
hAb-10D3
BPS Bioscience
U.S. Pat. No. 10,759,859


Unknown

Isis Innovation
WO 2010/029434


ANB030

AnaptysBio Inc.
Grebinoski et al., Current






Opinion in Immunol. 67: 1-






9 (2020)


MCLA-134

Merus N.V.
WO 2019/009727


hAb21

Suzhou Stainwei
U.S. 2020/0277376




Biotech Inc.




Ampsource
U.S. 2019/0367617




Reyoung(Suzhou)
U.S. 2019/0071501




Biology Science &




Technology Co.




Xencor, Inc.
WO 2018/071918




Crown Bio science Inc.
WO 2016/014688




Beijing Hanmi
U.S. 2020/0299412




Beijing Hanmi
U.S. 2021/0032343




Beijing Hanmi
U.S. 2019/0367615




MabQuest
WO 2016/020856




Fuso Pharma,
WO 2018/034226




Hokkaido U.




Eureka Therapeutics,
WO 2016/210129




Inc., Sloan-Kettering




Sutro Biopharma, Inc.
WO 2016/077397




Y-Biologics
U.S. 2019/0248900




Harbour Biomed Ltd.
WO 2017/016497




Shanghai
Liu et al., Sci. Rep 9(1)




PharmaExplorer Co.
(2019)




Bio X Cell
Grasselly et al., Front






Immunol 9: 2100 (2018)





Tongji University
Cai et al., Invest. New






Drugs 37(5): 799-809






(2019)




Janssen Biotech
U.S. 2018/0355061




Shanghai Henlius
U.S. 2019/0218295




Biotech, Inc.




Ultrahuman Nine Ltd.
WO 2019/170898




CytomX
WO 2017/011580




Nanjing Legend
WO 2017/133633




Biotech




Aduro Biotech
U.S. Pat. No. 10,494,436




Sutro Biopharm
U.S. Pat. No. 10,822,414




Versitech Ltd.
U.S. Pat. No. 10,047,137




STCube & Co.,
WO 2016/160792




University of Texas




Tayu Huaxia Biotech
U.S. 2019/0144543




Medivcal Group Co.




Hangzhou Sumgen
U.S. 2019/0016799




Biotechnology




Jiangsu Hengrui
WO 2015/085847




Medicine Co, Shanghai




Hengrui




Pharmaceutical Co




Salubris (Chengdu)
WO 2019/201169




Biotech Co.




Kadmon Corp.
U.S. Pat. No. 10,407,502




Augusta University
WO 2019/051164




Research Institute




Abbvie Biotherapeutics
WO 2018/053106




Inc.




Lyvgen Biopharma
WO 2017/087599




Holdings Ltd.




Crescendo Biologies
WO 2018/127711




Ltd.




Zhejiang Teruisi
U.S. 2019/0322747




Pharmaceutics Inc.




Biosion Inc.
WO 2019/204132




Celgene Corp.
U.S. 2017/0088618




Asia Biotech Pte. Ltd.
WO 2017/058115




The Brigham and
U.S. Pat. No. 10,934,352




Women's Hospital




Singapore Immunology
Seidel et al., Front. Oncol.




Network (SIgN),
28 (2018)




Institute of Medical




Biology, Agency for




Science, Technology




and Research




(A*STAR)




Chinese People
Sun et al., J Gastrointest




Liberation Army

Oncol. 11(6): 1421-1430





General Hospital
(2020)




Immunomedics Inc.
U.S. Pat. No. 10,669,338




Xiangtan Tenghua Bio
WO 2018/162944




Beijing Dongfang
U.S. 2019/0144541




Biotech




REMD Biotherapeutics
WO 2018/119474




Rinat Neuroscience
U.S. Pat. No. 10,155,037




Corp.




Medimmune, Wyeth
WO 2004/056875




Fudan University
Yuan et al., Invest. New






Drugs 39(1): 34-51 (2021)










XI. Gene Switch

Provided herein are gene switch polypeptides, polynucleotides encoding ligand-inducible gene switch polypeptides, and methods and systems incorporating these polypeptides and/or polynucleotides. The term “gene switch” refers to the combination of a response element associated with a promoter, and for instance, an ecdysone receptor (EcR) based system which, in the presence of one or more ligands, modulates the expression of a gene into which the response element and promoter are incorporated. Tightly regulated inducible gene expression systems or gene switches are useful for various applications such as gene therapy, large scale production of proteins in cells, cell based high throughput screening assays, functional genomics and regulation of traits in transgenic plants and animals. Such inducible gene expression systems can include ligand inducible heterologous gene expression systems.


In some embodiments, the polynucleotide or an additional polynucleotide encodes polypeptides for a gene switch system for ligand-inducible control of heterologous gene expression, wherein the gene switch polypeptides include: (a) a first gene switch polypeptide that comprises a DNA binding domain fused to a first nuclear receptor ligand binding domain; and (b) a second gene switch polypeptide that comprises a transactivation domain fused to a second nuclear receptor ligand binding domain; wherein the first gene switch polypeptide and the second gene switch polypeptide are connected by a linker.


In some embodiments, the gene switch system comprises: (a) a first gene switch polypeptide comprising a transactivation domain; (b) a second gene switch polypeptide comprising a DNA binding domain fused to a ligand binding domain; and (c) at least one heterologous polypeptide; wherein one of said first gene switch polypeptide, said second gene switch polypeptide and said heterologous polypeptide is connected by a linker to another one of said first gene switch polypeptide, said second gene switch polypeptide and said heterologous polypeptide, and wherein said polypeptide linker comprises a cleavable linker or ribosome skipping linker sequence.


The heterologous polypeptide may, for example, be a chimeric receptor (e.g., a CAR), a cell tag, or a cytokine as described herein.


In certain embodiments, the gene switch system is of the type described in WO 2018/132494.


In some embodiments, the DNA binding domain comprises at least one of GAL4 (GAL4 DBD), a LexA DBD, a transcription factor DBD, a steroid/thyroid hormone nuclear receptor superfamily member DBD, a bacterial LacZ DBD, and a yeast DBD. In other embodiments, the DNA binding domain comprises the amino acid sequence of SEQ ID NO: 638 or a functional fragment or variant thereof. In some embodiments, the transactivation domain comprises at least one of a VP16 transactivation domain and a B42 acidic activator transactivation domain. In other cases, the transactivation domain comprises an amino acid sequence as shown in SEQ ID NO: 632 or a functional fragment or variant thereof. In some embodiments, at least one of the first nuclear receptor ligand binding domain and the second nuclear receptor ligand binding domain comprises at least one of an ecdysone receptor (EcR), a ubiquitous receptor, an orphan receptor 1, a NER-1, a steroid hormone nuclear receptor 1, a retinoid X receptor interacting protein-15, a liver X receptor (3, a steroid hormone receptor like protein, a liver X receptor, a liver X receptor α, a farnesoid X receptor, a receptor interacting protein 14, and a farnesol receptor, or a functional fragment or variant thereof. In some embodiments, at least one of the first nuclear receptor ligand binding domain, the second nuclear receptor ligand binding domain, and the ligand binding domain is derived from the Ecdysone Receptor polypeptide sequence of SEQ ID NOs: 640 or 642 or a functional fragment or variant thereof. In some embodiments, the nuclear receptor ligand binding domain is a RxR domain of SEQ ID NO: 634 or a functional fragment or variant thereof.


In some embodiments, the polynucleotide encodes at least one of GAL4 (GAL4 DBD), a LexA DBD, a transcription factor DBD, a steroid/thyroid hormone nuclear receptor superfamily member DBD, a bacterial LacZ DBD, and a yeast DBD, or a functional fragment or variant thereof. In other embodiments, the DNA binding domain is encoded by a nucleotide sequence as shown in SEQ ID NO: 639 or a functional fragment or variant thereof. In some embodiments, the polynucleotide encodes at least one of a VP16 transactivation domain and a B42 acidic activator transactivation domain. In other cases, the transactivation domain is encoded by a nucleotide sequence as shown in SEQ ID NO: 633 or a functional fragment or variant thereof. In some embodiments, at least one of the first nuclear receptor ligand binding domain, the second nuclear receptor ligand binding domain, and the ligand binding domain is encoded by SEQ ID NO: 641 or 643 or a functional fragment or variant thereof. In some embodiments, at least one of the first nuclear receptor ligand binding domain, the second nuclear receptor ligand binding domain, and the ligand binding domain is encoded by SEQ ID NO: 635 or a functional fragment or variant thereof.


In yet another embodiment, the first gene switch polypeptide comprises a GAL4 DBD, or a functional fragment or variant thereof, fused to an EcR nuclear receptor ligand binding domain, or a functional fragment or variant thereof, and the second gene switch polypeptide comprises a VP16 transactivation domain, or a functional fragment or variant thereof, fused to a retinoid receptor X (RXR) nuclear receptor ligand binding domain, or a functional fragment or variant thereof. In some cases, the first gene switch polypeptide and the second gene switch polypeptide are connected by a linker, which is selected from the group consisting of 2A, GSG-2A, GSG linker (SEQ ID NO: 531), SGSG linker (SEQ ID NO: 533), furinlink variants and derivatives thereof.


In some cases, at least one of the first nuclear receptor ligand binding domain and the second nuclear receptor ligand binding domain comprise any one of amino acid sequences as shown in SEQ ID NOs: 640 or 642 or a functional fragment or variant thereof. In some embodiments, the first gene switch polypeptide comprises a GAL4 DBD, or a functional fragment or variant thereof, fused to an EcR nuclear receptor ligand binding domain, or a functional fragment or variant thereof, and the second gene switch polypeptide comprises a VP16 transactivation domain, or a functional fragment or variant thereof, fused to a retinoid receptor X (RXR) nuclear receptor ligand binding domain, or a functional fragment or variant thereof. In some embodiments, the Gal4 DBD, or a functional fragment or variant thereof, fused to the EcR nuclear receptor ligand binding domain, or a functional fragment or variant thereof, comprises an amino acid sequence as shown in SEQ ID NOs: 644 or 646, or a functional fragment or variant thereof, and the VP16 transactivation domain, or a functional fragment or variant thereof, fused to the retinoid receptor X (RXR) nuclear receptor ligand binding domain, or a functional fragment or variant thereof, comprises an amino acid sequence as shown in SEQ ID NO: 636, or a functional fragment or variant thereof.


In some cases, at least one of the first nuclear receptor ligand binding domain and the second nuclear receptor ligand binding domain are encoded by the amino acid sequences as shown in SEQ ID NOs: 641 or 643, or a functional fragment or variant thereof. In some embodiments, the Gal4 DBD, or a functional fragment or variant thereof, fused to the EcR nuclear receptor ligand binding domain, or a functional fragment or variant thereof, is encoded by the nucleotide sequence as shown in SEQ ID NOs: 645 or 647, or a functional fragment or variant thereof, and the VP16 transactivation domain, or a functional fragment or variant thereof, fused to the retinoid receptor X (RXR) nuclear receptor ligand binding domain, or a functional fragment or variant thereof, is encoded by the nucleotide sequence of SEQ ID NO: 637, or a functional fragment or variant thereof.


In any of the foregoing gene switch embodiments, the linker can be a cleavable linker, a ribosome skipping linker sequence or an IRES linker. In some cases, the linker is an IRES linker and is encoded by a nucleic acid comprising the sequence of SEQ ID NOs: 702 or 703 or a functional fragment or variant thereof. In other cases, the linker is a cleavable linker or ribosome skipping linker sequence. In some embodiments, the cleavable linker or the ribosome skipping linker sequence comprises one or more of a 2A linker, p2A linker, T2A linker, F2A linker, E2A linker, GSG-2A linker, GSG linker, SGSG linker, furinlink linker variants and derivatives thereof. In other embodiments, the cleavable linker or said ribosome skipping linker sequence has a sequence as shown in any one of SEQ ID NOs: 527, 529, 531, 533, 535, 537, 539, 541, 543, 545, 547, 549, 551, 553, 555, 557 and 559 or is encoded by the sequence as shown in any one of SEQ ID NOs: 528, 530, 532, 534, 536, 538, 540, 542, 544, 546, 548, 550, 552, 554, 556, 558, and 560.


In any of the gene switch embodiments, expression of at least one of the first gene switch polypeptide, the second gene switch polypeptide, the antigen-binding polypeptide, and the heterologous polypeptide of any of the compositions as provided herein can be modulated by a promoter, where the promoter is a tissue-specific promoter or an EF1A promoter or functional fragment or variant thereof. In some cases, the promoter comprises the sequence of SEQ ID NOs: 58 and 59 of WO 2018/132494, or a functional fragment or variant thereof. In other cases, the promoter is a tissue-specific promoter comprising a T-cell-specific response element. In another case, the tissue-specific promoter comprises one or more NFAT response element(s). In yet another case, the NFAT response element has a sequence of any one of SEQ ID NOs: 51 to 57 of WO 2018/132494 or a functional fragment or variant thereof.


In an embodiment, expression of the at least one heterologous polypeptide is modified by an inducible promoter. In some embodiments, the inducible promoter has a sequence of any one of SEQ ID NOs: 40 to 64 of WO 2018/132494 or a functional fragment or variant thereof. In other embodiments, the inducible promoter is modulated by at least one of the first gene switch polypeptide and the second gene switch polypeptide.


It should be understood that any of the foregoing polynucleotides can be included in a vector as described herein.


Also provided herein is a method of regulating the expression of a heterologous gene in an effector cell, the method comprising: (a) introducing into the effector cell one or more polynucleotides encoding the polypeptides of the first and second gene switch polypeptides as described herein and the heterologous polypeptide; and (b) contacting the effector cell with a ligand in an amount sufficient to induce expression of the gene encoding the heterologous polypeptide.


In certain embodiments, the ligand in the method of regulating the expression of the heterologous gene in the effector cell as provided herein comprises at least one of: (2S,3R,5R,9R,10R,13R,14S,17R)-17-[(2S,3R)-3,6-dihydroxy-6-methylheptan-2-yl]-2,3,14-trihydroxy-10,13-dimethyl-2,3,4,5,9,11,12,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-6-one; N′-(3,5-Dimethylbenzoyl)-N′-[(3R)-2,2-dimethyl-3-hexanyl]-2-ethyl-3-methoxybenzohydrazide; 5-Methyl-2,3-dihydro-benzo[1,4] dioxine-6-carboxylic acid N′-(3,5-dimethyl-benzoyl)-N′-(1-ethyl-2,2-dimethyl-propyl)-hydrazide; 5-Methyl-2,3-dihydro-benzo [1,4] dioxine-6-carboxylic acid N′-(3,5-dimethoxy-4-methyl-benzoyl)-N′-(1-ethyl-2,2-dimethyl-propyl)-hydrazide; 5-Methyl-2,3-dihydro-benzo[1,4]dioxine-6-carboxylic acid N′-(1-tert-butyl-butyl)-N′-(3,5-dimethyl-benzoyl)-hydrazide; 5-Methyl-2,3-dihydro-benzo[1,4]dioxine-6-carboxylic acid N′-(1-tert-butyl-butyl)-N′-(3,5-dimethoxy-4-methyl-benzoyl)-hydrazide; 5-Ethyl-2,3-dihydro-benzo[1,4]dioxine-6-carboxylic acid N′-(3,5-dimethyl-benzoyl)-N′-(1-ethyl-2,2-dimethyl-propyl)-hydrazide; 5-Ethyl-2,3-dihydro-benzo[1,4]dioxine-6-carboxylic acid N′-(3,5-dimethoxy-4-methyl-benzoyl)-N ‘-(1-ethyl-2,2-dimethyl-propyl)-hydrazide; 5-Ethyl-2,3-dihydro-benzo[1,4]dioxine-6-carboxylic acid N’-(1-tert-butyl-butyl)-N ‘-(3,5-dimethyl-benzoyl)-hydrazide; 5-Ethyl-2,3-dihydro-benzo[1,4]dioxine-6-carboxylic acid N’-(1-tert-butyl-butyl)-N ‘-(3,5-dimethoxy-4-methyl-benzoyl)-hydrazide; 3,5-Dimethyl-benzoic acid N-(1-ethyl-2,2-dimethyl-propyl)-N’-(3-methoxy-2-methyl-benzoyl)-hydrazide; 3,5-Dimethoxy-4-methyl-benzoic acid N-(1-ethyl-2,2-dimethyl-propyl)-N′-(3-methoxy-2-methyl-benzoyl)-hydrazide; 3,5-Dimethyl-benzoic acid N-(1-tert-butyl-butyl)-N′-(3-methoxy-2-methyl-benzoyl)-hydrazide; 3,5-Dimethoxy-4-methyl-benzoic acid N-(1-tert-butyl-butyl)-N′-(3-methoxy-2-methyl-benzoyl)-hydrazide; 3,5-Dimethyl-benzoic acid N-(1-ethyl-2,2-dimethyl-propyl)-N′-(2-ethyl-3-methoxy-benzoyl)-hydrazide; 3,5-Dimethoxy-4-methyl-benzoic acid N-(1-ethyl-2,2-dimethyl-propyl)-N′-(2-ethyl-3-methoxy-benzoyl)-hydrazide; 3,5-Dimethyl-benzoic acid N-(1-tert-butyl-butyl)-N′-(2-ethyl-3-methoxy-benzoyl)-hydrazide; 3,5-Dimethoxy-4-methyl-benzoic acid N-(1-tert-butyl-butyl)-N′-(2-ethyl-3-methoxy-benzoyl)-hydrazide; 2-Methoxy-nicotinic acid N-(1-tert-butyl-pentyl)-N′-(4-ethyl-benzoyl)-hydrazide; 3,5-Dimethyl-benzoic acid N-(2,2-dimethyl-1-phenyl-propyl)-N′-(4-ethyl-benzoyl)-hydrazide; 3,5-Dimethyl-benzoic acid N-(1-tert-butyl-pentyl)-N′-(3-methoxy-2-methyl-benzoyl)-hydrazide; and 3,5-Dimethoxy-4-methyl-benzoic acid N-(1-tert-butyl-pentyl)-N′-(3-methoxy-2-methyl-benzoyl)-hydrazide.


In some cases, the expression of the gene encoding the polypeptide in the effector cell as provided herein is reduced or eliminated in the absence of the ligand, as compared to the expression in the presence of the ligand. In certain cases, the expression of said heterologous polypeptide is restored by providing additional amounts of the ligand.


In some embodiments, the one or more expression cassettes of the gene switch system further comprise one or more of the following: (a) one or more recombinase attachment sites; and (b) a sequence encoding a serine recombinase. In other embodiments, the one or more expression cassettes further comprise one or more of the following: (a) a non-inducible promoter; and (b) an inducible promoter.


In some embodiments, one of the first and second gene switch polypeptides can be connected to the heterologous polypeptide by a linker.


In some embodiments, the first and second gene switch polypeptides are connected by a polypeptide linker that is an IRES linker.


In some embodiments, the expression cassette can further comprise a second gene encoding a second heterologous polypeptide.


In some embodiments, the gene switch system as provided is for integrating a heterologous gene in a host cell, wherein upon contacting the host cell with the one or more expression cassettes in the presence of the serine recombinase and the one or more recombinase attachment sites, the heterologous gene is integrated in the host cell. In certain embodiments, the gene switch system further comprises a ligand, wherein the heterologous gene is expressed in the host cell upon contact of the host cell by the ligand. In certain embodiments, the one or more recombinase attachment sites can comprise a phage genomic recombination attachment site (attP) or a bacterial genomic recombination attachment site (attB). In some cases, the serine recombinase can be SF370.


In some cases, the expression cassette has the sequence of any one of SEQ ID NOs: 131 to 126 of WO 2018/132494 or a functional fragment or variant thereof.


In certain embodiments, the inducible promoter of the gene switch system can be activated by the transactivation domain. It should be understood that the gene switch system can be included in a single vector or in multiple vectors.


An early version of EcR-based gene switch used Drosophila melanogaster EcR (DmEcR) and Mus musculus RXR (MmRXR) polypeptides and showed that these receptors in the presence of steroid, ponasteroneA, transactivate reporter genes in mammalian cell lines and transgenic mice (Christopherson et al., 1992; No et al., 1996). Later, Suhr et al., 1998 showed that non-steroidal ecdysone agonist, tebufenozide, induced high level of transactivation of reporter genes in mammalian cells through Bombyx mori EcR (BmEcR) in the absence of exogenous heterodimer partner.


International Patent Applications No. PCT/US97/05330 (WO 97/38117) and PCT/US99/08381 (WO99/58155) disclose methods for modulating the expression of an exogenous gene in which a DNA construct comprising the exogenous gene and an ecdysone response element is activated by a second DNA construct comprising an ecdysone receptor that, in the presence of a ligand therefor, and optionally in the presence of a receptor capable of acting as a silent partner, binds to the ecdysone response element to induce gene expression. In this example, the ecdysone receptor was isolated from Drosophila melanogaster. Typically, such systems require the presence of the silent partner, preferably retinoid X receptor (RXR), in order to provide optimum activation. In mammalian cells, insect ecdysone receptor (EcR) is capable of heterodimerizing with mammalian retinoid X receptor (RXR) and, thereby, be used to regulate expression of target genes or heterologous genes in a ligand dependent manner. International Patent Application No. PCT/US98/14215 (WO 99/02683) discloses that the ecdysone receptor isolated from the silk moth Bombyx mori is functional in mammalian systems without the need for an exogenous dimer partner.


U.S. Pat. No. 6,265,173 discloses that various members of the steroid/thyroid superfamily of receptors can combine with Drosophila melanogaster ultraspiracle receptor (USP) or fragments thereof comprising at least the dimerization domain of USP for use in a gene expression system. U.S. Pat. No. 5,880,333 discloses a Drosophila melanogaster EcR and ultraspiracle (USP) heterodimer system used in plants in which the transactivation domain and the DNA binding domain are positioned on two different hybrid proteins. In each of these cases, the transactivation domain and the DNA binding domain (either as native EcR as in International Patent Application No. PCT/US98/14215 or as modified EcR as in International Patent Application No. PCT/US97/05330) were incorporated into a single molecule and the other heterodimeric partners, either USP or RXR, were used in their native state.


International Patent Application No. PCT/US01/0905 discloses an ecdysone receptor-based inducible gene expression system in which the transactivation and DNA binding domains are separated from each other by placing them on two different proteins results in greatly reduced background activity in the absence of a ligand and significantly increased activity over background in the presence of a ligand. This two-hybrid system is a significantly improved inducible gene expression modulation system compared to the two systems disclosed in applications PCT/US97/05330 and PCT/US98/14215. The two-hybrid system is believed to exploit the ability of a pair of interacting proteins to bring the transcription activation domain into a more favorable position relative to the DNA binding domain such that when the DNA binding domain binds to the DNA binding site on the gene, the transactivation domain more effectively activates the promoter (see, for example, U.S. Pat. No. 5,283,173). The two-hybrid gene expression system comprises two gene expression cassettes; the first encoding a DNA binding domain fused to a nuclear receptor polypeptide, and the second encoding a transactivation domain fused to a different nuclear receptor polypeptide. In the presence of ligand, it is believed that a conformational change is induced which promotes interaction of the antibody with the TGF-β cytokine trap, thereby resulting in dimerization of the DNA binding domain and the transactivation domain. Since the DNA binding and transactivation domains reside on two different molecules, the background activity in the absence of ligand is greatly reduced.


Certain modifications of the two-hybrid system could also provide improved sensitivity to non-steroidal ligands for example, diacylhydrazines, when compared to steroidal ligands for example, ponasterone A (“PonA”) or muristerone A (“MurA”). That is, when compared to steroids, the non-steroidal ligands provided higher gene transcription activity at a lower ligand concentration. Furthermore, the two-hybrid system avoids some side effects due to overexpression of RXR that can occur when unmodified RXR is used as a switching partner. In a preferred two-hybrid system, native DNA binding and transactivation domains of EcR or RXR are eliminated and as a result, these hybrid molecules have less chance of interacting with other steroid hormone receptors present in the cell, thereby resulting in reduced side effects.


The ecdysone receptor (EcR) is a member of the nuclear receptor superfamily and is classified into subfamily 1, group H (referred to herein as “Group H nuclear receptors”). The members of each group share 40-60% amino acid identity in the E (ligand binding) domain (Laudet et al., A Unified Nomenclature System for the Nuclear Receptor Subfamily, 1999; Cell 97: 161-163). In addition to the ecdysone receptor, other members of this nuclear receptor subfamily 1, group H include: ubiquitous receptor (UR), Orphan receptor 1 (OR-1), steroid hormone nuclear receptor 1 (NER-1), RXR interacting protein-15 (RIP-15), liver x receptor β (LXRβ), steroid hormone receptor like protein (RLD-1), liver x receptor (LXR), liver x receptor α (LXRα), farnesoid x receptor (FXR), receptor interacting protein 14 (RIP-14), and farnesol receptor (HRR-1).


In some cases, an inducible promoter (“IP”) can be a small molecule ligand-inducible two polypeptide ecdysone receptor-based gene switch, such as Intrexon Corporation's RHEOSWITCH® gene switch. In some cases, a gene switch can be selected from ecdysone-based receptor components as described in, but without limitation to, any of the systems described in: PCT/US2001/009050 (WO 2001/070816); U.S. Pat. Nos. 7,091,038; 7,776,587; 7,807,417; 8,202,718; PCT/US2001/030608 (WO 2002/029075); U.S. Pat. Nos. 8,105,825; 8,168,426; PCT/US52002/005235 (WO 2002/066613); U.S. application Ser. No. 10/468,200 (U.S. Pub. No. 20120167239); PCT/US2002/005706 (WO 2002/066614); U.S. Pat. Nos. 7,531,326; 8,236,556; 8,598,409; PCT/US2002/005090 (WO 2002/066612); U.S. Pat. No. 8,715,959 (U.S. Pub. No. 20060100416); PCT/US2002/005234 (WO 2003/027266); U.S. Pat. Nos. 7,601,508; 7,829,676; 7,919,269; 8,030,067; PCT/US2002/005708 (WO 2002/066615); U.S. application Ser. No. 10/468,192 (U.S. Pub. No. 20110212528); PCT/US2002/005026 (WO 2003/027289); U.S. Pat. Nos. 7,563,879; 8,021,878; 8,497,093; PCT/US2005/015089 (WO 2005/108617); U.S. Pat. Nos. 7,935,510; 8,076,454; PCT/US2008/011270 (WO 2009/045370); U.S. application Ser. No. 12/241,018 (U.S. Pub. No. 20090136465); PCT/US2008/011563 (WO 2009/048560); U.S. application Ser. No. 12/247,738 (U.S. Pub. No. 20090123441); PCT/US2009/005510 (WO 2010/042189); U.S. application Ser. No. 13/123,129 (U.S. Pub. No. 20110268766); PCT/US2011/029682 (WO 2011/119773); U.S. application Ser. No. 13/636,473 (U.S. Pub. No. 20130195800); PCT/US2012/027515 (WO 2012/122025); WO 2018/132494 (PCT/US2018/013196); and, U.S. Pat. No. 9,402,919.


As used herein, the term “ligand,” as applied to ligand-activated ecdysone receptor-based gene switches are small molecules of varying solubility (such as diacylhydrazine compounds) having the capability of activating a gene switch to stimulate gene expression (i.e., therein providing ligand inducible expression of polynucleotides (e.g., mRNAs, miRNAs, etc) and/or polypeptides). Examples of such ligands include, but are not limited to those described in: WO 2004/072254 (PCT/US2004/003775); WO 2004/005478 (PCT/US2003/021149); WO 2005/017126 (PCT/US2004/005149); WO 2004/078924 (PCT/US2004/005912); WO 2008/153801 (PCT/US2008/006757); WO 2009/114201 (PCT/US2009/001639); WO 2013/036758 (PCT/US2012/054141); WO 2014/144380 (PCT/US2014/028768); and, WO 2016/044390 (PCT/US2015/050375).


Examples of ligands also include, without limitation, an ecdysteroid, such as ecdysone, 20-hydroxyecdysone, ponasterone A, muristerone A, and the like, 9-cis-retinoic acid, synthetic analogs of retinoic acid, N, N′-diacylhydrazines such as those disclosed in U.S. Pat. Nos. 6,258,603; 6,013,836; 5,117,057; 5,530,028; 5,378,726; 7,304,161; 7,851,220; 8,748,125; 9,272,986; 7,456,315; 7,563,928; 8,524,948; 9,102,648; 9,169,210; 9,255,273; and, 9,359,289; oxadiazolines as described in U.S. Pat. Nos. 8,669,072; and, 8,895,306; dibenzoylalkyl cyanohydrazines such as those disclosed in European Application No. 2,461,809; N-alkyl-N,N′-diaroylhydrazines such as those disclosed in U.S. Pat. No. 5,225,443; N-acyl-N-alkylcarbonylhydrazines such as those disclosed in European Application No. 234,994; N-aroyl-N-alkyl-N′-aroylhydrazines such as those described in U.S. Pat. No. 4,985,461; amidoketones such as those described in U.S. Pat. Nos. 7,375,093; 8,129,355; and, 9,802,936; and other similar materials including 3,5-di-tert-butyl-4-hydroxy-N-isobutyl-benzamide, 8-O-acetylharpagide, oxysterols, 22(R) hydroxycholesterol, 24(S) hydroxycholesterol, 25-epoxycholesterol, T0901317, 5-alpha-6-alpha-epoxycholesterol-3-sulfate (ECHS), 7-ketocholesterol-3-sulfate, famesol, bile acids, 1,1-biphosphonate esters, juvenile hormone III, and the like. Examples of diacylhydrazine ligands useful in the present invention include RG-115819 (3,5-Dimethyl-benzoic acid N-(1-ethyl-2,2-dimethyl-propyl)-N′-(2-methyl-3-methoxybenzoyl)-hydrazide), RG-115932 ((R)-3,5-Dimethyl-benzoic acid N-(1-tert-butyl-butyl)N′-(2-ethyl-3-methoxy-benzoyl)-hydrazide), and RG-115830 (3,5-Dimethyl-benzoic acid N-(1-tert-butyl-butyl)-N′-(2-ethyl-3-methoxy-benzoyl)-hydrazide). See, e.g., WO 2008/153801 (PCT/US2008/006757); and, WO 2013/036758 (PCT/US2012/054141).


For example, a ligand for the ecdysone receptor-based gene switch may be selected from any suitable ligands. Both naturally occurring ecdysone or ecdysone analogs (e.g., 20-hydroxyecdysone, muristerone A, ponasterone A, ponasterone B, ponasterone C, 26-iodoponasterone A, inokosterone or 26-mesylinokosterone) and non-steroid inducers may be used as a ligand for gene switch of the present invention. U.S. Pat. No. 6,379,945, describes an insect steroid receptor isolated from Heliothis virescens (“HEcR”) which is capable of acting as a gene switch responsive to both steroid and certain non-steroidal inducers. Non-steroidal inducers have a distinct advantage over steroids, in this and many other systems which are responsive to both steroids and non-steroid inducers, for several reasons including, for example: lower manufacturing cost, metabolic stability, absence from insects, plants, or mammals, and environmental acceptability. U.S. Pat. No. 6,379,945 describes the utility of two dibenzoylhydrazines, 1,2-dibenzoyl-1-tert-butyl-hydrazine and tebufenozide (N-(4-ethylbenzoyl)-N′-(3,5-dimethylbenzoyl)-N′-tert-butyl-hydrazine) as ligands for an ecdysone-based gene switch. Also included in the present invention as a ligand are other dibenzoylhydrazines, such as those disclosed in U.S. Pat. No. 5,117,057. Use of tebufenozide as a chemical ligand for the ecdysone receptor from Drosophila melanogaster is also disclosed in U.S. Pat. No. 6,147,282. Additional, non-limiting examples of ecdysone ligands are 3,5-di-tert-butyl-4-hydroxy-N-isobutyl-benzamide, 8-O-acetylharpagide, a 1,2-diacyl hydrazine, an N′-substituted-N,N′-disubstituted hydrazine, a dibenzoylalkyl cyanohydrazine, an N-substituted-N-alkyl-N,N-diaroyl hydrazine, an N-substituted-N-acyl-N-alkyl, carbonyl hydrazine or an N-aroyl-N′-alkylN′-aroyl hydrazine. (See U.S. Pat. No. 6,723,531).


In one embodiment, the ligand for an ecdysone-based gene switch system is a diacylhydrazine ligand or chiral diacylhydrazine ligand. The ligand used in the gene switch system may be compounds of Formula I




embedded image


wherein A is alkoxy, arylalkyloxy or aryloxy; B is optionally substituted aryl or optionally substituted heteroaryl; and R1 and R2 are independently optionally substituted alkyl, arylalkyl, hydroxyalkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclo, optionally substituted aryl or optionally substituted heteroaryl; or pharmaceutically acceptable salts, hydrates, crystalline forms or amorphous forms thereof.


In another embodiment, the ligand may be enantiomerically enriched compounds of Formula II




embedded image


wherein A is alkoxy, arylalkyloxy, aryloxy, arylalkyl, optionally substituted aryl or optionally substituted heteroaryl; B is optionally substituted aryl or optionally substituted heteroaryl; and R1 and R2 are independently optionally substituted alkyl, arylalkyl, hydroxyalkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclo, optionally substituted aryl or optionally substituted heteroaryl; with the proviso that R1 does not equal R2; wherein the absolute configuration at the asymmetric carbon atom bearing R1 and R2 is predominantly S; or pharmaceutically acceptable salts, hydrates, crystalline forms or amorphous forms thereof.


In certain embodiments, the ligand may be enantiomerically enriched compounds of Formula III




embedded image


wherein A is alkoxy, arylalkyloxy, aryloxy, arylalkyl, optionally substituted aryl or optionally substituted heteroaryl; B is optionally substituted aryl or optionally substituted heteroaryl; and R1 and R2 are independently optionally substituted alkyl, arylalkyl, hydroxyalkyl, haloalkyl, optionally substituted cycloalkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heterocyclo, optionally substituted aryl or optionally substituted heteroaryl; with the proviso that R1 does not equal R2; wherein the absolute configuration at the asymmetric carbon atom bearing R1 and R2 is predominantly R; or pharmaceutically acceptable salts, hydrates, crystalline forms or amorphous forms thereof.


In one embodiment, a ligand may be (R)-3,5-dimethyl-benzoic acid N-(1-tertbutyl-butyl)-N′-(2-ethyl-3-methoxy-benzoyl)-hydrazide having an enantiomeric excess of at least 95% or a pharmaceutically acceptable salt, hydrate, crystalline form or amorphous form thereof.


The diacylhydrazine ligands of Formula I and chiral diacylhydrazine ligands of Formula II or III, when used with an ecdysone-based gene switch system, provide the means for external temporal regulation of expression of a therapeutic polypeptide or therapeutic polynucleotide of the present invention. See U.S. Pat. Nos. 8,076,517; 8,884,060; and, 9,598,355.


The ligands used in the present invention may form salts. The term “salt(s)” as used herein denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases. In addition, when a compound of Formula I, II or III contains both a basic moiety and an acidic moiety, zwitterions (“inner salts”) may be formed and are included within the term “salt(s)” as used herein. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are used, although other salts are also useful, e.g., in isolation or purification steps which may be employed during preparation. Salts of the compounds of Formula I, II or III may be formed, for example, by reacting a compound with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.


The ligands which contain a basic moiety may form salts with a variety of organic and inorganic acids. Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example, trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides (formed with hydrochloric acid), hydrobromides (formed with hydrogen bromide), hydroiodides, 2-hydroxyethanesulfonates, lactates, maleates (formed with maleic acid), methanesulfonates (formed with methanesulfonic acid), 2-naphthalenesulfonates, nicotinates, nitrates, oxalates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates (such as those formed with sulfuric acid), sulfonates (such as those mentioned herein), tartrates, thiocyanates, toluenesulfonates such as tosylates, undecanoates, and the like.


The ligands which contain an acidic moiety may form salts with a variety of organic and inorganic bases. Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines (formed with N,N-bis(dehydroabietyl)ethylenediamine), N-methyl-D-glucamines, N-methyl-D-glucamides, t-butyl amines, and salts with amino acids such as arginine, lysine and the like.


Non-limiting examples of the ligands for the inducible gene expression system also includes those utilizing the FK506 binding domain are FK506, Cyclosporin A, or Rapamycin. FK506, rapamycin, and their analogs are disclosed in U.S. Pat. Nos. 6,649,595; 6,187,757; 7,276,498; and, 7,273,874.


In some embodiments, a diacylhydrazine ligand for inducible gene expression is administered at unit daily dose of about 5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100 or 120 mg. In some embodiments, the diacylhydrazine ligand is administered at a unit daily dose of about 5 mg. In some embodiments, the diacylhydrazine ligand is administered at a unit daily dose of about 10 mg. In some embodiments, the diacylhydrazine ligand is administered at a unit daily dose of about 15 mg. In some embodiments, the diacylhydrazine ligand is administered daily at a unit daily dose of about 20 mg.


In some embodiments, the cytokine, cell tag and/or CAR can be under the control of an inducible promoter for gene transcription. In one aspect, the inducible promoter can be a gene switch ligand inducible promoter. In some cases, an inducible promoter can be a small molecule ligand-inducible two polypeptide ecdysone receptor-based gene switch, such as RHEOSWITCH® gene switch. In some cases, the gene switch system used may be the one described in WO 2018/132494.


In some embodiments, the cytokines described above can be under the control of an inducible promoter for gene transcription. In one aspect, the inducible promoter can be a gene switch ligand inducible promoter. In some cases, an inducible promoter can be a small molecule ligand-inducible two polypeptide ecdysone receptor-based gene switch, such as RHEOSWITCH® gene switch. In some cases, the gene switch system used may be the one described in WO 2018/132494.


In some embodiments, the modified immune effector cells as described herein can be under the control of an inducible promoter for gene transcription. In one aspect, the inducible promoter can be a gene switch ligand inducible promoter. In some cases, an inducible promoter can be a small molecule ligand-inducible two polypeptide ecdysone receptor-based gene switch, such as RHEOSWITCH® gene switch as described in WO2018/132494.


XII. Modified Immune Effector Cells

Provided herein are modified immune effector cells expressing one or more miRNAs, CARs, cytokines, and/or cell tags as described herein. In some embodiments, modified immune effector cells are modified T cells and/or natural killer (NK) cells. T cells or T lymphocytes are a subtype of white blood cells that are involved in cell-mediated immunity. Exemplary T cells include T helper cells, cytotoxic T cells, TH17 cells, stem memory T cells (TSCM), naïve T cells, memory T cells, effector T cells, regulatory T cells, or natural killer T cells. In certain aspects, the embodiments described herein include making and/or expanding the modified immune effector cells (e.g., T-cells, Tregs, NK-cell or NK T-cells). Such may be accomplished by transfecting the cells with an expression vector containing a DNA (or RNA) construct encoding the one or more miRNAs, CARs, cytokines, and/or cell tags as described herein. It should be understood that the cells of the present disclosure can be human or animal cells.


T helper cells (TH cells) assist other white blood cells in immunologic processes, including maturation of B cells into plasma cells and memory B cells, and activation of cytotoxic T cells and macrophages. In some instances, TH cells are known as CD4+ T cells due to expression of the CD4 glycoprotein on the cell surfaces. T helper cells become activated when they are presented with peptide antigens by MHC class II molecules, which are expressed on the surface of antigen-presenting cells (APCs). Once activated, they divide rapidly and secrete small proteins called cytokines that regulate or assist in the active immune response. These cells can differentiate into one of several subtypes, including TH1, TH2, TH3, TH17, Th9, or TFH, which secrete different cytokines to facilitate different types of immune responses. Signaling from the APC directs T cells into particular subtypes.


Cytotoxic T cells (TC cells or CTLs) destroy virus-infected cells and tumor cells, and are also implicated in transplant rejection. These cells are also known as CD8+ T cells since they express the CD8 glycoprotein on their surfaces. These cells recognize their targets by binding to antigen associated with MHC class I molecules, which are present on the surface of all nucleated cells. Through IL-10, adenosine, and other molecules secreted by regulatory T cells, the CD8+ cells can be inactivated to an anergic state, which prevents autoimmune diseases.


Memory T cells are a subset of antigen-specific T cells that persist long-term after an infection has resolved. They quickly expand to large numbers of effector T cells upon re-exposure to their cognate antigen, thus providing the immune system with “memory” against past infections. Memory T cells comprise subtypes: stem memory T cells (TSCM), central memory T cells (TCM cells) and two types of effector memory T cells (TEM cells and TEMRA cells). Memory cells can be either CD4+ or CD8+. Memory T cells can express the cell surface proteins CD45RO, CD45RA and/or CCR7.


Regulatory T cells (Treg cells), formerly known as suppressor T cells, play a role in the maintenance of immunological tolerance. Their major role is to shut down T cell-mediated immunity toward the end of an immune reaction and to suppress autoreactive T cells that escaped the process of negative selection in the thymus.


Natural killer T cells (NKT cells) bridge the adaptive immune system with the innate immune system. Unlike conventional T cells that recognize peptide antigens presented by major histocompatibility complex (MHC) molecules, NKT cells recognize glycolipid antigen presented by a molecule called CD1d. Once activated, these cells can perform functions ascribed to both Th and Tc cells (i.e., cytokine production and release of cytolytic/cell killing molecules). They are also able to recognize and eliminate some tumor cells and cells infected with herpes viruses.


Natural killer (NK) cells are a type of cytotoxic lymphocyte of the innate immune system. In some instances, NK cells provide a first line defense against viral infections and/or tumor formation. NK cells can detect MHC presented on infected or cancerous cells, triggering cytokine release, and subsequently induce lysis and apoptosis. NK cells can further detect stressed cells in the absence of antibodies and/or MHC, thereby allowing a rapid immune response.


A. Immune Effector Cell Sources


In certain aspects, the embodiments described herein include methods of making and/or expanding the modified (antigen-specific redirected) immune effector cells (e.g., T-cells, Tregs, NK-cell or NK T-cells) that comprise transfecting the cells with an expression vector containing a DNA (or RNA) construct encoding the CAR, then, optionally, stimulating the cells with feeder cells, recombinant antigen, or an antibody to the receptor to cause the cells to proliferate. In certain aspects, the cell (or cell population) engineered to express a CAR is a stem cell, CD34+ cord blood cells, iPS cell, T cell differentiated from iPS cell, immune effector cell or a precursor of these cells.


Sources of immune effector cells can include both allogeneic and autologous sources. In some cases immune effector cells can be differentiated from stem cells or induced pluripotent stem cells (iPSCs). Thus, cells for engineering according to the embodiments can be isolated from umbilical cord blood, peripheral blood, human embryonic stem cells, or iPSCs. For example, allogeneic T cells can be modified to include a chimeric antigen receptor (and optionally, to lack functional TCR). In some aspects, the immune effector cells are primary human T cells such as T cells derived from human peripheral blood mononuclear cells (PBMC). PBMCs can be collected from the peripheral blood or after stimulation with G-CSF (Granulocyte colony stimulating factor) from the bone marrow, or umbilical cord blood. In one aspect, the immune effector cells are Pan T cells. Following transfection or transduction (e.g., with a CAR expression construct), the cells can be immediately infused or can be cryo-preserved. In certain aspects, following transfection or transduction, the cells can be preserved in a cytokine bath that can include IL-2 and/or IL-21 until ready for infusion. In certain aspects, following transfection, the cells can be propagated for days, weeks, or months ex vivo as a bulk population within about 1, 2, 3, 4, 5 days or more following gene transfer into cells. In a further aspect, following transfection, the transfectants are cloned and a clone demonstrating presence of a single integrated or episomally maintained expression cassette or plasmid, and expression of the chimeric antigen receptor is expanded ex vivo. The clone selected for expansion demonstrates the capacity to specifically recognize and lyse antigen-expressing target cells. The recombinant T cells can be expanded by stimulation with IL-2, or other cytokines that bind the common gamma-chain (e.g., IL-7, IL-12, IL-15, IL-21, and others). The recombinant T cells can be expanded by stimulation with artificial antigen presenting cells. The recombinant T cells can be expanded on artificial antigen presenting cell or with an antibody, such as OKT3, which cross links CD3 on the T cell surface. Subsets of the recombinant T cells can be further selected with the use of magnetic bead based isolation methods and/or fluorescence activated cell sorting technology and further cultured with the AaPCs. In a further aspect, the genetically modified cells can be cryopreserved. In some embodiments, the genetically modified cells are not cryopreserved.


T cells can also be obtained from a number of sources, including bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In certain embodiments of the present invention, any number of T cell lines available in the art, can be used. In certain embodiments of the present invention, T cells can be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled artisan, such as Ficoll® separation. In some embodiments, cells from the circulating blood of an individual are obtained by apheresis. The apheresis product typically contains lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets. In one embodiment, the cells collected by apheresis can be washed to remove the plasma fraction and to place the cells in an appropriate buffer or media for subsequent processing steps. In one embodiment of the invention, the cells are washed with phosphate buffered saline (PBS). In an alternative embodiment, the wash solution lacks calcium and can lack magnesium or can lack many if not all divalent cations. Initial activation steps in the absence of calcium lead to magnified activation. As those of ordinary skill in the art would readily appreciate a washing step can be accomplished by methods known to those in the art, such as by using a semi-automated “flow-through” centrifuge (for example, the Cobe 2991 cell processor, the Baxter CytoMate, or the Haemonetics Cell Saver 5) according to the manufacturer+s instructions. After washing, the cells can be resuspended in a variety of biocompatible buffers, such as, for example, Ca2+-free, Mg2+-free PBS, PlasmaLyte A, or other saline solution with or without buffer. Alternatively, the undesirable components of the apheresis sample can be removed and the cells directly resuspended in culture media.


In another embodiment, T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLL® gradient or by counterflow centrifugal elutriation. A specific subpopulation of T cells, such as CD3+, CD28+, CD4+, CD8+, CD45RA+, and CD45RO+ T cells, can be further isolated by positive or negative selection techniques. In another embodiment, CD14+ cells are depleted from the T-cell population. For example, in one embodiment, T cells are isolated by incubation with anti-CD3/anti-CD28 (i.e., 3×28)-conjugated beads, such as DYNABEADS® M-450 CD3/CD28 T, for a time period sufficient for positive selection of the desired T cells. In one embodiment, the time period is about 30 minutes. In a further embodiment, the time period ranges from 30 minutes to 36 hours or longer and all integer values there between. In a further embodiment, the time period is at least 1, 2, 3, 4, 5, or 6 hours. In yet another embodiment, the time period is 10 to 24 hours. In one embodiment, the incubation time period is 24 hours. For isolation of T cells from patients with leukemia, use of longer incubation times, such as 24 hours, can increase cell yield. Longer incubation times can be used to isolate T cells in any situation where there are few T cells as compared to other cell types, such in isolating tumor infiltrating lymphocytes (TIL) from tumor tissue or from immune-compromised individuals. Further, use of longer incubation times can increase the efficiency of capture of CD8+ T cells. Thus, by simply shortening or lengthening the time T cells are allowed to bind to the CD3/CD28 beads and/or by increasing or decreasing the ratio of beads to T cells (as described further herein), subpopulations of T cells can be preferentially selected for or against at culture initiation or at other time points during the process. Additionally, by increasing or decreasing the ratio of anti-CD3 and/or anti-CD28 antibodies on the beads or other surface, subpopulations of T cells can be preferentially selected for or against at culture initiation or at other desired time points. The skilled artisan would recognize that multiple rounds of selection can also be used in the context of this invention. In certain embodiments, it can be desirable to perform the selection procedure and use the “unselected” cells in the activation and expansion process. “Unselected” cells can also be subjected to further rounds of selection.


Enrichment of a T cell population by negative selection can be accomplished with a combination of antibodies directed to surface markers unique to the negatively selected cells. One method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected. For example, to enrich for CD4+ cells by negative selection, a monoclonal antibody cocktail typically includes antibodies to CD14, CD20, CD11b, CD16, HLA-DR, and CD8. In certain embodiments, it can be desirable to enrich for or positively select for regulatory T cells which typically express CD4+, CD25+, CD62Lhi, GITR+, and FoxP3+. Alternatively, in certain embodiments, T regulatory cells are depleted by anti-CD25 conjugated beads or other similar method of selection.


For isolation of a desired population of cells by positive or negative selection, the concentration of cells and surface (e.g., particles such as beads) can be varied. In certain embodiments, it can be desirable to significantly decrease the volume in which beads and cells are mixed together (i.e., increase the concentration of cells), to ensure maximum contact of cells and beads. For example, in one embodiment, a concentration of 2 billion cells/ml is used. In one embodiment, a concentration of 1 billion cells/ml is used. In a further embodiment, greater than 100 million cells/ml is used. In a further embodiment, a concentration of cells of 10, 15, 20, 25, 30, 35, 40, 45, or 50 million cells/ml is used. In yet another embodiment, a concentration of cells from 75, 80, 85, 90, 95, or 100 million cells/ml is used. In further embodiments, concentrations of 125 or 150 million cells/ml can be used. Using high concentrations can result in increased cell yield, cell activation, and cell expansion. Further, use of high cell concentrations allows more efficient capture of cells that can weakly express target antigens of interest, such as CD28-negative T cells, or from samples where there are many tumor cells present (i.e., leukemic blood, tumor tissue, etc.). Such populations of cells can have therapeutic value and would be desirable to obtain. For example, using high concentration of cells allows more efficient selection of CD8+ T cells that normally have weaker CD28 expression.


In a related embodiment, it can be desirable to use lower concentrations of cells. By significantly diluting the mixture of T cells and surface (e.g., particles such as beads), interactions between the particles and cells is minimized. This selects for cells that express high amounts of desired antigens to be bound to the particles. For example, CD4+ T cells express higher levels of CD28 and are more efficiently captured than CD8+ T cells in dilute concentrations. In one embodiment, the concentration of cells used is 5×106/ml. In other embodiments, the concentration used can be from about 1×105/ml to 1×106/ml, and any integer value in between.


In other embodiments, the cells can be incubated on a rotator for varying lengths of time at varying speeds at either 2-10° C. or at room temperature.


T cells for stimulation can also be frozen after a washing step. After the washing step that removes plasma and platelets, the cells can be suspended in a freezing solution. While many freezing solutions and parameters are known in the art and will be useful in this context, one method involves using PBS containing 20% DMSO and 8% human serum albumin, or culture media containing 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin and 7.5% DMSO, or 31.25% Plasmalyte-A, 31.25% Dextrose 5%, 0.45% NaCl, 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin, and 7.5% DMSO or other suitable cell freezing media containing for example, Hespan and PlasmaLyte A, the cells then are frozen to −80° C. at a rate of 1° C. per minute and stored in the vapor phase of a liquid nitrogen storage tank. Other methods of controlled freezing can be used as well as uncontrolled freezing immediately at −20° C. or in liquid nitrogen. In certain embodiments, cryopreserved cells are thawed and washed as described herein and allowed to rest for one hour at room temperature prior to activation using the methods of the present invention.


Also provided in certain embodiments is the collection of blood samples or apheresis product from a subject at a time period prior to when the expanded cells as described herein might be needed. As such, the source of the cells to be expanded can be collected at any time point necessary, and desired cells, such as T cells, isolated and frozen for later use in T cell therapy for any number of diseases or conditions that would benefit from T cell therapy, such as those described herein. In one embodiment a blood sample or an apheresis is taken from a generally healthy subject. In certain embodiments, a blood sample or an apheresis is taken from a generally healthy subject who is at risk of developing a disease, but who has not yet developed a disease, and the cells of interest are isolated and frozen for later use. In certain embodiments, the T cells can be expanded, frozen, and used at a later time. In certain embodiments, samples are collected from a patient shortly after diagnosis of a particular disease as described herein but prior to any treatments. In a further embodiment, the cells are isolated from a blood sample or an apheresis from a subject prior to any number of relevant treatment modalities, including but not limited to treatment with agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxan, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, and irradiation. These drugs inhibit either the calcium dependent phosphatase calcineurin (cyclosporine and FK506) or inhibit the p70S6 kinase that is important for growth factor induced signaling (rapamycin) (Liu et al., Cell 66:807-815, (1991); Henderson et al., Immun 73:316-321, (1991); Bierer et al., Curr. Opin. Immun 5:763-773, (1993)). In a further embodiment, the cells are isolated for a patient and frozen for later use in conjunction with (e.g., before, simultaneously or following) bone marrow or stem cell transplantation, T cell ablative therapy using either chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide, or antibodies such as OKT3 or CAMPATH. In another embodiment, the cells are isolated prior to and can be frozen for later use for treatment following B-cell ablative therapy such as agents that react with CD20, e.g., Rituxan.


In a further embodiment of the present invention, T cells are obtained from a patient directly following treatment. In this regard, it has been observed that following certain cancer treatments, in particular treatments with drugs that damage the immune system, shortly after treatment during the period when patients would normally be recovering from the treatment, the quality of T cells obtained can be optimal or improved for their ability to expand ex vivo. Likewise, following ex vivo manipulation using the methods described herein, these cells can be in a preferred state for enhanced engraftment and in vivo expansion. Thus, it is contemplated within the context of the present invention to collect blood cells, including T cells, dendritic cells, or other cells of the hematopoietic lineage, during this recovery phase. Further, in certain embodiments, mobilization (for example, mobilization with GM-CSF) and conditioning regimens can be used to create a condition in a subject wherein repopulation, recirculation, regeneration, and/or expansion of particular cell types is favored, especially during a defined window of time following therapy. Illustrative cell types include T cells, B cells, dendritic cells, and other cells of the immune system.


B. Activation and Expansion of Immune Effector Cells


Whether prior to or after engineering of the immune effector cells to express a CAR described herein, the cells can be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005.


Generally, the immune effector cells described herein are expanded by contact with a surface having attached thereto an agent that stimulates a CD3/TCR complex associated signal and a ligand that stimulates a co-stimulatory molecule on the surface of the cells. In particular, cell populations can be stimulated as described herein, such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore. For co-stimulation of an accessory molecule on the surface of the cells, a ligand that binds the accessory molecule is used. For example, a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells. To stimulate proliferation of either CD4+ T cells or CD8+ T cells, an anti-CD3 antibody and an anti-CD28 antibody. Examples of an anti-CD28 antibody include 9.3, B-T3, XR-CD28 (Diaclone, Besancon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, (1998); Haanen et al., J. Exp. Med. 190(9):13191328, (1999); Garland et al., J. Immunol Meth. 227(1-2):53-63, (1999)).


In certain embodiments, the primary stimulatory signal and the co-stimulatory signal for the immune effector cell can be provided by different protocols. For example, the agents providing each signal can be in solution or coupled to a surface. When coupled to a surface, the agents can be coupled to the same surface (i.e., in “cis” formation) or to separate surfaces (i.e., in “trans” formation). Alternatively, one agent can be coupled to a surface and the other agent in solution. In one embodiment, the agent providing the co-stimulatory signal is bound to a cell surface and the agent providing the primary activation signal is in solution or coupled to a surface. In certain embodiments, both agents can be in solution. In another embodiment, the agents can be in soluble form, and then cross-linked to a surface, such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents. In this regard, see for example, U.S. Patent Application Publication Nos. 20040101519 and 20060034810 for artificial antigen presenting cells (aAPCs) that are contemplated for use in activating and expanding T cells in the present invention.


In one embodiment, the two agents are immobilized on beads, either on the same bead, i.e., “cis,” or to separate beads, i.e., “trans.” By way of example, the agent providing the primary activation signal is an anti-CD3 antibody or an antigen-binding fragment thereof and the agent providing the co-stimulatory signal is an anti-CD28 antibody or antigen-binding fragment thereof; and both agents are co-immobilized to the same bead in equivalent molecular amounts. In one embodiment, a 1:1 ratio of each antibody bound to the beads for CD4+ T cell expansion and T cell growth is used. In certain aspects of the present invention, a ratio of anti CD3:CD28 antibodies bound to the beads is used such that an increase in T cell expansion is observed as compared to the expansion observed using a ratio of 1:1. In one particular embodiment an increase of from about 1 to about 3 fold is observed as compared to the expansion observed using a ratio of 1:1. In one embodiment, the ratio of CD3:CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between. In one aspect of the present invention, more anti-CD28 antibody is bound to the particles than anti-CD3 antibody, i.e., the ratio of CD3:CD28 is less than one. In certain embodiments of the invention, the ratio of anti CD28 antibody to anti CD3 antibody bound to the beads is greater than 2:1. In one particular embodiment, a 1:100 CD3:CD28 ratio of antibody bound to beads is used. In another embodiment, a 1:75 CD3:CD28 ratio of antibody bound to beads is used. In a further embodiment, a 1:50 CD3:CD28 ratio of antibody bound to beads is used. In another embodiment, a 1:30 CD3:CD28 ratio of antibody bound to beads is used. In some embodiments, a 1:10 CD3:CD28 ratio of antibody bound to beads is used. In another embodiment, a 1:3 CD3:CD28 ratio of antibody bound to the beads is used. In yet another embodiment, a 3:1 CD3:CD28 ratio of antibody bound to the beads is used.


Ratios of particles to cells from 1:500 to 500:1 and any integer values in between can be used to stimulate T cells or other target cells. As those of ordinary skill in the art can readily appreciate, the ratio of particles to cells can depend on particle size relative to the target cell. For example, small sized beads could only bind a few cells, while larger beads could bind many. In certain embodiments the ratio of cells to particles ranges from 1:100 to 100:1 and any integer values in-between and in further embodiments the ratio comprises 1:9 to 9:1 and any integer values in between, can also be used to stimulate T cells. The ratio of anti-CD3- and anti-CD28-coupled particles to T cells that result in T cell stimulation can vary as noted above, however certain values include 1:100, 1:50, 1:40, 1:30, 1:20, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, and 15:1 with one ratio being at least 1:1 particles per T cell. In one embodiment, a ratio of particles to cells of 1:1 or less is used. In one particular embodiment, the particle:cell ratio is 1:5. In further embodiments, the ratio of particles to cells can be varied depending on the day of stimulation. For example, in one embodiment, the ratio of particles to cells is from 1:1 to 10:1 on the first day and additional particles are added to the cells every day or every other day thereafter for up to 10 days, at final ratios of from 1:1 to 1:10 (based on cell counts on the day of addition). In one particular embodiment, the ratio of particles to cells is 1:1 on the first day of stimulation and adjusted to 1:5 on the third and fifth days of stimulation. In another embodiment, particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:5 on the third and fifth days of stimulation. In another embodiment, the ratio of particles to cells is 2:1 on the first day of stimulation and adjusted to 1:10 on the third and fifth days of stimulation. In another embodiment, particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:10 on the third and fifth days of stimulation. One of skill in the art will appreciate that a variety of other ratios can be suitable for use in the present invention. In particular, ratios will vary depending on particle size and on cell size and type.


In further embodiments described herein, the immune effector cells, such as T cells, are combined with agent-coated beads, the beads and the cells are subsequently separated, and then the cells are cultured. In an alternative embodiment, prior to culture, the agent-coated beads and cells are not separated but are cultured together. In a further embodiment, the beads and cells are first concentrated by application of a force, such as a magnetic force, resulting in increased ligation of cell surface markers, thereby inducing cell stimulation.


By way of example, cell surface proteins can be ligated by allowing paramagnetic beads to which anti-CD3 and anti-CD28 are attached (3×28 beads) to contact the T cells. In one embodiment the cells (for example, 104 to 109 T cells) and beads (for example, DYNABEADS® M-450 CD3/CD28 T paramagnetic beads at a ratio of 1:1, or MACS® MicroBeads from Miltenyi Biotec) are combined in a buffer, for example, PBS (without divalent cations such as, calcium and magnesium). Again, those of ordinary skill in the art can readily appreciate any cell concentration can be used. For example, the target cell can be very rare in the sample and comprise only 0.01% of the sample or the entire sample (i.e., 100%) can comprise the target cell of interest. Accordingly, any cell number is within the context of the present invention. In certain embodiments, it can be desirable to significantly decrease the volume in which particles and cells are mixed together (i.e., increase the concentration of cells), to ensure maximum contact of cells and particles. For example, in one embodiment, a concentration of about 2 billion cells/ml is used. In another embodiment, greater than 100 million cells/ml is used. In a further embodiment, a concentration of cells of 10, 15, 20, 25, 30, 35, 40, 45, or 50 million cells/ml is used. In yet another embodiment, a concentration of cells from 75, 80, 85, 90, 95, or 100 million cells/ml is used. In further embodiments, concentrations of 125 or 150 million cells/ml can be used. Using high concentrations can result in increased cell yield, cell activation, and cell expansion. Further, use of high cell concentrations allows more efficient capture of cells that can weakly express target antigens of interest, such as CD28-negative T cells. Such populations of cells can have therapeutic value and would be desirable to obtain in certain embodiments. For example, using high concentration of cells allows more efficient selection of CD8+ T cells that normally have weaker CD28 expression.


In one embodiment described herein, the mixture can be cultured for several hours (about 3 hours) to about 14 days or any hourly integer value in between. In another embodiment, the mixture can be cultured for 21 days. In one embodiment of the invention the beads and the T cells are cultured together for about eight days. In another embodiment, the beads and T cells are cultured together for 2-3 days. Several cycles of stimulation may also be desired such that culture time of T cells can be 60 days or more. Conditions appropriate for T cell culture include an appropriate media (e.g., Minimal Essential Media or RPMI Media 1640 or, X-vivo 15, (Lonza)) that can contain factors necessary for proliferation and viability, including serum (e.g., fetal bovine or human serum), interleukin-2 (IL-2), insulin, IFN-.gamma., IL-4, IL-7, GM-CSF, IL-10, IL-12, IL-15, TGFbeta, and TNF-alpha or any other additives for the growth of cells known to the skilled artisan. Other additives for the growth of cells include, but are not limited to, surfactant, plasmanate, and reducing agents such as N-acetyl-cysteine and 2-mercaptoethanol. Media can include RPMI 1640, AIM-V, DMEM, MEM, alpha-MEM, F-12, X-Vivo 15, and X-Vivo 20, Optimizer, with added amino acids, sodium pyruvate, and vitamins, either serum-free or supplemented with an appropriate amount of serum (or plasma) or a defined set of hormones, and/or an amount of cytokine(s) sufficient for the growth and expansion of T cells. Antibiotics, e.g., penicillin and streptomycin, are included only in experimental cultures, not in cultures of cells that are to be infused into a subject. The target cells are maintained under conditions necessary to support growth, for example, an appropriate temperature (e.g., 37° C.) and atmosphere (e.g., air plus 5% CO2).


Ex vivo procedures are well known and are discussed more fully below. Briefly, cells are isolated from a mammal (for example, a human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein. The CAR-modified cell can be administered to a mammalian recipient to provide a therapeutic benefit. The mammalian recipient can be a human and the CAR-modified cell can be autologous with respect to the recipient. Alternatively, the cells can be allogeneic, syngeneic or xenogeneic with respect to the recipient.


The procedure for ex vivo expansion of hematopoietic stem and progenitor cells is described in U.S. Pat. No. 5,199,942, can be applied to the cells of the present invention. Other suitable methods are known in the art, therefore the present invention is not limited to any particular method of ex vivo expansion of the cells. Briefly, ex vivo culture and expansion of effector cells comprises: (1) collecting CD34+ hematopoietic stem and progenitor cells from a mammal from peripheral blood harvest or bone marrow explants; and (2) expanding such cells ex vivo. In addition to the cellular growth factors described in U.S. Pat. No. 5,199,942, other factors such as flt3-L, IL-1, IL-3 and c-kit ligand, can be used for culturing and expansion of the cells.


Effector cells that have been exposed to varied stimulation times can exhibit different characteristics. For example, typical blood or apheresed peripheral blood mononuclear cell products have a helper T cell population (TH, CD4±) that is greater than the cytotoxic or suppressor T cell population (Tc, CD8±). Ex vivo expansion of T cells by stimulating CD3 and CD28 receptors produces a population of T cells that prior to about days 8-9 consists predominately of TH cells, while after about days 8-9, the population of T cells comprises an increasingly greater population of Tc cells. Accordingly, depending on the purpose of treatment, infusing a subject with a T cell population comprising predominately of TH cells can be advantageous. Similarly, if an antigen-specific subset of Tc cells has been isolated it can be beneficial to expand this subset to a greater degree.


Further, in addition to CD4 and CD8 markers, other phenotypic markers vary significantly, but in large part, reproducibly during the course of the cell expansion process. Thus, such reproducibility enables the ability to tailor an activated T cell product for specific purposes.


In some cases, immune effector cells of the embodiments (e.g., T-cells) are co-cultured with activating and propagating cells (AaPCs), to aid in cell expansion. AaPCs can also be referred to as artificial Antigen Presenting cells (aAPCs). For example, antigen presenting cells (APCs) are useful in preparing therapeutic compositions and cell therapy products of the embodiments. In one aspect, the AaPCs can be genetically modified K562 cells. For general guidance regarding the preparation and use of antigen-presenting systems, see, e.g., U.S. Pat. Nos. 6,225,042, 6,355,479, 6,362,001 and 6,790,662; U.S. Patent Application Publication Nos. 2009/0017000 and 2009/0004142; and International Publication No. WO2007/103009. In yet a further aspect of the embodiments, culturing the genetically modified CAR cells comprises culturing the genetically modified CAR cells in the presence of dendritic cells or activating and propagating cells (AaPCs) that stimulate expansion of the CAR-expressing immune effector cells. In still further aspects, the AaPCs comprise a CAR-binding antibody or fragment thereof expressed on the surface of the AaPCs. The AaPCs can comprise additional molecules that activate or co-stimulate T-cells in some cases. The additional molecules can, in some cases, comprise membrane-bound Cy cytokines. In yet still further aspects, the AaPCs are inactivated or irradiated, or have been tested for and confirmed to be free of infectious material. In still further aspects, culturing the genetically modified CAR cells in the presence of AaPCs comprises culturing the genetically modified CAR cells in a medium comprising soluble cytokines, such as IL-15, IL-21 and/or IL-2. The cells can be cultured at a ratio of about 10:1 to about 1:10; about 3:1 to about 1:5; about 1:1 to about 1:3 (immune effector cells to AaPCs); or any range derivable therein. For example, the co-culture of T cells and AaPCs can be at a ratio of about 1:1, about 1:2 or about 1:3.


In one aspect, the AaPCs can express CD137L. In some aspects, the AaPCs can further express the antigen that is targeted by the CAR cell, for example MUC16, CD33, or ROR1 (full length, truncate or any variant thereof). In other aspects, the AaPCs can further express CD64, CD86, or mIL15. In certain aspects, the AaPCs can express at least one anti-CD3 antibody clone, such as, for example, OKT3 and/or UCHT1. In one aspect, the AaPCs can be inactivated (e.g., irradiated). In one aspect, the AaPCs have been tested and confirmed to be free of infectious material. Methods for producing such AaPCs are known in the art. In one aspect, culturing the CAR-modified T cell population with AaPCs can comprise culturing the cells at a ratio of about 10:1 to about 1:10; about 3:1 to about 1:5; about 1:1 to about 1:3 (T cells to AaPCs); or any range derivable therein. For example, the co-culture of T cells and AaPCs can be at a ratio of about 1:1, about 1:2 or about 1:3. In one aspect, the culturing step can further comprise culturing with an aminobisphosphonate (e.g., zoledronic acid).


In one aspect, the population of genetically modified CAR cells is immediately infused into a subject or cryopreserved. In another aspect, the population of genetically modified CAR cells is placed in a cytokine bath prior to infusion into a subject. In a further aspect, the population of genetically modified CAR cells is cultured and/or stimulated for no more than 1, 2, 3, 4, 5, 6, 7, 14, 21, 28, 35 42 days, 49, 56, 63 or 70 days. In some embodiments, the population of CAR-T cells is cultured and/or stimulated for at least 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30 or more days. In some embodiments, the population of CAR-T cells is cultured and/or stimulated for at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60 or more days. In some embodiments, the population of CAR expressing effector cells is cultured and/or stimulated for at least 7, 14, 21, 28, 35, 42, 49, 56, 63 or more days. In an embodiment, a stimulation includes the co-culture of the genetically modified CAR-T cells with AaPCs to promote the growth of CAR positive T cells. In another aspect, the population of genetically modified CAR cells is stimulated for not more than: 1× stimulation, 2× stimulation, 3× stimulation, 4× stimulation, 5× stimulation, 5× stimulation, 6× stimulation, 7× stimulation, 8× stimulation, 9× stimulation or 10× stimulation. In some instances, the genetically modified cells are not cultured ex vivo in the presence of AaPCs. In some specific instances, the method of the embodiment further comprises enriching the cell population for CAR-expressing immune effector cells (e.g., T-cells) after the transfection and/or culturing step. The enriching can comprise fluorescence-activated cell sorting (FACS) to sort for CAR-expressing cells. In a further aspect, the sorting for CAR-expressing cells comprises use of a CAR-binding antibody. The enriching can also comprise depletion of CD56+ cells. In yet still a further aspect of the embodiment, the method further comprises cryopreserving a sample of the population of genetically modified CAR cells.


In some cases, AaPCs are incubated with a peptide of an optimal length that allows for direct binding of the peptide to the MHC molecule without additional processing. Alternatively, the cells can express an antigen of interest (i.e., in the case of MHC-independent antigen recognition). Furthermore, in some cases, APCs can express an antibody that binds to either a specific CAR polypeptide or to CAR polypeptides in general (e.g., a universal activating and propagating cell (uAPC). Such methods are disclosed in WO2014/190273. In addition to peptide-MHC molecules or antigens of interest, the AaPC systems can also comprise at least one exogenous assisting molecule. Any suitable number and combination of assisting molecules can be employed. The assisting molecule can be selected from assisting molecules such as co-stimulatory molecules and adhesion molecules. Exemplary co-stimulatory molecules include CD70 and B7.1 (B7.1 was previously known as B7 and also known as CD80), which among other things, bind to CD28 and/or CTLA-4 molecules on the surface of T cells, thereby affecting, for example, T-cell expansion, Th1 differentiation, short-term T-cell survival, and cytokine secretion such as interleukin (IL)-2. Adhesion molecules can include carbohydrate-binding glycoproteins such as selectins, transmembrane binding glycoproteins such as integrins, calcium-dependent proteins such as cadherins, and single-pass transmembrane immunoglobulin (Ig) superfamily proteins, such as intercellular adhesion molecules (ICAMs) that promote, for example, cell-to-cell or cell-to-matrix contact. Exemplary adhesion molecules include LFA-3 and ICAMs, such as ICAM-1. Techniques, methods, and reagents useful for selection, cloning, preparation, and expression of exemplary assisting molecules, including co-stimulatory molecules and adhesion molecules, are exemplified in, e.g., U.S. Pat. Nos. 6,225,042, 6,355,479, and 6,362,001.


Cells selected to become AaPCs, preferably have deficiencies in intracellular antigen-processing, intracellular peptide trafficking, and/or intracellular MHC Class I or Class II molecule-peptide loading, or are poikilothermic (i.e., less sensitive to temperature challenge than mammalian cell lines), or possess both deficiencies and poikilothermic properties. Preferably, cells selected to become AaPCs also lack the ability to express at least one endogenous counterpart (e.g., endogenous MHC Class I or Class II molecule and/or endogenous assisting molecules as described above) to the exogenous MHC Class I or Class II molecule and assisting molecule components that are introduced into the cells. Furthermore, AaPCs preferably retain the deficiencies and poikilothermic properties that were possessed by the cells prior to their modification to generate the AaPCs. Exemplary AaPCs either constitute or are derived from a transporter associated with antigen processing (TAP)-deficient cell line, such as an insect cell line. An exemplary poikilothermic insect cells line is a Drosophila cell line, such as a Schneider 2 cell line (see, e.g., Schneider 1972 Illustrative methods for the preparation, growth, and culture of Schneider 2 cells, are provided in U.S. Pat. Nos. 6,225,042, 6,355,479, and 6,362,001.


In one embodiment, AaPCs are also subjected to a freeze-thaw cycle. In an exemplary freeze-thaw cycle, the AaPCs can be frozen by contacting a suitable receptacle containing the AaPCs with an appropriate amount of liquid nitrogen, solid carbon dioxide (i.e., dry ice), or similar low-temperature material, such that freezing occurs rapidly. The frozen APCs are then thawed, either by removal of the AaPCs from the low-temperature material and exposure to ambient room temperature conditions, or by a facilitated thawing process in which a lukewarm water bath or warm hand is employed to facilitate a shorter thawing time. Additionally, AaPCs can be frozen and stored for an extended period of time prior to thawing. Frozen AaPCs can also be thawed and then lyophilized before further use. Preferably, preservatives that might detrimentally impact the freeze-thaw procedures, such as dimethyl sulfoxide (DMSO), polyethylene glycols (PEGs), and other preservatives, are absent from media containing AaPCs that undergo the freeze-thaw cycle, or are essentially removed, such as by transfer of AaPCs to media that is essentially devoid of such preservatives.


In further embodiments, xenogenic nucleic acid and nucleic acid endogenous to the AaPCs, can be inactivated by crosslinking, so that essentially no cell growth, replication or expression of nucleic acid occurs after the inactivation. In one embodiment, AaPCs are inactivated at a point subsequent to the expression of exogenous MHC and assisting molecules, presentation of such molecules on the surface of the AaPCs, and loading of presented MHC molecules with selected peptide or peptides. Accordingly, such inactivated and selected peptide loaded AaPCs, while rendered essentially incapable of proliferating or replicating, retain selected peptide presentation function. Preferably, the crosslinking also yields AaPCs that are essentially free of contaminating microorganisms, such as bacteria and viruses, without substantially decreasing the antigen-presenting cell function of the AaPCs. Thus crosslinking maintains the important AaPC functions of while helping to alleviate concerns about safety of a cell therapy product developed using the AaPCs. For methods related to crosslinking and AaPCs, see for example, U.S. Patent Application Publication No. 20090017000.


In certain embodiments there are further provided an engineered antigen presenting cell (APC). Such cells can be used, for example, as described above, to propagate immune effector cells ex vivo. In further aspects, engineered APCs can, themselves be administered to a patient and thereby stimulate expansion of immune effector cells in vivo. Engineered APCs of the embodiments can, themselves, be used as a therapeutic agent. In other embodiments, the engineered APCs can used as a therapeutic agent that can stimulate activation of endogenous immune effector cells specific for a target antigen and/or to increase the activity or persistence of adoptively transferred immune effector cells specific to a target antigen.


As used herein the term “engineered APC” refers to cell(s) that comprises at least a first transgene, wherein the first transgene encodes a HLA. Such engineered APCs can further comprise a second transgene for expression of an antigen, such that the antigen is presented at the surface on the APC in complex with the HLA. In some aspects, the engineered APC can be a cell type that presented antigens (e.g., a dendritic cell). In further aspects, engineered APC can be produced from a cell type that does not normally present antigens, such a T-cell or T-cell progenitor (referred to as “T-APC”). Thus, in some aspects, an engineered APC of the embodiments comprises a first transgene encoding a target antigen and a second transgene encoding a human leukocyte antigen (HLA), such that the HLA is expressed on the surface of the engineered APC in complex with an epitope of the target antigen. In certain specific aspects, the HLA expressed in the engineered APC is HLA-A2.


In some aspects, an engineered APC of the embodiments can further comprise at least a third transgene encoding co-stimulatory molecule. The co-stimulatory molecule can be a co-stimulatory cytokine that can be a membrane-bound Cy cytokine. In certain aspects, the co-stimulatory cytokine is IL-15, such as membrane-bound IL-15. In some further aspects, an engineered APC can comprise an edited (or deleted) gene. For example, an inhibitory gene, such as PD-1, LIM-3, CTLA-4 or a TCR, can be edited to reduce or eliminate expression of the gene. An engineered APC of the embodiments can further comprise a transgene encoding any target antigen of interest. For example, the target antigen can be an infectious disease antigen or a tumor-associated antigen (TAA).


C. Rapid Manufacturing


In one embodiment of the present disclosure, the immune effector cells described herein are modified at a point-of-care site. In one embodiment of the present disclosure, the immune effector cells described herein are modified at or near a point-of-care site. In some cases, modified immune effector cells are also referred to as engineered T cells. In some cases, the facility or treatment site is at a hospital, at a facility (e.g., a medical facility), or at a treatment site near a subject in need of treatment. The subject undergoes apheresis and peripheral blood mononuclear cells (PBMCs) or a sub population of PBMC can be enriched for example, by elutriation or Ficoll separation. Enriched PBMC or a subpopulation of PBMC can be cryopreserved in any appropriate cryopreservation solution prior to further processing. In one instance, the elutriation process is performed using a buffer solution containing human serum albumin. Immune effector cells, such as T cells can be isolated by selection methods described herein. In one instance, the selection method for T cells includes beads specific for CD3 or beads specific for CD4 and CD8 on T cells. In one case, the beads can be paramagnetic beads. The harvested immune effector cells can be cryopreserved in any appropriate cryopreservation solution prior to modification. The immune effector cells can be thawed up to 24 hours, 36 hours, 48 hours, 72 hours or 96 hours ahead of infusion. The thawed cells can be placed in cell culture buffer, for example in cell culture buffer (e.g. RPMI) supplemented with fetal bovine serum (FBS) or human serum AB or placed in a buffer that includes cytokines such as IL-2 and IL-21, prior to modification. In another aspect, the harvested immune effector cells can be modified immediately without the need for cryopreservation. In one aspect, the elutriation step is eliminated completely.


In some cases, the immune effector cells are modified by engineering/introducing a one or more miRNA(s), a chimeric receptor, one or more cell tag(s), and/or cytokine(s) into the immune effector cells and then rapidly infused into a subject. In some cases, the sources of immune effector cells can include both allogeneic and autologous sources. In one case, the immune effector cells can be T cells or NK cells. In one case, the chimeric receptor can be a CAR. In another case, the cytokine can be IL-15 (for example, as part of a fusion protein with IL-15Rα) or IL-12. In yet another case, expression of cytokine is modulated by ligand inducible gene-switch expression systems described herein. For example, a ligand such as veledimex can be delivered to the subject to modulate the expression of the cytokine.


In another aspect, veledimex is provided at 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg or 100 mg. In a further aspect, lower doses of veledimex are provided, for example, 0.5 mg, 1 mg, 5 mg, 10 mg, 15 mg or 20 mg. In one embodiment, veledimex is administered to the subject 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days prior to infusion of the modified immune effector cells. In a further embodiment, veledimex is administered about once every 12 hours, about once every 24 hours, about once every 36 hours or about once every 48 hours, for an effective period of time to a subject post infusion of the modified immune effector cells. In one embodiment, an effective period of time for veledimex administration is about: 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 days. In other embodiments, veledimex can be re-administered after a rest period, after a drug holiday or when the subject experiences a relapse.


In certain cases, where an adverse effect on a subject is observed or when treatment is not needed, the cell tag can be activated, for example via cetuximab, for conditional in vivo ablation of modified immune effector cells comprising cell tags such as truncated epidermal growth factor receptor tags as described herein.


In some embodiments, such immune effectors cells are modified by the constructs as described herein through electroporation. In one instance, electroporation is performed with electroporators such as Lonza's Nucleofector™ electroporators. In other embodiments, the vector comprising the above-mentioned constructs is a non-viral or viral vector. In one case, the non-viral vector includes a Sleeping Beauty transposon-transposase system. In one instance, the immune effector cells are electroporated using a specific sequence. For example, the immune effector cells can be electroporated with one transposon followed by the DNA encoding the transposase followed by a second transposon. In another instance, the immune effector cells can be electroporated with all transposons and transposase at the same time. In another instance, the immune effector cells can be electroporated with a transposase followed by both transposons or one transposon at a time. While undergoing sequential electroporation, the immune effector cells can be rested for a period of time prior to the next electroporation step.


In some cases, the modified immune effector cells do not undergo a propagation and activation step. In some cases, the modified immune effector cells do not undergo an incubation or culturing step (e.g. ex vivo propagation). In some cases, the modified immune effector cells are place in PBS/EDTA buffer. In certain cases, the modified immune effector cells are placed in a buffer that includes IL-2 and IL-21 prior to infusion. In other instances, the modified immune effector cells are placed or rested in cell culture buffer, for example in cell culture buffer (e.g. RPMI) supplemented with fetal bovine serum (FBS) prior to infusion. Prior to infusion, the modified immune effector cells can be harvested, washed and formulated in saline buffer in preparation for infusion into the subject.


In one instance, the subject has been lymphodepleted prior to infusion. In other instances, lymphodepletion is not required and the modified immune effector cells are rapidly infused into the subject.


In a further instance, the subject undergoes minimal lymphodepletion. Minimal lymphodepletion herein refers to a reduced lymphodepletion protocol such that the subject can be infused within 1 day, 2 days or 3 days following the lymphodepletion regimen. In one instance, a reduced lymphodepletion protocol can include lower doses of fludarabine and/or cyclophosphamide. In another instance, a reduced lymphodepletion protocol can include a shortened period of lymphodepletion, for example 1 day or 2 days.


In some embodiments, the subject is not lymphodepleted prior to infusion.


In one embodiment, the immune effector cells are modified by engineering/introducing one or more miRNA(s), a chimeric receptor and a cytokine into said immune effector cells and then rapidly infused into a subject. In other cases, the immune effector cells are modified by engineering/introducing one or more miRNA(s), a chimeric receptor and a cytokine into said cells and then infused within at least: 0, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 hours into a subject. In other cases, immune effector cells are modified by engineering/introducing one or more miRNA(s), a chimeric receptor and a cytokine into the immune effector cells and then infused in 0 days, <1 day, <2 days, <3 days, <4 days, <5 days, <6 days or <7 days into a subject.


In other embodiments, a method of stimulating the proliferation and/or survival of engineered cells comprises obtaining a sample of cells from a subject, and transfecting cells of the sample of cells with one or more polynucleotides that comprise one or more transposons. In one embodiment, the transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, and a transposase effective to integrate said one or more polynucleotides into the genome of said cells, to provide a population of engineered cells. In an embodiment, the transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, gene switch polypeptides for ligand-inducible control of the cytokine and a transposase effective to integrate said one or more polynucleotides into the genome of said cells, to provide a population of engineered cells. In an embodiment, the gene switch polypeptides comprise i) a first gene switch polypeptide that comprises a DNA binding domain fused to a first nuclear receptor ligand binding domain, and ii) a second gene switch polypeptide that comprises a transactivation domain fused to a second nuclear receptor ligand binding domain. In some embodiments, the first gene switch polypeptide and the second gene switch polypeptide are connected by a linker. In one instance, lymphodepletion is not required prior to administration of the engineered cells to a subject.


In one instance, a method of in vivo propagation of engineered cells comprises obtaining a sample of cells from a subject, and transfecting cells of the sample of cells with one or more polynucleotides that comprise one or more transposons. In one embodiment, the transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, and a transposase effective to integrate said one or more polynucleotides into the genome of said cells, to provide a population of engineered cells. In an embodiment, the transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, gene switch polypeptides for ligand-inducible control of the cytokine and a transposase effective to integrate said one or more polynucleotides into the genome of said cells, to provide a population of engineered cells. In an embodiment, the gene switch polypeptides comprise i) a first gene switch polypeptide that comprises a DNA binding domain fused to a first nuclear receptor ligand binding domain, and ii) a second gene switch polypeptide that comprises a transactivation domain fused to a second nuclear receptor ligand binding domain. In some embodiments, the first gene switch polypeptide and the second gene switch polypeptide are connected by a linker. In one instance, lymphodepletion is not required prior to administration of the engineered cells to a subject.


In another embodiment, a method of enhancing in vivo persistence of engineered cells in a subject in need thereof comprises obtaining a sample of cells from a subject, and transfecting cells of the sample of cells with one or more polynucleotides that comprise one or more transposons. In some cases, one or more transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, and a transposase effective to integrate the DNA into the genome of said cells, to provide a population of engineered cells. In some cases, one or more transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, gene switch polypeptides for ligand-inducible control of the cytokine and a transposase effective to integrate the DNA into the genome of said cells, to provide a population of engineered cells. In some cases, the gene switch polypeptides comprise i) a first gene switch polypeptide that comprises a DNA binding domain fused to a first nuclear receptor ligand binding domain, and ii) a second gene switch polypeptide that comprises a transactivation domain fused to a second nuclear receptor ligand binding domain, wherein the first gene switch polypeptide and the second gene switch polypeptide are connected by a linker. In one instance, lymphodepletion is not required prior to administration of the engineered cells to a subject.


XIII. Kits and Compositions

One aspect of the disclosure relates to kits and compositions that comprise: a CAR, a cytokine, a cell tag, and/or one or more miRNAs as described previously, or nucleic acids encoding the same. In another aspect, the kits and compositions can include RHEOSWITCH® gene switch components. These kits and compositions can include multiple vectors each encoding different proteins or subsets of proteins. These vectors can be viral, non-viral, episomal, or integrating. In some embodiments, the vectors are transposons, e.g., Sleeping Beauty transposons. In other embodiments, the vectors can comprise sequences for serine recombinase-mediated integration.


In some embodiments, the kits and compositions include not only vectors but also cells and agents such as interleukins, cytokines, interleukins and chemotherapeutics, adjuvants, wetting agents, or emulsifying agents. In one embodiment the cells are T cells. In one embodiment the kits and composition includes IL-2. In one embodiment, the kits and compositions include IL-21. In one embodiment, the kits and compositions include Bcl-2, STAT3 or STATS inhibitors. In some embodiments, the kit includes IL-15, for example as part of a fusion protein with IL-15Rα.


Disclosed herein, in certain embodiments, are kits and articles of manufacture for use with one or more methods described herein. Such kits include a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. In one embodiment, the containers are formed from a variety of materials such as glass or plastic.


The articles of manufacture provided herein contain packaging materials. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, bags, containers, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.


For example, the container(s) include CAR-T cells (e.g., MUC16-, CD33-, and ROR1-specific CAR-T cells described herein), and optionally in addition with cytokines and/or chemotherapeutic agents disclosed herein. Such kits optionally include an identifying description or label or instructions relating to its use in the methods described herein.


A kit typically includes labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included.


In some embodiments, a label is on or associated with the container. In one embodiment, a label is on a container when letters, numbers or other characters forming the label are attached, molded or etched into the container itself; a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In one embodiment, a label is used to indicate that the contents are to be used for a specific therapeutic application. The label also indicates directions for use of the contents, such as in the methods described herein.


The present invention relates also to pharmaceutical compositions comprising a modified immune effector cell as described above. In certain embodiments, the composition comprising an immune effector cell (e.g., T cell) modified with sequences comprising one or more miRNA(s), a CAR, one or more cell tags, and/or one or more cytokines and optionally, components of the gene switch system as described herein. In certain embodiments, the immune effector cell is modified with Sleeping Beauty transposon(s) and Sleeping Beauty transposase. For example, the Sleeping Beauty transposon or transposons can include one or more miRNA(s), a CAR, one or more cell tags, one or more cytokines and optionally, components of the gene switch system as described herein. Therefore, in some instances, the modified T-cell can elicit a CAR-mediated T-cell response.


The cells activated and expanded as described herein can be utilized in the treatment and prevention of diseases that arise in individuals who are immunocompromised. In particular, the modified T cells of the invention are used in the treatment of malignancies. In certain embodiments, the cells of the invention are used in the treatment of patients at risk for developing malignancies. Thus, the methods for the treatment or prevention of malignancies comprising administering to a subject in need thereof, a therapeutically effective amount of the modified T cells of the invention. In embodiments, the cells activated and expanded as described herein can be utilized in the treatment of malignancies.


In addition to using a cell-based vaccine in terms of ex vivo immunization, the present invention also provides compositions and methods for in vivo immunization to elicit an immune response directed against an antigen in a patient.


Briefly, pharmaceutical compositions described herein can comprise a population comprising modified immune effector cells as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. Such compositions can comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione;


adjuvants (e.g., aluminum hydroxide); and preservatives. In some embodiments, compositions of the present invention are formulated for intravenous administration.


Pharmaceutical compositions described herein can be administered in a manner appropriate to the disease to be treated (or prevented). The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease.


When “an immunologically effective amount”, or “therapeutic amount” is indicated, the precise amount of the compositions described herein to be administered can be determined by a physician with consideration of individual differences in age, weight, and condition of the patient (subject). It can generally be stated that a pharmaceutical composition comprising the T cells described herein can be administered at a dosage of 104 to 109 cells/kg body weight, 105 to 106 cells/kg body weight, including all integer values within those ranges. T cell compositions can also be administered multiple times at these dosages. The cells can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al., New Eng. J. of Med. 319:1676, (1988)). The optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly.


In certain embodiments, it can be desired to administer activated T cells to a subject and then subsequently redraw blood (or have an apheresis performed), activate T cells therefrom, and reinfuse the patient with these activated and expanded T cells. This process can be carried out multiple times every few weeks. In certain embodiments, T cells can be activated from blood draws of from 10 cc to 400 cc. In certain embodiments, T cells are activated from blood draws of 20 cc, 30 cc, 40 cc, 50 cc, 60 cc, 70 cc, 80 cc, 90 cc, or 100 cc. Not to be bound by theory, using this multiple blood draw/multiple reinfusion protocol can serve to select out certain populations of T cells. In another embodiment, it can be desired to administer activated T cells of the subject composition following lymphodepletion of the patient, either via radiation or chemotherapy.


Formulations described herein can benefit from antioxidants, metal chelating agents, thiol containing compounds and other general stabilizing agents. Examples of such stabilizing agents, include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1% to about 1% w/v methionine, (c) about 0.1% to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v. polysorbate 20, (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (1) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.


A “carrier” or “carrier materials” include any commonly used excipients in pharmaceutics and should be selected on the basis of compatibility with the polynucleotides, vectors, and/or cells disclosed herein, and the release profile properties of the desired dosage form. Exemplary carrier materials include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like. “Pharmaceutically compatible carrier materials” can include, but are not limited to, acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, polyvinylpyrrollidone (PVP), cholesterol, cholesterol esters, sodium caseinate, soy lecithin, taurocholic acid, phosphotidylcholine, sodium chloride, tricalcium phosphate, dipotassium phosphate, cellulose and cellulose conjugates, sugars sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, and the like. See, e.g., Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999).


“Dispersing agents,” and/or “viscosity modulating agents” include materials that control the diffusion and homogeneity of a drug through liquid media or a granulation method or blend method. In some embodiments, these agents also facilitate the effectiveness of a coating or eroding matrix. Exemplary diffusion facilitators/dispersing agents include, e.g., hydrophilic polymers, electrolytes, Tween® 60 or 80, PEG, polyvinylpyrrolidone (PVP; commercially known as Plasdone®), and the carbohydrate-based dispersing agents such as, for example, hydroxypropyl celluloses (e.g., HPC, HPC-SL, and HPC-L), hydroxypropyl methylcelluloses (e.g., HPMC K100, HPMC K4M, HPMC K15M, and HPMC K100M), carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose acetate stearate (HPMCAS), noncrystalline cellulose, magnesium aluminum silicate, triethanolamine, polyvinyl alcohol (PVA), vinyl pyrrolidone/vinyl acetate copolymer (S630), 4-(1,1,3,3-tetramethylbutyl)-phenol polymer with ethylene oxide and formaldehyde (also known as tyloxapol), poloxamers (e.g., Pluronics F68®, F88®, and F108®, which are block copolymers of ethylene oxide and propylene oxide); and poloxamines (e.g., Tetronic 908®, also known as Poloxamine 908®, which is a tetrafunctional block copolymer derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine (BASF Corporation, Parsippany, N.J.)), polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, polyvinylpyrrolidone/vinyl acetate copolymer (S-630), polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, sodium carboxymethylcellulose, methylcellulose, polysorbate-80, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, polysorbate-80, sodium alginate, polyethoxylated sorbitan monolaurate, polyethoxylated sorbitan monolaurate, povidone, carbomers, polyvinyl alcohol (PVA), alginates, chitosans and combinations thereof. Plasticizers such as cellulose or triethyl cellulose can also be used as dispersing agents. Dispersing agents particularly useful in liposomal dispersions and self-emulsifying dispersions are dimyristoyl phosphatidyl choline, natural phosphatidyl choline from eggs, natural phosphatidyl glycerol from eggs, cholesterol and isopropyl myristate.


Combinations of one or more erosion facilitator with one or more diffusion facilitator can also be used in the present compositions.


The term “diluent” refers to chemical compounds that are used to dilute the compound of interest prior to delivery. Diluents can also be used to stabilize compounds because they can provide a more stable environment. Salts dissolved in buffered solutions (which also can provide pH control or maintenance) are utilized as diluents in the art, including, but not limited to a phosphate buffered saline solution. In certain embodiments, diluents increase bulk of the composition to facilitate compression or create sufficient bulk for homogenous blend for capsule filling. Such compounds include e.g., lactose, starch, mannitol, sorbitol, dextrose, microcrystalline cellulose such as Avicel®; dibasic calcium phosphate, dicalcium phosphate dihydrate; tricalcium phosphate, calcium phosphate; anhydrous lactose, spray-dried lactose; pregelatinized starch, compressible sugar, such as Di-Pac® (Amstar); mannitol, hydroxypropylmethylcellulose, hydroxypropylmethylcellulose acetate stearate, sucrose-based diluents, confectioner's sugar; monobasic calcium sulfate monohydrate, calcium sulfate dihydrate; calcium lactate trihydrate, dextrates; hydrolyzed cereal solids, amylose; powdered cellulose, calcium carbonate; glycine, kaolin; mannitol, sodium chloride; inositol, bentonite, and the like.


“Filling agents” include compounds such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.


“Lubricants” and “glidants” are compounds that prevent, reduce or inhibit adhesion or friction of materials. Exemplary lubricants include, e.g., stearic acid, calcium hydroxide, talc, sodium stearyl fumerate, a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil (Sterotex®), higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, glycerol, talc, waxes, Stearowet®, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol (e.g., PEG-4000) or a methoxypolyethylene glycol such as Carbowax™ sodium oleate, sodium benzoate, glyceryl behenate, polyethylene glycol, magnesium or sodium lauryl sulfate, colloidal silica such as Syloid™, Cab-O-Sil®, a starch such as corn starch, silicone oil, a surfactant, and the like.


“Plasticizers” are compounds used to soften the microencapsulation material or film coatings to make them less brittle. Suitable plasticizers include, e.g., polyethylene glycols such as PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, triethyl cellulose and triacetin. In some embodiments, plasticizers can also function as dispersing agents or wetting agents.


“Solubilizers” include compounds such as triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, sodium lauryl sulfate, sodium doccusate, vitamin E TPGS, dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropylmethyl cellulose, hydroxypropyl cyclodextrins, ethanol, n-butanol, isopropyl alcohol, cholesterol, bile salts, polyethylene glycol 200-600, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide and the like.


“Stabilizers” include compounds such as any antioxidation agents, buffers, acids, preservatives and the like.


“Suspending agents” include compounds such as polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, vinyl pyrrolidone/vinyl acetate copolymer (S630), polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, hydroxymethylcellulose acetate stearate, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, hydroxyethylcellulose, polysorbate-80, sodium alginate, polyethoxylated sorbitan monolaurate, polyethoxylated sorbitan monolaurate, povidone and the like.


“Surfactants” include compounds such as sodium lauryl sulfate, sodium docusate, Tween 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like. Some other surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40. In some embodiments, surfactants can be included to enhance physical stability or for other purposes.


“Viscosity enhancing agents” include, e.g., methyl cellulose, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose acetate stearate, hydroxypropylmethyl cellulose phthalate, carbomer, polyvinyl alcohol, alginates, acacia, chitosans and combinations thereof.


“Wetting agents” include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium docusate, sodium oleate, sodium lauryl sulfate, sodium doccusate, triacetin, Tween 80, vitamin E TPGS, ammonium salts and the like.


XIV. Methods of Treatment

The present invention also relates to a method of treating a disease or disorder comprising administering a modified immune effector cell of the present disclosure to a subject in need thereof. In certain embodiments, the cell is administered in a therapeutically effective amount.


The present invention also relates to the use of a modified immune effector cell of the present disclosure in the manufacture of a medicament for the treatment of a disease or disorder.


In some embodiments, the disease can be cancer. In certain aspects, the cancer can be hematological or a solid tumor. In other instances, the cancer is a hematologic malignancy. In some cases, the cancer is a metastatic cancer. In some cases, the cancer is a relapsed or refractory cancer. In certain aspects, the cancer is selected from B cell cancer, e.g., multiple myeloma, Waldenstrom's macroglobulinemia, the heavy chain diseases, such as, for example, alpha chain disease, gamma chain disease, and mu chain disease, benign monoclonal gammopathy, and immunocytic amyloidosis, melanomas, breast cancer, lung cancer, bronchus cancer, colorectal cancer, prostate cancer (e.g., metastatic, hormone refractory prostate cancer), pancreatic cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues, and the like. Other non-limiting examples of types of cancers that can be treated by the methods of the present disclosure include human sarcomas and carcinomas, e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's sarcoma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, colorectal cancer, pancreatic cancer, breast cancer, beast adenocarcinomas e.g. triple negative breast cancer, ovarian cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, liver cancer, hepatocellular carcinoma (HCC), choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, bone cancer, brain tumor, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma; leukemias, e.g., acute myeloid leukemia, acute lymphocytic leukemia, mantle cell lymphoma, acute lymphoblastic leukemia, and acute myelocytic leukemia (myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia); diffuse large B-cell lymphoma; and polycythemia vera, lymphoma (Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, and heavy chain disease.


In some instances, the cancer is a solid tumor. Exemplary solid tumors include, but are not limited to, anal cancer; appendix cancer; bile duct cancer (i.e., cholangiocarcinoma); bladder cancer; brain tumor; breast cancer; cervical cancer; colon cancer; cancer of Unknown Primary (CUP); esophageal cancer; eye cancer; fallopian tube cancer; gastroenterological cancer; kidney cancer; liver cancer; lung cancer; medulloblastoma; melanoma; oral cancer; ovarian cancer; pancreatic cancer; parathyroid disease; penile cancer; pituitary tumor; prostate cancer; rectal cancer; skin cancer; stomach cancer; testicular cancer; throat cancer; thyroid cancer; uterine cancer; vaginal cancer; or vulvar cancer.


In some instances, the cancer is a hematologic malignancy. In some cases, a hematologic malignancy comprises a lymphoma, a leukemia, a myeloma, or a B-cell malignancy. In some cases, a hematologic malignancy comprises a lymphoma, a leukemia or a myeloma. In some instances, exemplary hematologic malignancies include chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), high risk CLL, non-CLL/SLL lymphoma, prolymphocytic leukemia (PLL), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia, multiple myeloma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, Burkitt's lymphoma, non-Burkitt high grade B cell lymphoma, primary mediastinal B-cell lymphoma (PMBL), immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, B cell prolymphocytic leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis. In some embodiments, the hematologic malignancy comprises a myeloid leukemia. In some embodiments, the hematologic malignancy comprises acute myeloid leukemia (AML) or chronic myeloid leukemia (CML).


In some instances, disclosed herein are methods of administering to a subject having a hematologic malignancy selected from chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), high risk CLL, non-CLL/SLL lymphoma, prolymphocytic leukemia (PLL), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia, multiple myeloma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, Burkitt's lymphoma, non-Burkitt high grade B cell lymphoma, primary mediastinal B-cell lymphoma (PMBL), immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, B cell prolymphocytic leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis a modified effector cell described herein. In some instances, disclosed herein are methods of administering to a subject having a hematologic malignancy selected from AML or CML a modified effector cell to the subject.


In still other embodiments, the epithelial cancer is non-small-cell lung cancer, nonpapillary renal cell carcinoma, cervical carcinoma, ovarian carcinoma (e.g., serous ovarian carcinoma), or breast carcinoma. The epithelial cancers can be characterized in various other ways including, but not limited to, serous, endometrioid, mucinous, clear cell, brenner, or undifferentiated. In some embodiments, the methods and compositions of the present disclosure are used in the treatment, diagnosis, and/or prognosis of lymphoma or its subtypes, including, but not limited to, mantle cell lymphoma.


In some embodiments, the disease or disorder is associated with the overexpression of an antigen. In certain embodiments, the antigen is CD19, CD33, ROR1, MUC1, or MUC16. In some embodiments, the disease is ovarian cancer, a myelodysplastic syndrome (MDS).


In some embodiments, disclosed herein are methods of administering a modified effector cell comprising a polypeptide described herein to a subject having a disorder, for instance a cancer. In some cases, the cancer is a cancer associated with an expression of CD19, CD20, CD33, CD44, BCMA, CD123, EGFRvIII, α-Folate receptor, CAIX, CD30, ROR1, CEA, EGP-2, EGP-40, HER2, HER3, Folate-binding Protein, GD2, GD3, IL-13R-a2, KDR, EDB-F, mesothelin, GPC3, CSPG4, HER1/HER3, HER2, CD44v6, CD44v7/v8, CD20, CD174, CD138, L1-CAM, FAP, c-MET, PSCA, CS1, CD38, IL-11Rα, EphA2, CLL-1, CD22, EGFR, Folate receptor α, Mucins such as MUC1 or MUC16, MAGE-A1, h5T4, PSMA, CSPG4, TAG-72 or VEGF-R2.


In some embodiments, the disease is associated with the overexpression of MUC16. In certain such embodiments, the disease is ovarian cancer, breast cancer, pancreatic cancer, endometrial cancer, or lung cancer.


In some embodiments, the disease is associated with the overexpression of CD33. In certain such embodiments, the disease is acute myeloid leukemia (AML) or a myelodysplastic syndrome (MDS).


In some embodiments, the disease is associated with the overexpression of ROR1. In certain such embodiments, the disease involves a hematological tumor, for example chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), acute lymphoblastic leukemia (ALL), and diffuse large B-cell lymphoma (DLBCL). In certain such embodiments, the disease involves a solid tumor, for example breast adenocarcinomas encompassing triple negative breast cancer (TNBC), pancreatic cancer, ovarian cancer, and lung adenocarcinoma.


In another embodiment, a method of treating a subject with a solid tumor comprises obtaining a sample of cells from a subject, transfecting cells of the sample with one or more polynucleotides that comprise one or more transposons, and administering the population of engineered cells to the subject. In one instance, lymphodepletion is not required prior to administration of the engineered cells to a subject. In some embodiments, genetically modified T cells can be expanded and transferred into patients treated with or without preconditioning lymphodepletion according to well-known protocols. In some cases, the one or more transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, and a transposase effective to integrate the DNA into the genome of the cells. In some cases, the one or more transposons encode one or more miRNA(s), a chimeric antigen receptor (CAR), a cytokine, one or more cell tags, gene switch polypeptides for ligand-inducible control of the cytokine and a transposase effective to integrate the DNA into the genome of the cells. In some cases, the gene switch polypeptides comprise: i) a first gene switch polypeptide that comprises a DNA binding domain fused to a first nuclear receptor ligand binding domain, and ii) a second gene switch polypeptide that comprises a transactivation domain fused to a second nuclear receptor ligand binding domain, wherein the first gene switch polypeptide and second gene switch polypeptide are connected by a linker. In some cases, the cells are transfected via electroporation. In some cases, the polynucleotides encoding the gene switch polypeptides are modulated by a promoter. In some cases, the promoter is a tissue-specific promoter or an EF1A promoter or functional variant thereof. In some cases, the tissue-specific promoter comprises a T cell specific response element or an NFAT response element. In some cases, the cytokine comprises at least one of IL-1, IL-2, IL-15, IL-12, IL-21, a fusion of IL-15, IL-15R or an IL-15 variant. In some cases, the cytokine is in secreted form. In some cases, the cytokine is in membrane-bound form. In some cases, the cells are NK cells, NKT cells, T-cells or T-cell progenitor cells. In some cases, the cells are administered to a subject (e.g. by infusing the subject with the engineered cells). In some cases, the method further comprises administering an effective amount of a ligand (e.g. veledimex) to induce expression of the cytokine. In some cases, the transposase is salmonid-type Tc 1-like transposase. In some cases, the transposase is SB11 or SB100x transposase. In other cases, the transposase is PiggyBac. In some cases, the cell tag comprises at least one of a HER1 truncated variant or a CD20 truncated variant.


Studies conducted in patients with metastatic melanoma demonstrated that lymphodepletion prior to adoptive cell transfer dramatically improves the efficacy of therapy with in vitro expanded tumor-infiltrating lymphocytes (TILs). Muranski P, et al., “Increased intensity lymphodepletion and adoptive immunotherapy—how far can we go?” Nat Clin Pract Oncol. 2006; 3(12):668-681. Lymphodepletion likely works by multiple mechanisms, including eliminating sinks for homeostatic cytokines, such as interleukin-2 (IL-2), IL-7, and IL-15, eradicating immunosuppressive elements, such as regulatory T cells and myeloid-derived suppressor cells, inducing costimulatory molecules and downregulating indoleamine 2,3-dioxygenase in tumor cells, and promoting expansion, function, and persistence of adoptively transferred T cells. These experiences led to the use of lymphodepletion in clinical trials with CAR T-cell therapy. Kochenderfer et al showed an association between an increase in serum IL-15 levels post-lymphodepletion and clinical response after anti-CD19 CAR T-cell therapy. Kochenderfer J N, et al., “Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels,” J Clin Oncol. 2017; 35(16):1803-1813. Turtle and colleagues showed that the addition of fludarabine to cyclophosphamide (Cy/Flu) in the lymphodepleting regimen was associated with improved anti-CD19 CAR T-cell expansion and persistence and better clinical outcome compared with non-Flu lymphodepleting regimens in patients with non-Hodgkin lymphoma. Turtle C J, et al., “Immunotherapy of non-Hodgkin's lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells,” Sci Transl Med. 2016; (8)355-355ra116. Other studies indicated that the magnitude of the CAR T-cell expansion in vivo with respect to both the peak and the area under the curve over the first month may be associated with response and/or durability. Neelapu S S, et al., “Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma,” N Engl J Med. 2017; 377(26):2531-2544.


In some embodiments, the subject is subjected to lymphodepletion before the step of administering the modified immune effector cells to the subject. As used herein, “lymphodepletion” involves methods that reduce the number of lymphocytes in a subject, for example by administration of a lymphodepletion agent. Examples of lymphodepletion include nonmyeloablative lymphodepleting chemotherapy, myeloablative lymphodepleting chemotherapy. Lymphodepletion can also be attained by partial body or whole body fractioned radiation therapy. A lymphodepletion agent can be a chemical compound or composition capable of decreasing the number of functional lymphocytes in a mammal when administered to the mammal. One example of such an agent is one or more chemotherapeutic agents. Such agents and dosages are known, and can be selected by a treating physician depending on the subject to be treated. Examples of lymphodepletion agents include, but are not limited to, fludarabine, cyclophosphamide, cladribine, denileukin diftitox, or combinations thereof. In some embodiments, the subject is not subjected to lymphodepletion before the step of administering the modified immune effector cells to the subject.


In some embodiments, lymphodepletion is performed by administration of cyclophosphamide at a dose of about 10 mg/kg to about 100 mg/kg, preferably about 40 mg/kg to about 80 mg/kg, for example about 60 mg/kg. In such embodiments, the cyclophosphamide can be administered concomitantly with fludarabine at a dose of about 10 mg/m2 to about 50 mg/m2, preferably about 20 mg/m2 to about 40 mg/m2, for example about 30 mg/m2. In some embodiments, lymphodepletion is performed by administration of fludarabine at a dose of about 10 mg/m2 to about 50 mg/m2, preferably about 20 mg/m2 to about 40 mg/m2, for example about 30 mg/m2. In such embodiments, the fludarabine can be administered concomitantly with cyclophosphamide at a dose of about 200 mg/m2 to about 900 mg/m2, preferably about 400 to about 600 mg/m2, for example 500 mg/m2.


In some embodiments, patients or subjects are not lymphodepleted prior to blood being withdrawn to produce the autologous modified immune effector cells.


In some embodiments, the modified immune effector cells are autologous to the subject. In some embodiments, the modified immune effector cells are allogeneic to the subject.


In some embodiments, an amount of modified immune effector cells that is administered to a subject in need thereof and the amount is determined based on the efficacy and the potential of inducing a cytokine-associated toxicity.


The administration of compositions described herein can be carried out in any convenient manner, including by aerosol inhalation, injection, ingestion, transfusion, implantation or transplantation. The compositions described herein can be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally. In one embodiment, the T cell compositions of the present invention are administered to a patient by intradermal or subcutaneous injection. In another embodiment, the immune effector cell compositions of the present invention are administered by i.v. injection. The compositions of T cells can be injected directly into a lymph node, or site of primary tumor or metastasis.


The dosage of the above treatments to be administered to a patient will vary with the precise nature of the condition being treated and the recipient of the treatment. The scaling of dosages for human administration can be performed according to art-accepted practices. For example, the dose of the above treatment can be in the range of 1×104 CAR+ cells/kg to 5×106 CAR+ cells/kg. Exemplary doses can be 1×102 CAR+ cells/kg, 1×103 CAR+ cells/kg, 1×104 CAR+ cells/kg, 1×105 CAR+ cells/kg, 3×105 CAR+ cells/kg, 1×106 CAR+ cells/kg, 5×106 CAR+ cells/kg, 1×107 CAR+ cells/kg, 1×108 CAR+ cells/kg or 1×109 CAR+ cells/kg. The appropriate dose can be adjusted accordingly for an adult or a pediatric patient.


Alternatively, a typical amount of immune effector cells administered to a mammal (e.g., a human) can be, for example, in the range of one hundred, one thousand, ten thousand, one million to 100 billion cells; however, amounts below or above this exemplary range are within the scope of the invention. For example, the dose of inventive host cells can be about 1 million to about 50 billion cells (e.g., about 5 million cells, about 25 million cells, about 500 million cells, about 1 billion cells, about 5 billion cells, about 20 billion cells, about 30 billion cells, about 40 billion cells, or a range defined by any two of the foregoing values), about 10 million to about 100 billion cells (e.g., about 20 million cells, about 30 million cells, about 40 million cells, about 60 million cells, about 70 million cells, about 80 million cells, about 90 million cells, about 10 billion cells, about 25 billion cells, about 50 billion cells, about 75 billion cells, about 90 billion cells, or a range defined by any two of the foregoing values), about 100 million cells to about 50 billion cells (e.g., about 120 million cells, about 250 million cells, about 350 million cells, about 450 million cells, about 650 million cells, about 800 million cells, about 900 million cells, about 3 billion cells, about 30 billion cells, about 45 billion cells, or a range defined by any two of the foregoing values).


Therapeutic or prophylactic efficacy can be monitored by periodic assessment of treated patients. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs. However, other dosage regimens can be useful and are within the scope of the invention. The desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition.


In some embodiments, an amount of modified effector cells is administered to a subject in need thereof and the amount is determined based on the efficacy and the potential of inducing a cytokine-associated toxicity. In another embodiment, the modified effector cells are CARP and CD3+ cells. In some cases, an amount of modified effector cells comprises about 104 to about 109 modified effector cells/kg. In some cases, an amount of modified effector cells comprises about 104 to about 105 modified effector cells/kg. In some cases, an amount of modified effector cells comprises about 105 to about 106 modified effector cells/kg. In some cases, an amount of modified effector cells comprises about 106 to about 107 modified effector cells/kg. In some cases, an amount of modified effector cells comprises >104 but ≤105 modified effector cells/kg. In some cases, an amount of modified effector cells comprises >105 but ≤106 modified effector cells/kg. In some cases, an amount of modified effector cells comprises >106 but ≤107 modified effector cells/kg.


In one embodiment, the modified immune effector cells are targeted to the cancer via regional delivery directly to the tumor tissue. For example, in ovarian cancer, the modified immune effector cells can be delivered intraperitoneally (IP) to the abdomen or peritoneal cavity. Such IP delivery can be performed via a port or pre-existing port placed for delivery of chemotherapy drugs. Other methods of regional delivery of modified immune effector cells can include catheter infusion into resection cavity, ultrasound guided intratumoral injection, hepatic artery infusion or intrapleural delivery.


In one embodiment, a subject in need thereof, can begin therapy with a first dose of modified immune effector cells delivered via IP followed by a second dose of modified immune effector cells delivered via IV. In a further embodiment, the second dose of modified immune effector cells can be followed by subsequent doses which can be delivered via IV or IP. In one embodiment, the duration between the first and second or further subsequent dose can be about: 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 days. In one embodiment, the duration between the first and second or further subsequent dose can be about: 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, or 36 months. In another embodiment, the duration between the first and second or further subsequent dose can be about: 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 years.


In another embodiment, a catheter can be placed at the tumor or metastasis site for further administration of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 doses of modified immune effector cells. In some cases, doses of modified effector cells can comprise about 102 to about 109 modified effector cells/kg. In cases where toxicity is observed, doses of modified effector cells can comprise about 102 to about 105 modified effector cells/kg. In some cases, doses of modified effector cells can start at about 102 modified effector cells/kg and subsequent doses can be increased to about: 104, 105, 106, 107, 108 or 109 modified effector cells/kg.


The immune effector cells expressing the disclosed nucleic acid sequences, or a vector comprising the those nucleic acid sequences, can be administered with one or more additional therapeutic agents, which can be co-administered to the mammal. By “co-administering” is meant administering one or more additional therapeutic agents and the inventive host cells or the inventive vector sufficiently close in time to enhance the effect of one or more additional therapeutic agents, or vice versa. In this regard, the immune effector cells described herein or a vector described herein can be administered simultaneously with one or more additional therapeutic agents, or first, and the one or more additional therapeutic agents can be administered second, or vice versa. Alternatively, the disclosed immune effector cells or the vectors described herein and the one or more additional therapeutic agents can be administered simultaneously.


An example of a therapeutic agents that can be included in or co-administered with the inventive host cells and/or the inventive vectors are interleukins, cytokines, interferons, adjuvants and chemotherapeutic agents. In some embodiments, the additional therapeutic agents are IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G-CSF, M-CSF, LT-beta, TNF-alpha, growth factors, and hGH, a ligand of human Toll-like receptor TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, and TLR10.


A. Modified Immune Effector Cell Doses


In some embodiments, an amount of modified immune effector cells administered comprises about 102 to about 109 modified effector cells/kg of the subject's body weight. In some embodiments, an amount of modified immune effector cells administered comprises about 103 to about 109 modified effector cells/kg of the subject's body weight. In some embodiments, an amount of modified immune effector cells administered comprises about 104 to about 109 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 105 to about 109 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 105 to about 108 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 105 to about 107 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 106 to about 109 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 106 to about 108 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 107 to about 109 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 105 to about 106 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 106 to about 107 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 107 to about 108 modified effector cells/kg of the subject's body weight. In some cases, an amount of modified effector cells comprises about 108 to about 109 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 109 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 108 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 107 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 106 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 105 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 104 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 103 modified effector cells/kg of the subject's body weight. In some instances, an amount of modified effector cells comprises about 102 modified effector cells/kg of the subject's body weight.


In some embodiments, the modified immune effector cells are modified CAR-T cells. In some instances, the modified CAR-T cells further comprise one or more miRNA(s) as described herein. In some cases, an amount of modified CAR-T cells comprises about 104 to about 109 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 105 to about 109 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 105 to about 108 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 105 to about 107 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 106 to about 109 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 106 to about 108 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 107 to about 109 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 105 to about 106 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 106 to about 107 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 107 to about 108 modified CAR-T cells/kg of the subject's body weight. In some cases, an amount of modified CAR-T cells comprises about 108 to about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of modified CAR-T cells comprises about 109 modified CAR-T cells/kg of the subject's body weight. In some instances, an amount of modified CAR-T cells comprises about 108 modified CAR-T cells/kg of the subject's body weight. In some instances, an amount of modified CAR-T cells comprises about 107 modified CAR-T cells/kg of the subject's body weight. In some instances, an amount of modified CAR-T cells comprises about 106 modified CAR-T cells/kg of the subject's body weight. In some instances, an amount of modified CAR-T cells comprises about 105 modified CAR-T cells/kg of the subject's body weight.


In some embodiments, the modified CAR-T cells are CD19-specific CAR-T cells. In some cases, an amount of CD19-specific CAR-T cells comprises about 105 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 105 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 105 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 106 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 106 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 107 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 105 to about 106 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 106 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 107 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD19-specific CAR-T cells comprises about 108 to about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD19-specific CAR-T cells comprises about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD19-specific CAR-T cells comprises about 108 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD19-specific CAR-T cells comprises about 107 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD19-specific CAR-T cells comprises about 106 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD19-specific CAR-T cells comprises about 105 CAR-T cells/kg of the subject's body weight.


In some embodiments, the modified CAR-T cells are CD33-specific CAR-T cells. In some cases, an amount of CD33-specific CAR-T cells comprises about 105 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 105 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 105 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 106 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 106 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 107 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 105 to about 106 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 106 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 107 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of CD33-specific CAR-T cells comprises about 108 to about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD33-specific CAR-T cells comprises about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD33-specific CAR-T cells comprises about 108 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD33-specific CAR-T cells comprises about 107 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD33-specific CAR-T cells comprises about 106 CAR-T cells/kg of the subject's body weight. In some instances, an amount of CD33-specific CAR-T cells comprises about 105 CAR-T cells/kg of the subject's body weight.


In some embodiments, the modified CAR-T cells are MUC1-specific CAR-T cells. In some cases, an amount of MUC1-specific CAR-T cells comprises about 105 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 105 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 105 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 106 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 106 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 107 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 105 to about 106 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 106 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 107 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC1-specific CAR-T cells comprises about 108 to about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC1-specific CAR-T cells comprises about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC1-specific CAR-T cells comprises about 108 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC1-specific CAR-T cells comprises about 107 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC1-specific CAR-T cells comprises about 106 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC1-specific CAR-T cells comprises about 105 CAR-T cells/kg of the subject's body weight.


In some embodiments, the modified CAR-T cells are MUC16-specific CAR-T cells. In some cases, an amount of MUC16-specific CAR-T cells comprises about 105 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 105 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 105 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 106 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 106 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 107 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 105 to about 106 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 106 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 107 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of MUC16-specific CAR-T cells comprises about 108 to about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC16-specific CAR-T cells comprises about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC16-specific CAR-T cells comprises about 108 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC16-specific CAR-T cells comprises about 107 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC16-specific CAR-T cells comprises about 106 CAR-T cells/kg of the subject's body weight. In some instances, an amount of MUC16-specific CAR-T cells comprises about 105 CAR-T cells/kg of the subject's body weight.


In some embodiments, the modified CAR-T cells are ROR1-specific CAR-T cells. In some cases, an amount of ROR1-specific CAR-T cells comprises about 105 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 105 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 105 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 106 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 106 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 107 to about 109 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 105 to about 106 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 106 to about 107 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 107 to about 108 CAR-T cells/kg of the subject's body weight. In some cases, an amount of ROR1-specific CAR-T cells comprises about 108 to about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of ROR1-specific CAR-T cells comprises about 109 CAR-T cells/kg of the subject's body weight. In some instances, an amount of ROR1-specific CAR-T cells comprises about 108 CAR-T cells/kg of the subject's body weight. In some instances, an amount of ROR1-specific CAR-T cells comprises about 107 CAR-T cells/kg of the subject's body weight. In some instances, an amount of ROR1-specific CAR-T cells comprises about 106 CAR-T cells/kg of the subject's body weight. In some instances, an amount of ROR1-specific CAR-T cells comprises about 105 CAR-T cells/kg of the subject's body weight.


In some embodiments, the modified T cells are engineered TCR T-cells. In some cases, an amount of engineered TCR T-cells comprises about 105 to about 109 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 105 to about 108 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 105 to about 107 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 106 to about 109 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 106 to about 108 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 107 to about 109 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 105 to about 106 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 106 to about 107 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 107 to about 108 TCR cells/kg of the subject's body weight. In some cases, an amount of engineered TCR cells comprises about 108 to about 109 TCR cells/kg of the subject's body weight. In some instances, an amount of engineered TCR cells comprises about 109 TCR cells/kg of the subject's body weight. In some instances, an amount of engineered TCR cells comprises about 108 TCR cells/kg of the subject's body weight. In some instances, an amount of engineered TCR cells comprises about 107 TCR cells/kg of the subject's body weight. In some instances, an amount of engineered TCR cells comprises about 106 TCR cells/kg of the subject's body weight. In some instances, an amount of engineered TCR cells comprises about 105 TCR cells/kg of the subject's body weight.


It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.


In some embodiments, the cancer whose phenotype is determined by the method of the present disclosure is an epithelial cancer such as, but not limited to, bladder cancer, breast cancer, cervical cancer, colon cancer, gynecologic cancers, renal cancer, laryngeal cancer, lung cancer, oral cancer, head and neck cancer, ovarian cancer, pancreatic cancer, prostate cancer, or skin cancer. In other embodiments, the cancer is breast cancer, prostate cancer, lung cancer, or colon cancer.


B. Combination Therapies


In certain embodiments, the modified immune effector cells and compositions thereof described herein may be used, alone or with other therapies, to treat cancers that have about 0, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95%, 96%, 97%, 98%, or 99% average response rate to standard therapy (including but not limited to chemotherapy, chemotherapy and current clinical trial therapies). Such cancers include but are not limited to, Hodgkin's lymphoma, melanoma, non-small cell lung cancer (NSCLC), microsatellite instability (MSI)-high or mismatch repair (MMR)-deficient solid tumors, CSCC, RCC, CRC, melanoma, Merkel cell cancer, bladder cancer, RCC, hepatocellular carcinoma (HCC), head & neck cancer (H&N), cervical cancer, gastric cancer, small cell lung cancer (SCLC), endometrial cancer, mesothelioma, ovarian cancer, triple negative breast cancer (TNBC), breast cancer, colorectal cancer (CRC), pancreatic cancer, prostate cancer.


In some embodiments, the compositions described herein can be administered as a combination therapy with an additional therapeutic agent. In some embodiments, an additional therapeutic agent comprises a biological molecule, such as an antibody. For example, treatment can involve the combined administration of the modified immune effector cells as described herein with antibodies against tumor-associated antigens including, but not limited to, antibodies that bind EGFR, HER2/ErbB2, and/or VEGF. In certain embodiments, the additional therapeutic agent is an antibody specific for a cancer stem cell marker. In certain embodiments, the additional therapeutic agent is an antibody that is an angiogenesis inhibitor (e.g., an anti-VEGF or VEGF receptor antibody). In certain embodiments, the additional therapeutic agent is bevacizumab (AVASTIN), ramucirumab, trastuzumab (HERCEPTIN), pertuzumab (OMNITARG), panitumumab (VECTIBIX), nimotuzumab, zalutumumab, or cetuximab (ERBITUX). In some embodiments, an additional therapeutic agent comprises an agent such as a small molecule. For example, treatment can involve the combined administration of the modified immune effector cells as described herein with a small molecule that acts as an inhibitor against tumor-associated antigens including, but not limited to, EGFR, HER2 (ErbB2), and/or VEGF. In some embodiments, the modified immune effector cells as described herein administered in combination with a protein kinase inhibitor selected from the group consisting of: gefitinib (IRESSA), erlotinib (TARCEVA), sunitinib (SUTENT), lapatanib, vandetanib (ZACTIMA), AEE788, CI-1033, cediranib (RECENTIN), sorafenib (NEXAVAR), and pazopanib (GW786034B). In some embodiments, an additional therapeutic agent comprises an mTOR inhibitor. In another embodiment, the additional therapeutic agent is chemotherapy or other inhibitors that reduce the number of TREG cells. In certain embodiments, the therapeutic agent is cyclophosphamide or an anti-CTLA4 antibody. In another embodiment, the additional therapeutic reduces the presence of myeloid-derived suppressor cells. In a further embodiment, the additional therapeutic is carbotaxol. In a further embodiment, the additional therapeutic agent is ibrutinib. In some embodiments, an additional therapeutic agent comprises an anti-PD-1 or anti-PDL1 inhibitor. In some embodiments, the method can further comprise one or more checkpoint inhibitors in combination with modified immune effector cells as described herein. In some embodiments, the additional checkpoint inhibitor can be an anti-CTLA-4 antibody. The anti-CTLA-4 antibody (e.g., ipilimumab) has shown durable anti-tumor activities and prolonged survival in participants with advanced melanoma, resulting in its Food and Drug Administration (FDA) approval in 2011. See Hodi et al., Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. (2010) Aug. 19; 363(8):711-23. In some embodiments, the one or more checkpoint inhibitors can be an anti-PD-L1 antibody. In some embodiments, the anti-PD-L1 antibody can be a full length atezolizumab (anti-PD-L1), avelumab (anti-PD-L1), durvalumab (anti-PD-L1), or a fragment or a variant thereof. In some embodiments, the one or more checkpoint inhibitors can be any one or more of CD27 inhibitor, CD28 inhibitor, CD40 inhibitor, CD122 inhibitor, CD137 inhibitor, OX40 (also known as CD134) inhibitor, GITR inhibitor, ICOS inhibitor, or any combination thereof. In some embodiments, the one or more checkpoint inhibitors can be any one or more of A2AR inhibitor, B7-H3 (also known as CD276) inhibitor, B7-H4 (also known as VTCN1) inhibitor, BTLA inhibitor, IDO inhibitor, KIR inhibitor, LAG3 inhibitor, TIM-3 inhibitor, VISTA inhibitor, or any combination thereof.


In certain embodiments, an additional therapeutic agent comprises a second immunotherapeutic agent. In some embodiments, the additional immunotherapeutic agent includes, but is not limited to, a colony stimulating factor, an interleukin, an antibody that blocks immunosuppressive functions (e.g., an anti-CTLA-4 antibody, anti-CD28 antibody, anti-CD3 antibody, anti-PD-L1 antibody, anti-TIGIT antibody), an antibody that enhances immune cell functions (e.g., an anti-GITR antibody, an anti-OX-40 antibody, an anti-CD40 antibody, or an anti-4-1BB antibody), a toll-like receptor (e.g., TLR4, TLR7, TLR9), a soluble ligand (e.g., GITRL, GITRL-Fc, OX-40L, OX-40L-Fc, CD40L, CD40L-Fc, 4-1BB ligand, or 4-1BB ligand-Fc), or a member of the B7 family (e.g., CD80, CD86). In some embodiments, the additional immunotherapeutic agent targets CTLA-4, CD28, CD3, PD-L1, TIGIT, GITR, OX-40, CD-40, or 4-1BB.


In some embodiments, the additional therapeutic agent is an additional immune checkpoint inhibitor. As used herein, “an immune checkpoint inhibitor” is an agent that inhibits the activity of an immune checkpoint protein. In some embodiments, the additional immune checkpoint inhibitor is an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-CD28 antibody, an anti-TIGIT antibody, an anti-LAG3 antibody, an anti-TIM3 antibody, an anti-GITR antibody, an anti-4-1BB antibody, or an anti-OX-40 antibody. In some embodiments, the additional therapeutic agent is an anti-TIGIT antibody. In some embodiments, the additional therapeutic agent is an anti-PD-L1 antibody selected from the group consisting of: BMS935559 (MDX-1105), atexolizumab (MPDL3280A), durvalumab (MEDI4736), and avelumab (MSB0010718C). In some embodiments, the additional therapeutic agent is an anti-CTLA-4 antibody selected from the group consisting of: ipilimumab (YERVOY) and tremelimumab. In some embodiments, the additional therapeutic agent is an anti-LAG-3 antibody selected from the group consisting of: BMS-986016 and LAG525. In some embodiments, the additional therapeutic agent is an anti-OX-40 antibody selected from the group consisting of: MEDI6469, MEDI0562, and MOXR0916. In some embodiments, the additional therapeutic agent is an anti-4-1BB antibody selected from the group consisting of: PF-05082566. In some embodiments, the modified immune effector cells as described herein can be administered in combination with a biologic molecule selected from the group consisting of: cytokines, adrenomedullin (AM), angiopoietin (Ang), BMPs, BDNF, EGF, erythropoietin (EPO), FGF, GDNF, granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), stem cell factor (SCF), GDF9, HGF, HDGF, IGF, migration-stimulating factor, myostatin (GDF-8), NGF, neurotrophins, PDGF, thrombopoietin, TGF-α, TGF-β, TNF-α, VEGF, P1GF, gamma-IFN, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-12, IL-15, and IL-18.


XVI. Personalized Therapy

In an embodiment, the invention involves the detection of a disease or disorder associated with the overexpression of an antigen in a subject. The method of detection comprises: a) contacting a sample from a subject with one or more of the antibodies, or antigen-binding fragments thereof, that are described herein; and b) detecting an increased level of binding of the antibody or fragment thereof to the sample as compared to such binding to a control sample lacking the disease, thereby detecting the disease in the subject. In certain embodiments, the disease is cancer. In certain such embodiments, the cancer is selected from the group of ovarian cancer and breast cancer. While not intending to limit the method of detection, in one embodiment detection of binding is immunohistochemical, for example by enzyme-linked immunosorbent assay (ELISA), fluorescence-activated cell sorting (FACS), Western blot, immunoprecipitation, and/or radiographic imaging.


In an embodiment of the invention, a collection approach is used to transform the personalized cell therapy landscape for patients, including cancer patients. The approach includes the use of a developed and validated collection of non-viral plasmids to target tumor-associated antigens. These design and manufacturing advantages include the use of UltraCAR-T™ technology for generation of genetically modified cells, coupled with the capabilities of the UltraPorator™ electroporation system. These methods, compounds and compositions allow therapies and treatment options to assist cancer centers and physicians deliver personalized, autologous genetically modified cell treatments with overnight manufacturing to cancer patients. Based on the patient's cancer indication and/or biomarker profile, one or more non-viral plasmids may be selected from the library to build a personalized UltraCAR-T cell treatment. After initial treatment, this approach has potential to allow re-dosing of UltraCAR-T cells targeting the same or new tumor antigen(s) based on the treatment response and the changes in antigen expression of the patient's tumor. The combination of the advanced UltraVector™ DNA construction platform and the ease of overnight manufacturing gives this library approach an advantage over traditional T-cell therapies.


Provided herein is a method for the treatment of a disease or disorder comprising the serial administration of polynucleotides encoding a chimeric antigen receptor or a cell comprising the same. The disease or disorder may, for example, be a cancer or auto-immune disease or disorders. The polynucleotides may be present in a viral or non-viral vector. The chimeric antigen receptors selected from a collection of chimeric antigen receptors having different structural compositions and binding specificities for an array of antigen targets.


In certain embodiments, the collection of chimeric antigen receptors comprises a chimeric antigen receptor that targets at least one of the following antigens: BCMA, CAIX, CA125, CCR4, CD3, CD4, CD5, CD7, CD16, CD19, CD20, CD22, CD24, CD25, CD28, CD30, CD33, CD38, CD40, CD44, CD44v6, CD44v7/v8, CD47, CD52, CD56, CD70, CD79b, CD80, CD81, CD86, CD123, CD133, CD138, CD151, CD171, CD174, CD276, CEA, CEACAM6, CLL-1, c-MET, CS1, CSPG4, CTLA-4, DLL3, EDB-F, EGFR, EGFR2, EGFRvIII, EGP-2, EGP-40, EphA2, FAP, FLT1, FLT4, Folate-binding Protein, Folate Receptor, Folate receptor α, α-Folate receptor, Frizzled, GD2, GD3, GHR, GHRHR, GITR, GPC3, Gp100, gp130, HBV antigens, HER1, HER2, HER3, HER4, h5T4, HVEM, IGF1R, IgKAppa, IL-1-RAP, IL-2R, IL6R, IL-11Rα, IL-13R-a2, KDR, KRASG12V, LewisA, LewisY, L1-CAM, LIFRP, LRP5, LTPR, MAGE-A, MAGE-A1, MAGE-A10, MAGE-A3, MAGEA3/A6, MAGE-A4, MAGE-A6, MART-1, MCAM, mesothelin, PSCA, Mucins such as MUC1, MUC4 or MUC16, NGFR, NKG2D, Notch-1-4, NY-ESO-1, O-acetylGD2, O-acetylGD3, OX40, P53, PD1, PDE10A, PD-L1, PD-L2, PRAME, PSMA, PTCH1, RANK, Robol, ROR1, ROR1R, ROR-2, TACI, TAG-72, TCRα, TCRp, TGF, TGFBeta, TGFBeta-II, TGFBR1, TGFBR2, Titin, TLR7, TLR9, TNFR1, TNFR2, TNFRSF4, TRBC1, TWEAK-R, VEGF, VEGF-R2, and WT-1. In certain embodiments, the collection of chimeric antigen receptors comprises a chimeric antigen receptor that targets at least one of the following antigens: B7H4, BCMA, BTLA, CA125, CAIX, CCR4, CD123, CD133, CD137, CD138, CD151, CD16, CD171, CD174, CD19, CD20, CD22, CD24, CD25, CD276, CD28, CD3, CD30, CD33, CD38, CD4, CD40, CD44, CD44v6, CD44v7/v8, CD47, CD5, CD52, CD56, CD7, CD70, CD79b, CD80, CD81, CD86, CEA, CEACAM6, CLL-1, c-MET, CS1, CSPG4, CTLA-4, DLL3, EDB-F, EGFR, EGFR2, EGFRvIII, EGP-2, EGP-40, EphA2, HER1, HER2, HER3, HER4, FAP, FLT1, FLT4, Folate receptor α, Folate-binding protein, folate receptor, Frizzled, GD2, GD3, GHR, GHRHR, GITR, Gp100, gp130, GPC3, h5T4, HBV antigens, HER1/HER3, HPV antigens, HVEM, IGF1R, IgKAppa, IL-11Rα, IL-13R-a2, IL-1-RAP, IL-2R, IL6R, KDR, KRASG12V, L1-CAM, LewisA, LewisY, LIFRP, LRP5, LTPR, MAGE-A, MAGE-A1, MAGE-A10, MAGE-A3, MAGEA3/A6, MAGE-A4, MAGE-A6, MART-1, MCAM, Mesothelin, mucins such as MUC1 and MUC16, NGFR, NKG2D, Notch 1-4, NY-ESO-1, O-acetylGD2, O-acetylGD3, OX40, P53, PD1, PDE10A, PD-L1, PD-L2, PMSA, PRAME, PSCA, PSMA, PTCH1, RANK, Robol, ROR1, ROR1R, ROR-2, TACI, TAG-72, TCRα, TCRp, TGF, TGFβ, TGFβ-II, TGFBR1, TGFBR2, Titin, TLR7, TLR9, TNFR1, TNFR2, TNFRSF4, TRBC1, TWEAK-R, VEGF, VEGF-R2, and WT-1.


In certain embodiments, the method comprises a first administration of cells expressing one or more chimeric antigen receptors from the collection followed by a second administration of cells expressing one or more chimeric antigen receptors from the collection, wherein a period of time elapses between the first and second administrations. In certain embodiments, the same one or more CARs in the first administration are administered again. In certain other embodiments, at least one of the CARs expressed by cells in the second administration is different from CARs in the first administration. In certain embodiments, the invention comprises a third, fourth, fifth, sixth, seventh, eighth, ninth, tenth or any additional number of rounds of administration of cells expressing CARs selected from the collection, wherein in each subsequent round of administration a different CAR is administered which was not administered in a previous round of treatment.


In certain embodiments, the dose of the cells is autologous. In certain embodiments, the dose of cells is allogenic. In certain embodiments, one dose of the cells is autologous and another dose is allogenic.


In certain embodiments, prior to a second or subsequent administration of a CAR or CARs, a period of time is allowed to elapse that is sufficient for biologic or therapeutic activity of one or more CARs in a preceding administration to become diminished from peak biologic or therapeutic activity, become negligible or become undetectable. In certain embodiments, a subsequent administration of a CAR takes place at least one day following the previous administration of a CAR.


The polynucleotide encoding the CAR may be introduced into the cells of a subject by way of viral transduction, non-viral transfection or electroporation methods of delivery.


In certain embodiments, the polynucleotide encoding the CAR may be administered as part of a vector, such as those described herein. In certain embodiments of the invention, one or more polynucleotides encoding a CAR further comprises nucleotide sequences encoding a cytokine, a cell tag, and/or a gene switch, as described previously. In certain embodiments, the vector comprises the polynucleotide encoding the CAR and one or more polynucleotides encoding the cytokine, cell tag, and/or a gene switch.


In certain embodiments, the vector is a “regulatory approved vector,” meaning that it has been approved by a Regulatory Authority, national, supra-national (e.g., the U.S. Food and Drug Administration (FDA), the European Commission or the Council of the EU), regional, state or local regulatory agency, department, bureau, commission, council or other governmental entity, wherein such approval is necessary or sufficient for the manufacture, distribution, use or sale of a vector in a regulatory jurisdiction. In certain embodiments, such a “regulatory approved vector” means a vector which has been approved, at a minimum, for use in human clinical safety trials (sometimes referred to as “phase 1 clinical trial”) in a regulatory jurisdiction. “Clinical safety” or “Phase 1” trial means a trial to assess at least safety, optionally to assess safety and dosages, for analysis of side effects associated with the test article, optionally to assess side effects in conjunction with varying dosages. In certain embodiments, “regulatory approved vector” means a vector which has been approved, at a minimum, for use in “Phase 2” studies. A “Phase 2” study is one in which the test article is administered to a larger group human subjects (as compared to a smaller Phase 1 group of subjects) for evaluation of a larger group of patients (typically up to a few hundred) with a disease, disorder or condition for which the test article is developed, to initially assess its effectiveness and to further study its safety. In certain embodiments, a Phase 2 study is to assess the optimal dose or doses of a test article to maximize benefits, while minimizing risks. In certain embodiments, “regulatory approved vector” means a vector which has been approved, at a minimum, for use in “Phase 3” studies. A Phase 3 study (sometimes referred to as “pivotal trials”) typically involves about 300 to 3,000 participants from a patient population for which the test article is intended to be used. Participants in a Phase 3 study are typically assigned to receive either the test article or a placebo (a substance that has no therapeutic effect). A Phase 3 study is intended to demonstrate whether or not a test article offers a treatment benefit to a specific population and to provide more detailed safety data, and to serve as the basis for product labeling in regard to diseases or conditions for which the test article may be used. In certain embodiments, “regulatory approved vector” means a vector which has been approved for commercial manufacturing, use or sale for treatment of a disease, disorder or condition in humans by a regulatory authority in the regulatory jurisdiction (e.g., approval by U.S. FDA for manufacturing, use or sale in the United States of America). In certain embodiments, the “regulatory approved vector” is a regulatory approved lentivirus vector, a regulatory approved retroviral vector, or a regulatory approved non-viral vector. In some embodiments, the regulatory approved vector is a non-viral vector that is a Sleeping Beauty vector.


In certain embodiments, the regulatory authority governs approval of pharmaceuticals, biologics, or other medicines or medical treatments in a country selected from the United Arab Emirates, Antigua and Barbuda, Albania, Armenia, Angola, Austria, Australia, Azerbaijan, Bosnia and Herzegovina, Barbados, Belgium, Burkina Faso, Bulgaria, Bahrain, Benin, Brunei Darussalam, Brazil, Botswana, Belarus, Belize, Canada, Central African Republic, Congo, Switzerland, Cote d'Ivoire, Chile, Cameroon, China (People's Republic of China (PCR)), Colombia, Costa Rica, Cuba, Cyprus, Czech, Germany, Djibouti, Denmark, Dominica, Dominican Republic, Algeria, Ecuador, Estonia, Egypt, Spain, Finland, France, Gabon, United Kingdom, Grenada, Georgia, Ghana, Gambia, Guinea, Equatorial Guinea, Greece, Guatemala, Guinea-Bissau, Honduras, Croatia, Hungary, Indonesia, Ireland, Israel, India, Iran (Islamic Republic of), Iceland, Italy, Jordan, Japan, Kenya, Kyrgyzstan, Cambodia, Comoros, Saint Kitts and Nevis, Democratic People's Republic of Korea, Republic of Korea, Kuwait, Kazakhstan, Lao People's Democratic Republic, Saint Lucia, Liechtenstein, Sri Lanka, Liberia, Lesotho, Lithuania, Luxembourg, Latvia, Libya, Morocco, Monaco, Republic of Moldova, Montenegro, Madagascar, North Macedonia, Mali, Mongolia, Mauritania, Malta, Malawi, Mexico, Malaysia, Mozambique, Namibia, Niger, Nigeria, Nicaragua, Netherlands, Norway, New Zealand, Oman, Panama, Peru, Papua New Guinea, Philippines, Poland, Portugal, Qatar, Romania, Serbia, Russian Federation, Rwanda, Saudi Arabia, Seychelles, Sudan, Sweden, Singapore, Slovenia, Slovakia, Sierra Leone, San Marino, Senegal, Sao Tome and Principe, El Salvador, Syrian Arab Republic, Eswatini, Chad, Togo, Thailand, Tajikistan, Turkmenistan, Tunisia, Turkey, Trinidad and Tobago, United Republic of Tanzania, Ukraine, Uganda, United States of America, Uzbekistan, Saint Vincent and the Genadines, Viet Nam, Samoa, South Africa, Zambia, Zimbabwe, Afghanistan, Andorra, ARIPO, Bangladesh, Bolivia, Cayman Islands, Ethiopia, Fiji, Gibraltar, Guam, Haiti, Iraq, Lebanon, Martinique, Micronesia, federated states of, Nauru, OAPI, Palestinian territory, occupied (Gaza), Pitcairn, Saint Barthélemy, Somalia, Taiwan, Tonga, Vanuatu, Yemen, Åland Islands, Anguilla, Aruba, Bermuda, Burundi, Congo (Democratic Republic of the), Eurasian Patent Organization, French Guiana, Greenland, Guernsey, Holy see (Vatican), Jamaica, Macau, Mauritius, Montserrat, Nepal, Pakistan, Palestinian territory, occupied (West Bank), Puerto Rico, Saint Helena, South Sudan, Timor-Leste, Tuvalu, Venezuela, American Samoa, Argentina, Bahamas, Bhutan, Cape Verde, Eritrea, European Union, French Polynesia, Guadeloupe, Guyana, Hong Kong, Kiribati, Maldives, Mayotte, Myanmar, New Caledonia, Palau, Paraguay, Réunion, Saint Pierre and Miquelon, Suriname, Tokelau, Uruguay, and Western Sahara.


In certain embodiments, the regulatory authority is selected from a multinational organization, such as the World Health Organization, the Pan-American Health Organization, the World Trade Organization (WTO), the International Conference on Harmonization (ICH), and the World Intellectual Property Organization (WIPO); and a national health authority, such as, in Asia and the Pacific, the Australian Government: Department of Health and Ageing, Australian Government: Therapeutic Goods Administration (TGA), Brunei: Ministry of Health, People's Republic of China: State Food and Drug Administration, People's Republic of China: Ministry of Health, People's Republic of China: National Medical Products Administration, People's Republic of China: National Institute for the Control of Pharmaceutical and Biological Products, People's Republic of China: Ministry of Agriculture, Fiji: Ministry of Health, Hong Kong: Department of Health, India: Ministry of Consumer Affairs, Food & Public Distribution, India: Central Drug Standards Control Organization (CDSCO), India: Ministry of Food and Consumer Affairs, Indonesia: Ministry of Health, Japan: Ministry of Health, Labor and Welfare, Japan: Pharmaceuticals and Medical Devices Evaluation Agency, Korea: Food and Drug Administration, Malaysia: Ministry of Health, Malaysia: National Pharmaceutical Regulatory Agency, New Zealand: Ministry of Health, New Zealand: Medicines and Medical Devices Safety Authority, New Zealand: Food Safety Authority, Papua New Guinea: Department of Health, Philippines: Department of Health, Philippines: National Food Authority, Singapore: Ministry of Health, Singapore: Health Sciences Authority, Sri Lanka: Ministry of Health, Taiwan: Department of Health, Taiwan: National Laboratories of Foods and Drugs, Thailand: Ministry of Public Health, Thailand: Food and Drug Administration, Thailand: Ministry of Agriculture and Co-operatives, in Europe, the European Medicines Agency (EMA), European Commission Directorate General: Medicinal Products for Veterinary Use, Andorra: Ministry of Health and Welfare, Armenia: Ministry of Health, Armenia: Drug and Medical Technology Agency, Austria: Secretariat of Health, Belgium: Health, Food Chain Safety and Environment, Belgium: Pharmaceutical Inspectorate, Belgium: Federal Agency for the Safety of the Food Chain, Bulgaria: Ministry of Health, Bulgaria: Drug Agency, Croatia: Ministry of Health and Social Care, Czech Republic: Ministry of Health, Czech Republic: State Institute for Drug Control, Denmark: Ministry of Health, Denmark: Danish Medicines Agency, Denmark: Veterinary and Food Administration, Estonia: State Agency of Medicines, Finland: Ministry of Social Affairs and Health, Finnish Medicines Agency, France: Ministry of Health, France: National Agency for Veterinary Medicinal Products, Georgia: Ministry of Labor, Health and Social Affairs, Germany: Ministry of Health, Germany: Federal Institute for Drugs and Medical Devices, Germany: Robert Koch Institute, Germany: Paul Ehrlich Institute, Germany: Federal Institute for Risk Assessment, Germany: Ministry of Consumer Protection, Food and Agriculture, Greece: Ministry of Health and Social Solidarity, Greece: National Organization for Medicines, Hungary: Ministry of Health, Social and Family Affairs, Hungary: National Institute of Pharmacy, Iceland: Ministry for Health and Social Security, Icelandic Medicines Agency, Iceland: The Environment Agency, Ireland: Department of Health and Children, Irish Medicines Board, Italy: Ministry of Health, Italy: National Institute of Health, Latvia: State Agency of Medicines, Lithuania: Ministry of Health, Lithuania: State Medicines Control Agency, Luxembourg: Ministry of Health, Malta: Ministry of Health, Elderly and Community Care, Netherlands: Ministry of Health, Welfare and Sport, Netherlands: Medicines Evaluation Board, Netherlands: Inspectorate for Health Protection and Veterinary Public Health, Norway: Ministry of Health and Care Services, Norway: Norwegian Board of Health Supervision, Norway: Norwegian Medicines Agency, Norway: Ministry of Agriculture and Food, Poland: Ministry of Health and Social Security, Poland: Drug Institute, Portugal: Ministry of Health, Portugal: National Authority of Medicines and Health Products, Romania: Ministry of Health and the Family, Russian Federation: Scientific Centre for Expert Evaluation of Medicinal Products, Russian Federation: State Institute of Drugs and Good Practices, San Marino, Ministry of Health and Social Security, National Insurance and Gender Equality (in Italian), Slovak Republic: Ministry of Health, Slovak Republic: State Institute for Drug Control, Slovak Republic: State Veterinary and Food Administration, Slovenia: Ministry of Health, Slovenia: Institute of Public Health, Spain: Ministry of Health and Consumption; Spanish Agency for Medicines and Health Products, Sweden: Medical Products Agency, Sweden: National Board of Health and Welfare, Sweden: National Food Administration, Switzerland: Federal Office of Public Health, Switzerland: Agency for Therapeutic Products, Switzerland: Federal Veterinary Office, Turkey: Ministry of Health (in Turkish), Ukraine: Ministry of Health, UK: Department of Health, UK: Health Protection Agency, UK: Medicines and Healthcare Products Regulatory Agency (MHRA), UK: National Institute for Biological Standards and Control, UK: Veterinary Medicines Directorate, in the Middle East, the Bahrain: Ministry of Health, Israel: Ministry of Health, Israel: Ministry of Industry, Trade and Labor, Jordan: Ministry of Health, Lebanon: Ministry of Public Health, Palestinian Authority: Ministry of Health, Saudi Arabia: Ministry of Health, United Arab Emirates: Ministry of Health, United Arab Emirates: Federal Department of Pharmacies, Yemen: Ministry of Public Health & Population, in Africa, the Benin: Ministry of Health, Botswana: Ministry of Health, Egypt: Ministry of Agriculture and Land Reclamation, Ghana: Ministry of Health, Ghana: Ministry of Food and Agriculture, Kenya: Ministry of Health, Maldives: Ministry of Health, Mauritius: Ministry of Health & Quality of Life, Mauritius: Ministry of Agro Industry and Food Security, Morocco: Ministry of Public Health, Namibia: Ministry of Health and Social Services, Senegal: Ministry of Health and Prevention, South Africa: Department of Health, Swaziland: Ministry of Health and Social Welfare, Tanzania: Ministry of Health, Tunisia: Ministry of Public Health, Tunisia: Office of Pharmacy and Medicines, Uganda: Ministry of Health, Zimbabwe: Ministry of Health and Child Welfare, and in the Americas, the Argentina: Ministry of Health, Argentina: National Administration of Drugs, Foods and Medical Technology, Belize: Ministry of Health, Bolivia: Ministry of Health and Social Welfare, Brazil: Ministry of Health, Brazil: National Health Surveillance Agency, Brazil: Fundacao Oswaldo Cruz, Canada: Health Canada, Canada: Health Products and Food Branch, Chile: Health Ministry, Chile: Institute of Public Health, Colombia: Ministry of Health, Colombia: INVIMA Instituto Nacional de Vigilancia de Medicamentos y Alimentos, Costa Rica: Ministry of Health, Dominican Republic: Ministry of Agriculture, Ecuador: Ministry of Public Health, El Salvador: Ministry of Public Health and Social Assistance, El Salvador: Ministry of Agriculture, Guatemala: Ministry of Health, Guyana: Ministry of Health, Guyana: National Bureau of Standards, Jamaica: Ministry of Health, Mexico: Ministry of Health, Mexico: Federal Commission for the Protection Against Sanitary Risks, Netherlands Antilles: Department of Public Health and Environmental Protection, Nicaragua: Ministry of Health, Panama: Ministry of Health, Peru: Ministry of Health, Peru: General Directorate of Medicines, Supplies and Drugs, St. Lucia: Ministry of Agriculture, Lands Forestry and Fisheries, Trinidad and Tobago: Ministry of Health, Trinidad & Tobago: Bureau of Standards, United States of America (USA/U.S.): Food and Drug Administration (FDA), Uruguay: Ministry of Public Health, and the Venezuela: Ministry of Health and Social Development. Other exemplary regulatory authorities can also be found at http://iaocr.com/clinical-research-regulations/regulatory-authority-links/ and https://www.pda.org/scientific-and-regulatory-resources/global-regulatory-authority-websites.


In certain embodiments, cells expressing one or more CARs are isolated for continued ex vivo expansion and/or are cryopreserved for future use.


The present invention also relates to a composition comprising a chimeric antigen receptor as described above.


The present invention further relates to a kit comprising a first vector encoding a CAR as described above and a second vector encoding a CAR as described above.


EXAMPLES

These Examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein. The following table includes abbreviations and special terms that apply to the Examples only. These abbreviations and special terms are not otherwise limiting, and neither replace nor narrow the broader definitions set forth above, which shall continue to apply to the claims.









TABLE 9







Abbreviations and Special Terms for Use in the Examples








Abbreviation of



Special Term
Explanation





Artificial
A non-naturally occurring miRNA designed to target


miRNA
a specific gene


CAR-T
Chimeric Antigen Receptor T Cell


Guide miRNA
The mature miRNA processed from the pre-miRNA that



is complementary to an intended target gene


LFC
Log2 fold change


Mature miRNA
The fully processed 21-23nt miRNA


mbIL15
A fusion protein comprising IL-15 and IL-15Rα,



the protein comprising the amino acid sequence of



SEQ ID NO: 525


miRNA
microRNA, a small RNA species generally 21-23nt



in length that is complementary to a target mRNA



transcript and reduces expression of the target



transcript through either RNA degradation or



inhibition of translation


Passenger
A mature miRNA that may be processed from the side


miRNA
opposite of the guide miRNA in the RNA stem-loop


PD1 Silencer
A genetic module encoded within the VVN-5351 trans-


Module
gene cassette designed to reduce expression of PD1



within ROR1 + PD1 silencer cells. The module



consists of a splice unit containing a non-coding



RNA that is processed by internal cellular machinery



into 2 unique microRNAs with homology to PD1 mRNA.


Pre-miRNA
Precursor miRNA, which is the intermediate miRNA



species that has undergone the first processing step



to remove the RNA stem-loop structure from the primary



mRNA transcript. Pre-miRNA undergoes a second pro-



cessing step in which the mature miRNA is cleaved and



loaded into the RNA-induced silencing complex (RISC).


R0R1 + PD1
ROR1 CAR-T cells expressing mbIL15 and HER1t with


silencer cells
anti-PD-1 miRNAs


Pri-miRNA/
The primary transcript encoding RNA stem-loop struc-


pri-miRNA
tures that are processed by the endogenous cellular


scaffold
machinery into precursor and then mature miRNA. The



artificial pri-miRNA sequence and structures are



based on endogenous human pri-miRNAs, herein referred



to as a scaffold, with the natural guide and passenger



strands replaced with a guide miRNA to target PD1



and a passenger miRNA to maintain the predicted



folded RNA structure.


UTR
Untranslated region


VVN-5351
A DNA transposon plasmid encoding the ROR1



UltraCAR-T transgene cassette plus a dual PD1-



targeting miRNA in the 5′UTR encoded within a



synthetic splice unit.


VVN-5355
A DNA transposon plasmid encoding the ROR1



UltraCAR-T transgene cassette.









Example 1. PD1 Module Design

The PD1-silencing module of miRNA-expressing ROR1 UltraCAR-T cells encodes two artificial miRNAs designed to specifically reduce expression of PD-1 mRNA within UltraCAR-T cells while avoiding off-target silencing of other endogenous transcripts. The two artificial miRNAs of miRNA-expressing ROR1 UltraCAR-T cells are encoded within a dual primary miRNA (pri-miRNA) sequence placed within a 5′ UTR splice unit of the UltraCAR-T transgene cassette (FIG. 1B). The dual pri-miRNA forms stem-loop structures that are recognized and processed by cellular complexes to generate two unique 21-24 nucleotide mature guide miRNAs that are homologous to specific sequences within the PD1 target transcript. Interaction of the guide miRNAs with the PD1 target sequence is expected to trigger the silencing of PD1 by induction of RNA degradation or translational inhibition (Guo et al, 2010).


The guide miRNAs encoded within miRNA-expressing ROR1 UltraCAR-T cells were designed to be highly specific for the PD1 target transcript NM_005018.3 by implementation of an internally-designed computational workflow using a combination of twenty-one ranking parameters based on three validated rules-based siRNA prediction algorithms (Amarzguioui and Prydz, 2004; Reynolds et al, 2004; Ui-Tei et al, 2004). Multi-level specificity profiling was performed against the human reference exome (RefSeq) and the activated T-cell transcriptome (Zhao et al, 2014) to ensure that the mature miRNAs are highly specific for the PD1 target gene. To further reduce risk of off-target silencing by the non-PD1 targeting passenger strand miRNAs, pri-miRNA scaffolds were selected that produce a high ratio of guide:passenger miRNA (Miniarikova et al, 2016). The PD1-targeting guide miRNA PD1_1843 was incorporated into a pri-miRNA scaffold based on human miRNA hsa-miR-204 (accession #MI0000284) while the PD1_2061 guide miRNA was incorporated into the hsa-miR-206 (accession #MI0000490) scaffold. Mutations were created on the passenger strand side of each pri-miRNA to assure the specific miRNA structure was maintained and that thermodynamic stability was not substantially altered. RNA structures were predicted using CLC Main Workbench software. The PD1 silencing module contains the miR204 PD1_1843 pri-miRNA placed directly upstream of miR206 PD1_2061 pri-miRNA within a synthetic splice unit in the 5′ UTR of the CAR-T transgene expression cassette. The splice units were ordered as gBlock from IDT and can be cloned into Sleeping Beauty CAR vectors and can be cut using ClaI/NheI for cloning into the 5′UTR of any other Sleeping Beauty CAR vector.


Example 2. Reduction of PD1 Transcript Expression by miRNAs

Primary human T cells were transfected with the constructs listed in Table 10 and expanded in vitro with the use of AaPC cells expressing cognate antigen that were grown in large batches and then frozen into aliquots. The generated CAR-T cells were then further activated using anti-CD3/anti-CD28 beads at a bead:T cell ratio of 1:1 and 1×106 T cells/mL for 48 hrs prior to harvesting the cells for RNA isolation. RNA isolation was performed according to manufacturer's recommended protocol (Qiagen) then subjected to RT-qPCR analysis using the SuperScript VILO Master Mix with ezDNase (Invitrogen) and TaqMan FAST Advanced master mix for qPCR to evaluate PD-1 expression levels using specific primer/probes (Human PD1: Hs00169472_m1; Human TIGIT: Hs00545087_m1; and Human ACTb: Hs99999903_m1). All samples were also normalized to beta-actin expression levels. The relative expression values are based upon MET method by normalizing to construct #1 (MUC16 CAR-T cells only). Results are shown in FIG. 2. Shown is the mean±SD from 3 donors.









TABLE 10







Description of constructs 1-8 as utilized in FIG.


2 sequences of miRNA are as described in Table 4)









Construct




#
miRNA
Effector Genes





1
N/A
MUC16 CAR-mbIL15-HER1t


2
Scrambled 1 (SEQ ID NO:
MUC16 CAR-mbIL15-HER1t



586)


3
Scrambled 2 (SEQ ID NO:
MUC16 CAR-mbIL15-HER1t



582)


4
miR204 PD1 + miR206
MUC16 CAR-mbIL15-HER1t



PD1


5
miR17 TIGIT
MUC16 CAR-mbIL15-HER1t


6
miR150 TIGIT + miR206
MUC16 CAR-mbIL15-HER1t



PD1


7
miR204 PD1 + miR206
MUC16 CAR-mbIL15-HER1t



PD1 + miR17 TIGIT


8
N/A
MUC16 CAR + HER1t









The data demonstrates the specificity of PD-1 checkpoint inhibitor miRNA targeting the intended sequence. CAR constructs that expressed the scrambled miRNA (Constructs 2 and 3) along with the CAR construct that did not express mbIL-15 (construct #8) did not show any decrease in PD-1 expression whereas CAR constructs that contain PD-1 miRNA (construct 4) or a combination of PD-1 and TIGIT miRNA (constructs 6 and 7) did show a decrease in PD-1 expression. On the other hand, the CAR construct containing only a TIGIT miRNA sequence (construct 5) did not show any decrease in PD-1 mRNA expression levels further demonstrating the specificity in targeting.


Example 3. Downregulation of Targeted mRNA

Primary human T cells were transfected with a vector encoding CD33-specific CAR or a vector encoding MUC16-specific CAR. The vectors comprise a synthetic intron containing miRNA sequences that target PD-1, PD-1 and/or TIGIT or a non-targeting scrambled control miRNA. T cell cultures were in vitro expanded using antigen presenting cells with cognate tumor antigen that were K562 cells modified to express either CD33 or MUC16 plus other co-stimulatory molecules (based on “Clone 1”) and at a ratio of 1:1 (AaPC:T cell). The generated CAR+ cells were then stimulated with anti-CD3/anti-CD28 beads (1:1 bead:T cell ratio) in the absence of cytokines for 48 hrs. RNA was isolated using Qiagen kits (AllPrep Universal DNA/RNA/miRNA kit #80224) as per manufacturer's protocol and utilized for the Human PanCancer Immune gene set panel Kit from Nanostring. Briefly, the RNA was hybridized with capture and reporter probe sets and samples processed then hybridized to the slide using the nCounter Prep Station and transcript counts were generated by the nCounter Digital Analyzer according to the manufacturer's protocol. Data validation, QC and normalization were performed by nSolver software (Nanostring Technologies).


The distribution of the transcript count data on the graph would be a line (from lower left to top right corner) with the slope of 1 if the samples were identical. Data points falling off this line represent variations in transcript counts between the two samples. Counts that were found below the main line represent reduced expression whereas counts above the main line represent increased expression to the sample being compared. The transcript targeted by the miRNA, either PD-1 or TIGIT expressed in the CAR-T cells, specifically showed lower expression of the transcripts of their respective targeted genes compared to the CAR-T cells without the miRNA. See FIG. 3A-B. In addition, to further demonstrate the specificity of targeting, the CAR-T cells with the scrambled control miRNA had no alterations to the transcript levels of either PD-1 or TIGIT, as the data distribution in this graph remained close to the diagonal indicating very little variation in transcript levels. See FIG. 3C. Similar results were observed in T cells transfected with the MUC16 CAR vector comprising a synthetic intron containing miRNA sequences that target PD-1, PD-1 and/or TIGIT (see FIG. 4A-C).


Example 4. Enhancement of Tumor Cell Cytotoxicity Effect of miRNA MUC16 CAR-T Cells

Primary human T cells were transfected with a vector encoding MUC16-specific CAR and miRNA sequences that target two sequences within the PD-1 transcript, or with a vector that encodes the MUC16-specific CAR but not an miRNA. CAR-T cells used in this assay were in vitro expanded, normalized for CAR expression then seeded in triplicates with GFP+ K562 cells expressing MUC16 (“tumor cells”) in 96 well plates at the 3:1 E:T ratios. The plate was loaded into the IncuCyteS3 instrument and 4 images per well were taken every 2 hr for 7 days. The IncuCyte Software was used to analyze the data and normalize the GFP+ cell counts/image to the 0 hr time point.


The outgrowth assay determines the rate of target cell growth in the presence or absence of CAR-T cells over the course of 7 days in culture. The lower target cell count in cultures containing CAR-T cells indicate the CAR-T cytolytic activity. FIG. 5A depicts the difference between the tumor target cells only (black circle, filled) and the cells expressing the MUC16-specific CAR only (open square), demonstrating the killing capacity of the MUC16 CAR-T cells. The cells further expressing miRNA targeting PD-1 (grey circle filled) further demonstrates improved cytolytic activity, based upon the sustained low GFP+ counts over the time course evaluation, compared to the cells expressing MUC16 specific CAR only (open square). This data demonstrates the enhanced cytolytic activity of CAR-T cells containing PD-1 targeting miRNA.


A similar experiment was conducted utilizing GFP+ K562 cells expressing MUC16, PD-L1, and CD155. As shown in FIG. 5B, the difference between the tumor target cells only (square, filled) and cells expressing the CAR only (circle, open), demonstrates the killing capacity of the MUC16 CAR-T cells. The CAR-T cells incorporating miRNAs targeting both PD-1 and TIGIT (circle, filled) further demonstrates improved cytolytic activity, based upon the sustained low GFP+ counts over the time course evaluation, compared to the MUC16 CAR-T cells only (circle, filled). This data demonstrates the enhanced cytolytic activity of CAR-T cells containing 3 targeting miRNAs (2 for PD-1 and 1 for TIGIT) in a single construct.


Example 5. Improvement of Cytokine Expression

Primary human T cells were transfected with vectors encoding MUC16-specific CAR but not miRNA targeting sequences, and vectors encoding MUC16-specific CAR and single or combinations of miRNA targeting sequences of PD-1 and TIGIT (see Table 11). In vitro expanded CAR-T cells were in vitro expanded, normalized for CAR expression then seeded in triplicates in 96 well plates with K562 tumor target cells expressing truncated MUC16 9MUC16t) at an effector to target ratio of 1:1 or the CAR-T cells were cultured in media alone. Culture supernatants were collected after co-culturing for 3 days. Culture supernatants were collected and interferon-gamma (IFNγ) and granulocyte/macrophage-colony stimulating factor (GM-CSF) was assessed by multiplex cytokine analysis (Luminex) according to manufacturer's protocol. Shown in FIG. 6 is the mean±SD from duplicate wells.


For the CAR-T cell constructs cultured in media only had basal levels of IFNγ and GM-CSF detected. No cytokines were observed from the use of target cells or media only. The supernatants following co-culture of MUC16 CAR-T cells only (Vector 1) with the tumor cells provided a baseline value for expression levels of IFNγ and GM-CSF. With the inclusion of checkpoint inhibitor miRNA into the CAR constructs, improved cytokine expression may be observed, particularly through inhibition via the PD-1 pathway. In the constructs that contained dual PD-1 (Construct #3) or the combination of a single PD-1 and TIGIT miRNA (Construct #6) or dual PD-1 and a TIGIT miRNA (Constructs #10 and 11) provided higher levels of cytokine expression.









TABLE 11







Description of Constructs #1-11 in FIGS. 6A-D


(sequences of miRNA are as described in Table 4)









Constructs
miRNA
Effector genes












1
N/A
MUC16 CAR-mbIL15-HER1t


2
Scrambled control 1
MUC16 CAR-mbIL15-HER1t


3
miR204 PD1 + miR206 PD1
MUC16 CAR-mbIL15-HER1t


4
miR17 TIGIT
MUC16 CAR-mbIL15-HER1t


5
miR17 TIGIT + miR206
MUC16 CAR-mbIL15-HER1t



PD1


6
miR150 TIGIT + miR206
MUC16 CAR-mbIL15-HER1t



PD1


7
miR17 TIGIT + miR204
MUC16 CAR-mbIL15-HER1t



PD1 + miR206 PD1


8
miR17 TIGIT + miR204
MUC16 CAR-mbIL15-HER1t



PD1 + miR206 PD1extended



v1


9
miR17 TIGIT + miR204
MUC16 CAR-mbIL15-HER1t



PD1 + miR206 PD1extended



v2


10
miR204 PD1 + miR206
MUC16 CAR-mbIL15-HER1t



PD1 + miR17 TIGIT


11
miR204 PD1 + miR206
MUC16 CAR-mbIL15-HER1t



PD1 + miR17 TIGITextended



v1









Example 6. Tumor Burden in Treated Mice

Non-obese diabetic/severe combined immunodeficiency (NOD/SCID) gamma mice (NSG) mice were intraperitoneally implanted with fLUC-GFP+SK-OV-3 tumor cells expressing MUCt on Day 0. Tumor burden was monitored in these mice throughout the study using an in vivo imaging system (IVIS) by luminescence with an IVIS Spectrum instrument (Perkin Elmer). IVIS data was analyzed using the Living Image Software (Version 4.1) based upon a defined region of interest to obtain total flux values (photons/sec). Prior to administration of the CAR-T cells, mice were randomized based upon tumor burden and body weight into the different groups then administered the test articles on Day 6 All CARs tested expressed the MUC16-specific CAR along with mbIL15 and HER1t and are referred to as MUC16 CAR. All test articles were normalized to a 0.5×106 CAR-T cells/mouse and administered intraperitoneally. IVIS imaging was performed twice/week to monitor overall tumor burden in the mice. Data shown is the mean±SEM from n=4-8 mice/group.


As shown in FIG. 7, mice given saline only (gray-filled circles), had continuous tumor growth as evidenced by the increasing total flux levels observed throughout the course of the study. Eventually, these mice succumbed to the tumor burden and were euthanized. Mice given the MUC16 CAR only (black-filled squares) were able to control tumor burden. CAR-T cells expressing the checkpoint inhibitor miRNA to PD-1 and TIGIT within the constructs (open squares and circles) were found to maintain anti-tumor activity, based upon the decrease in tumor flux values to background levels. In addition, the CAR-T cells expressing the checkpoint inhibitor miRNA to PD-1 and TIGIT showed a faster time frame and the rate of tumor burden decrease compared to the MUC16 CAR only construct.


Example 7. In Vivo Phenotyping Experiment

SKOV-3/MUC16 tumor bearing mice were administered CAR-T cells (expressing MUC16-specific CAR, mbIL15 and HER1t) of either CAR only or CAR with PD-1/PD-1 miRNA on Study Day 6. Whole blood from mice were taken on Study Day 31 for phenotypic evaluation by flow cytometry of the administered CAR-T cells. Briefly, cocktails of fluorescently conjugated antibodies were used to stain the whole blood samples then concurrently fixed along with red blood cells lysis using a one-step Fix/Lyse buffer. The fixed samples were read on the flow cytometer (BD LSRFortessaX-20) instrument. CAR-T cells were identified based upon gating of hCD45/CD3+/HER1t+ expression. In, FIG. 8A, the sample of CAR only (dotted line) shows high PD-1 expression, whereas the CAR with PD-1/PD-1 miRNA (solid line) shows a significant decrease in the level of PD-1 expression detected. To further quantify the reduced expression of PD-1 detected on the CAR+ miRNA (PD-1/PD-1) group, the median fluorescent intensity (MFI) was examined (FIG. 8B). The mean MFI of PD1 expression in mice given CAR-T cells (stripe bar) only was ˜709 whereas the mean MFI of the CAR-T cells with a PD-1/PD-1 miRNA (solid bar) was reduced down to ˜236. The mean±SEM from 5-8 mice is shown.


Example 8. Specific PD-1 and TIGIT Downregulation

SKOV-3 tumor bearing mice were administered CAR-T cells (expressing MUC16-specific CAR (“MUC16 CAR”), mbIL15 and HER1t) of either CAR only or CAR with different checkpoint miRNA inhibitors (PD-1 and TIGIT) on Study Day 6. See Table 12.









TABLE 12







Description of Groups #1-9 in FIGS. 9A-B








Group
CAR-T Cells





1
Saline control


2
MUC16 CAR/mbIL15/HER1t


3
MUC16 CAR/mbIL15/HER1t + anti-PD1


4
MUC16 CAR/mbIL15/HER1t + scrambled miRNA 2


5
MUC16 CAR/mbIL15/HER1t + miR206 PD1


6
MUC16 CAR/mbIL15/HER1t + miR17 TIGIT


7
MUC16 CAR/mbIL15/HER1t + miR204 PD1/miR206



PD1


8
MUC16 CAR/mbIL15/HER1t + miR150 TIGIT/miR206



PD-1


9
MUC16 CAR/mbIL15/HER1t + miR204 PD1/miR206



PD1/miR150 TIGIT









Whole blood from mice were taken on Study Day 45 (D45) for phenotypic evaluation by flow cytometry of the administered CAR-T cells. Briefly, cocktails of fluorescently conjugated antibodies, which included specific antibodies for human PD-1 and human TIGIT, were used to stain the whole blood samples then fixed using a one step Fix/Lyse buffer. The fixed samples were read on the flow cytometer (BD LSRFortessaX-20) instrument. CAR-T cells were identified in the mouse based upon gating of hCD45/CD3+/HER1t+ expression. To further evaluate the specificity of miRNA used in the CAR vector for the checkpoint inhibitor, the median fluorescent intensity (MFI) for expression of PD-1 and TIGIT was analyzed (FIGS. 9A and B). The MFI for PD-1 is shown on the left and the MFI for TIGIT is shown on the right for a quantitative assessment of the expression levels for the same set of vectors. As shown in FIG. 9A, reduced expression of PD-1 was seen on the CAR-T cells for the groups indicated with the down arrows (solid line for PD-1), which are constructs with a PD-1 miRNA (single, double and in combination with another miRNA checkpoint inhibitor), when compared to the CAR vector only. On the right side (downward dashed arrows) highlights cell populations with a reduced expression of TIGIT expression seen on the CAR-T cells. The downregulated expression of TIGIT corresponded to the samples that contained a miRNA for TIGIT (either as a single or in combination with other checkpoint miRNA inhibitors). The mean±SEM from 5-8 mice is shown.


Example 9. Expression of PD1-Targeting miRNAs in ROR1-Targeted CAR-T Cells

Briefly, miRNA-expressing ROR1 UltraCAR-T cells or control ROR1 UltraCAR-T cells were generated from T cells from five donors. PanT cells from five healthy donors were transfected with an indicated transposon vector (VVN-5355 or VVN-5351) plus SB11 transposase vector, and expanded by weekly stimulations for 4 weeks (˜35 days before adding beads) with ROR1 antigen presenting cells. Following a rest period of 7-8 days, UltraCAR-T cells were activated with CD3/CD28 dynabeads (1:1 bead: T cell ratio, T cells at 1×106 cells/mL) for 48 hours prior to RNA harvest, 7-8 days after the last AaPC stimulation. Expression of PD1-targeting guide miRNAs and the impact on PD1 mRNA expression were verified by RT-qPCR. Small RNAseq, which is an established method to identify the predicted and alternate miRNA sequences that may arise from a pri-miRNA (Borel et al, 2018; Miniarikova et al, 2016), was performed to compare expression levels of the PD1-targeting guide miRNAs to additional small RNA species, such as passenger miRNA, that may be generated from the PD1 silencer module. Small RNAseq was also used to assess potential changes on global endogenous miRNA expression (Mueller et al, 2012). RNAseq analysis was performed to evaluate global transcript expression and to identify changes in molecular pathway signaling or off-target gene silencing attributed to the PD1 silencer. In silico miRNA target prediction was performed using the miRanda algorithm to identify most likely targets of miRNAs generated from the PD1 silencer module. Expression of the predicted target genes was evaluated by RNAseq. Details of each method is included in the sections below.


To characterize expression of miRNAs encoded by the PD1 silencer and impact on PD1 transcript levels, RT-qPCR and small RNAseq were performed. To characterize changes in specific genes or cellular pathways, RNAseq was performed.


Nucleic acids were purified from cell pellets using Qiagen's AllPrep DNA/RNA/miRNA Universal kit (Cat #80224) following the manufacturers protocol. Total RNA was eluted in 50 uL nuclease-free water and concentration measured on a Nanodrop™ 2000 spectrophotometer.


To quantify the expression of PD1 guide and passenger miRNAs, total RNA was used as input for cDNA synthesis using Qiagen's miRCURY LNA RT Kit (#339340). Per the manufacturer's protocol, cDNA was diluted 1:60 with nuclease-free water and 3 μL of the diluted cDNA was used as input for qPCR using miRCURY LNA SYBR Green PCR Kit (Qiagen #339345) with custom miRCURY LNA primers specific to the two PD1 guide miRNAs. An endogenous miRNA, hsa-let-7a-5p, was quantified as a reference small RNA to allow for input normalization (Qiagen product #339306 with custom #YP00205727). Samples were run in a 384 well format on a QuantStudio 6 Flex instrument. Relative quantification (dCT) calculations were performed in Microsoft Excel and data graphed in GraphPad Prism 9. Calculations for dCT were performed on each technical replicate as follows:






dCT=CT(guide miRNA)−CT(hsa-let-7a)





ddCT=dCT(miRNACART replicate)−dCT(average of VVN-5355 technical replicates)





Fold Change=2{circumflex over ( )}-ddCT


The VVN-5355 ROR1 UltraCAR-T control sample serves as the reference control sample for comparison to miRNA expressing ROR1 UltraCAR-T cells within each donor set. Average fold change and standard deviation of technical replicates was calculated and reported in FIG. 10A.


To quantify and compare the production of guide and passenger miRNAs originating from the PD1 silencing module, RT-qPCR was performed as described above, except this second experiment included primer assays to detect the passenger miRNAs as well as an additional endogenous reference small RNA, RNU1A1. Expression calculations were performed to compare expression of each guide or passenger strand mature miRNA relative to the average of the endogenous control small RNAs as follows:






dCT=CT(mature miRNA)−CT(average of hsa-let-7a and RNU1A1)





Fold change=2{circumflex over ( )}-dCT


Average fold change was calculated from three technical replicates. These values are plotted in FIG. 11 with mean and standard deviation shown for the donor sample sets tested.


To quantify the expression of endogenous PD1 mRNA, cDNA synthesis was performed using a final RNA concentration of 5 ng/μL using Invitrogen's SuperScript IV VILO Master Mix with ezDNase enzyme kit (#11766050). Multiplex Taqman qPCR was performed using Invitrogen's TaqMan MastAdvanced Master Mix (#4444963) with 1 microliter of cDNA and Invitrogen (ThermoFisher Scientific) Taqman assays. Human PD1 Taqman assay (Invitrogen #Hs00169472_m1) was FAM labeled and the internal normalizer gene, ACTb (Invitrogen #Hs99999903_m1), which was VIC-labelled. Samples were run in a 384 well format on a QuantStudio 6 Flex instrument. Relative quantification (dCT) calculations were performed in Microsoft Excel as described above and data graphed in GraphPad Prism 9.









TABLE 13







Test and Control Articles









Sample Name
Description
Designation





VVN-5355
CAR-T cells generated using Sleeping
Control




Beauty transposon plasmid VVN-5355,




which contains a transgene cassette



to express ROR1-specific CAR +



mbIL-15 + HER1t.


VVN-5351
CAR-T cells generated using Sleeping
Test




Beauty transposon plasmid VVN-5351,

Article



which contains a transgene cassette



to express ROR1-specific CAR +



mbIL-15 + HER1t plus a PD1



silencer module.









The PD1 Silencer Module is designed to produce two mature guide miRNAs that bind to the PD1 transcript to silence PD1 expression. Expression of the two PD1-targeting guide miRNAs, referred to as PD1_1843 and PD1_2061, was confirmed in miRNA expressing ROR1 UltraCAR-T cells generated from multiple donors (FIG. 10A). A corresponding reduction in PD1 mRNA expression was verified in the miRNA-expressing ROR1 UltraCAR-T cells from all donors tested (FIG. 10B). This result demonstrated that the PD1 Silencer module produces the intended guide miRNAs and functions as designed.


To reduce the risk of silencing genes other than PD1, the PD1 silencer module was designed using pri-miRNA scaffolds that preferentially produce PD1-targeting guide miRNA over the non-targeting passenger miRNA. Both guide and passenger mature miRNAs were quantified from miRNA expressing ROR1 UltraCAR-T cells by RT-qPCR, which verified PD1 targeting guide miRNAs as the predominant species compared to non-targeting passenger strand miRNA (FIG. 4). The strong processing preference for the PD1 targeted guide miRNA was confirmed by small RNAseq, with 99.7% of reads mapping to the PD1 Silencer Module matching the intended PD1 targeting guide miRNAs (FIGS. 12 A-E). Furthermore, the start and stop position of the miRNAs was as expected, with miRNAs of 21-23 nucleotides detected that had the same 5′ end and variable length at the 3′ end (FIGS. 12 A-E). The extremely low incidence of passenger strand miRNAs and lack of unexpected small RNAs generated by aberrant RNA processing substantially reduced the risk for off-target gene silencing.


To ensure that expression of the PD1 Silencer module does not overwhelm the internal cellular RNAi machinery, endogenous miRNA counts were compared from miRNA expressing ROR1 UltraCAR-T cells vs control ROR1 UltraCAR-T cells. Examination of the top twenty expressed endogenous miRNAs demonstrated no statistically significant changes in expression across the samples (Table 15). Furthermore, the mature miRNAs generated from the PD1 silencer module accounted for approximately 4% of all quantified small RNAs (FIG. 13). The data indicated that expression of miRNAs from the PD1 silencer module did not saturate the cellular RNAi machinery and had no detectable impact on global endogenous miRNA expression.









TABLE 14







Small RNAseq Comparison of Guide and Passenger miRNA Counts










Normalized



Mature miRNA_Length (Sequence)
Mean
% Total





Guide PD1_204_21nt (TTCAGGAATGGGTTCCAAGGA; SEQ
486,432.3
88.2%


ID NO: 704)




Guide PD1_204_22nt (TTCAGGAATGGGTTCCAAGGAT;
 12,763.5



SEQ ID NO: 72)




Guide PD1_204_23nt (TTCAGGAATGGGTTCCAAGGATG;
 59,759.4



SEQ ID NO: 705)







Passenger PD1_204_21nt (TCCTGGAAGCTATTCCTGACG;
    380.6
 0.1%


SEQ ID NO: 706)




Passenger PD1_204_22nt (TCCTGGAAGCTATTCCTGACGT;
     20.2



SEQ ID NO: 707)




Passenger PD1_204_23nt
     54.7



(TCCTGGAAGCTATTCCTGACGTT; SEQ ID NO: 708)







Guide PD1_206_21nt (TATAATATAATAGAACCACAG; SEQ
  3,752.7
11.5%


ID NO: 709)




Guide PD1_206_22nt (TATAATATAATAGAACCACAGG;
 49,181.3



SEQ ID NO: 74)




Guide PD1_206_23nt (TATAATATAATAGAACCACAGGA;
 19,695.3



SEQ ID NO: 710)







Passenger PD1_206_21nt (TGTGGTTCTGTTATATCCATA;
  1,581.0
 0.3%


SEQ ID NO: 711)




Passenger PD1_206_22nt (TGTGGTTCTGTTATATCCATAT;
      3.1



SEQ ID NO: 712)




Passenger PD1_206_23nt
     13.2



(TGTGGTTCTGTTATATCCATATA; SEQ ID NO: 713)
















TABLE 15







Top 20 Expressed Endogenous Mature


miRNAs Detected by Small RNAseq














Log2 Fold
P-value



Endogenous miRNA
Base Mean
Change
Adjusted
















hsa-miR-181a-5p
796180.25
0.11
0.7948



hsa-miR-191-5p
421094.34
−0.19
0.5622



hsa-miR-21-5p
386727.78
0.03
0.9411



hsa-miR-92a-3p
368149.34
0.26
0.2298



hsa-miR-142-5p
345451.62
0.08
0.8549



hsa-miR-146b-5p
286127.09
0.24
0.5471



hsa-miR-16-5p
261883.66
0.00
0.9954



hsa-miR-146a-5p
229059.23
−0.27
0.4569



hsa-let-7f-5p
188491.61
0.16
0.6018



hsa-miR-22-3p
123453.58
−0.01
0.9899



hsa-miR-26a-5p
110714.77
0.19
0.5166



hsa-let-7a-5p
104253.31
0.00
0.9921



hsa-miR-155-5p
72533.08
−0.29
0.2298



hsa-miR-181b-5p
67527.35
0.12
0.7687



hsa-let-7i-5p
64631.72
0.30
0.2116



hsa-miR-21-3p
52519.27
0.25
0.5358



hsa-miR-186-5p
52479.56
−0.20
0.4189



hsa-miR-25-3p
50411.16
0.02
0.9696



hsa-miR-27a-3p
47911.68
−0.10
0.7818



hsa-let-7g-5p
47159.42
0.01
0.9811










Example 10: PD1 Silencer Module Specifically Reduces Expression of PD-1

An in silico miRNA target prediction algorithm, miRanda (Betel et al, 2008; Betel et al, 2010), was used to predict the most likely target transcripts for the guide and passenger miRNAs generated from the PD1 silencer module. The algorithm assigned a score for each potential miRNA target gene, with higher scores indicating higher likelihood of silencing by the input miRNA sequence. A summary of the top ten hits for each mature miRNA generated from the PD1 silencing module is listed in Table 16. PD1 is the only gene with perfect homology to any of the guide and passenger miRNAs and has the highest predicted miRanda score of any potential target gene. Expression of each predicted target gene was characterized from the RNAseq differential expression data set. PD1 was the most downregulated of all predicted target genes, with a log2 fold change (LFC) of −2.63 (˜84% PD1 reduction in miRNA expressing ROR1 UltraCAR-T cells compared to control ROR1 CAR-T cells) and a highly significant adjusted p-value. The expression of other predicted target genes was unchanged; those genes with adjusted p-values below 0.05 had LFC in the range of −0.33 to 0.22, which is an expression decrease or increase of <20%. One exception is a weakly predicted target gene of PD1_2061 guide miRNA, HDAC9, which has a LFC of ˜1.51. HDAC9 is a transcriptional repressor that is mechanistically linked to PD1 expression through BCL6 (Xie et al, 2017; Gil et al, 2016). It is likely that HDAC9 is not directly targeted by PD1_2061 guide miRNA, and that the reduction in HDAC9 is an indirect effect of reduced PD1 expression.









TABLE 16







In silico Predicted miRNA Target Genes



















free
%
Base







Target
miRanda
energy
Identity
Mean
log2


Adjusted


miRNA
Gene
Score
kcal/mol
to Target
Count
FoldChange
lfcSE
P value
P Value



















PD1_1843
PD1
200
−41.9
100.0
84
−2.63
0.293
2.51E−19
2.64E−16


miR204
MIER3
184
−32.1
84.2
1073
0.01
0.063
0.9104
0.9688


Guide
SLFN12L
182
−34.2
89.5
1404
−0.22
0.068
0.0015
0.0134


miRNA
FMO4
180
−28.5
84.2
15
−0.18
0.461
0.6975
NA



GIGYF1
180
−33.5
84.2
2749
−0.07
0.046
0.1123
0.3211



FAM83G
180
−31.5
84.2
171
0.33
0.144
0.0214
0.1026



PPRC1
180
−31.2
84.2
2679
−0.18
0.045
0.0001
0.0011



PRMT9
180
−29.1
84.2
335
0.09
0.100
0.3704
0.6432



MGAT4B
179
−29.3
88.9
4480
−0.21
0.063
0.0007
0.0076



BRWD1
179
−25.0
88.9
1816
0.05
0.052
0.3581
0.6328


PD1_1843
DOCK9
179
−28.5
83.3
1239
−0.12
0.077
0.1240
0.3428


miR204
NFYA
179
−27.5
83.3
1615
−0.02
0.052
0.7446
0.8911


Passenger
IFIT5
178
−30.4
89.5
379
−0.11
0.102
0.2629
0.5351


miRNA
OTUB2
178
−33.7
84.2
248
−0.32
0.131
0.0154
0.0805



UBIAD1
177
−32.1
88.9
1064
−0.07
0.066
0.3085
0.5859



MYO15A
176
−29.5
93.3
11
−0.25
0.510
0.6263
NA



ADRB2
175
−27.1
83.3
62
0.61
0.264
0.0206
0.0997



HIC2
175
−28.5
83.3
312
−0.01
0.102
0.8962
0.9621



NECTIN3
175
−33.5
72.2
662
0.05
0.081
0.5587
0.7864



TBC1D10C
175
−27.3
77.8
7562
0.03
0.041
0.4409
0.7056


PD1_2061
PD1
200
−34.3
100.0
84
−2.63
0.293
2.51E−19
2.64E−16


miR206
LCOR
180
−16.7
84.2
1321
−0.19
0.064
0.0029
0.0230


Guide
STARD4
179
−16.7
88.9
6871
−0.18
0.055
0.0013
0.0120


miRNA
UQCRC2
179
−19.1
81.0
4759
0.03
0.043
0.4641
0.7216



EXD2
178
−17.5
88.2
58
0.18
0.265
0.5055
0.7511



NOL8
177
−18.6
88.9
2190
−0.08
0.050
0.1007
0.2996



ATP23
176
−18.6
84.2
318
0.22
0.100
0.0265
0.1201



ELAPOR2
176
−20.6
84.2
70
−0.08
0.211
0.7139
0.8793



HDAC9
176
−19.5
84.2
73
−1.51
0.280
6.77E−08
3.53E−06



WDR47
176
−19.6
79.0
625
0.08
0.089
0.3892
0.6617


PD1_2061
COG2
172
−18.9
93.3
1244
0.06
0.060
0.3399
0.6170


miR206
NRG4
171
−25.6
77.8


Passenger
AGBL3
170
−19.2
79.0
49
0.01
0.250
0.9811
0.9934


miRNA
PDK3
170
−20.9
100.0
605
0.18
0.081
0.0256
0.1169



PDXK
170
−21.7
82.4
1786
0.22
0.058
0.0002
0.0022



N4BP2
169
−25.1
77.8
2834
−0.33
0.051
1.86E−10
1.95E−08



ZNF271P
169
−23.8
83.3
777
0.03
0.074
0.7197
0.8812



NFKB1
168
−18.8
88.2
7889
−0.11
0.039
0.0036
0.0269



XYLT1
168
−26.9
82.4
5281
0.08
0.051
0.1100
0.3177



CNOT6
167
−20.1
72.2
2030
−0.24
0.052
0.0000
0.0001









It is expected that changes in PD1 expression will impact expression of other genes in downstream pathways. As expected, analysis of RNAseq data confirmed the differential expression of several genes in miRNA expressing ROR1 UltraCAR-T cells compared to the control ROR1 UltraCAR-T cells (FIGS. 14A and B, Table 17). To elucidate direct versus indirect changes in gene expression, the differential expression (LFC) in miRNA expressing ROR1 UltraCAR-T cells compared to control ROR1 CAR-T cells was plotted against the predicted binding potential (predicted free energy) of the PD1 miRNAs for genes, which were in silico predicted as potential PD1 miRNA targets (FIGS. 15A-D). Genes with a highly negative free energy and a statistically significant reduction in expression are likely to be directly targeted by miRNA, while downregulated genes with a weak free energy are likely to be indirectly impacted by the miRNAs. PD1 is clearly separated from all other genes in the plots with a strong reduction in expression and high miRNA binding potential, which suggests a strong and preferential direct targeting of PD1, but not other genes, by the PD1 silencer miRNAs.









TABLE 17







Top 10 Down- and Up-Regulated Genes in ROR1 + PD1


Silencer Cells Relative to Control ROR1 UltraCAR-T Cells
















base
Log2Fold






Ensembl_ID
Symbol
Mean
Change
lfcSE
stat
pvalue
padj

















ENSG00000188389
PDCD1
84.2
−2.6
0.3
−9.0
2.51E−19
2.64E−16


ENSG00000172020
GAP43
71.6
−2.2
0.4
−5.7
1.37E−08
9.21E−07


ENSG00000171951
SCG2
110.5
−2.2
0.4
−6.1
1.13E−09
9.89E−08


ENSG00000105974
CAV1
172.3
−2.1
0.5
−4.2
2.93E−05
5.78E−04


ENSG00000136193
SCRN1
37.4
−1.9
0.5
−3.8
0.000136
1.97E−03


ENSG00000130287
NCAN
206.1
−1.7
0.4
−4.4
1.23E−05
2.84E−04


ENSG00000156052
GNAQ
145.0
−1.7
0.2
−7.8
6.88E−15
2.18E−12


ENSG00000135898
GPR55
84.5
−1.6
0.3
−5.7
1.47E−08
9.48E−07


ENSG00000152495
CAMK4
856.5
−1.6
0.2
−7.5
9.08E−14
2.21E−11


ENSG00000102271
KLHL4
184.3
−1.6
0.3
−4.7
3.00E−06
8.90E−05


ENSG00000165025
SYK
120.2
1.6
0.4
4.0
6.65E−05
1.10E−03


ENSG00000276597
TRBV11-3
123.1
1.6
0.6
2.7
0.006913
4.44E−02


ENSG00000082781
ITGB5
329.1
1.7
0.3
5.7
1.45E−08
9.44E−07


ENSG00000244242
IFITM10
555.8
1.7
0.2
7.6
4.18E−14
1.10E−11


ENSG00000143842
SOX13
116.9
1.7
0.2
7.6
3.87E−14
1.06E−11


ENSG00000180549
FUT7
318.7
1.7
0.4
4.3
1.71E−05
3.71E−04


ENSG00000137474
MYO7A
1057.3
1.8
0.2
8.2
3.56E−16
1.62E−13


ENSG00000177508
IRX3
317.9
1.8
0.5
3.3
0.001046
1.03E−02


ENSG00000136689
IL1RN
242.1
1.9
0.5
4.0
5.65E−05
9.70E−04


ENSG00000189013
KIR2DL4
800.0
1.9
0.3
6.4
1.51E−10
1.62E−08









Example 11

In one instance a patient is diagnosed with a particular cancer. In this example the patient is diagnosed with breast cancer. The patient is identified to receive a particular chimeric antigen receptor therapy. The patient provides an initial blood sample for isolation of the desired cell type. In this example, the isolated cell type is a T-cell. In this example only a portion of the isolated T-cells are transfected with the desired initial CAR selected from the collection of CARs. The remaining T-cells are coded and stored in appropriate condition for potential future use by the patient. The remaining T-cells are transfected through an appropriate means. In this example, the transfection occurs through a non-viral means. In particular, the non-viral means utilizes a single sleeping beauty vector that encodes for at least the selected CAR, a T-cell expansion cytokine, and a termination switch. The patient is treated within 48 hours from the initial T-cell isolation.


Once the above round of treatment is complete the patient is followed in accordance with monitoring protocols. In the event there is an expansion of the cancer, in this example breast cancer, the patient is reevaluated. The patient may be redosed with the initial CAR, however, in the event the evaluation demonstrates that after the initial or potential redose there is antigen escape the protocol will call for a dosing of a new CAR from the CAR collection. In this instance, the patient's stored T-cells will be selected for use in the transfection of the newly selected CAR. The patient will be able to be redosed in a timely fashion. Further as the patient has already received each portion of the follow-on therapy except the newly selected CAR, the risk of toxicities and or adverse events is reduced. Further the knowledge of previous T-cell expansion for this patient is informed by the levels of expansion from the initial CAR selection. This further allows for a potential change in the dosage size, be it greater or less than the initial dose.


Example 12

In one instance a patient is not diagnosed with a particular cancer, but rather a specific antigen has been identified as causing undesired proliferation of cells or expansion of an infective agent. The initial CAR is selected based on antigen being presented in the patient. The patient provides an initial blood sample for isolation of the desired cell type. In this example, the isolated cell type is a T-cell. In this example all of the isolated T-cells are transfected with the desired initial CAR selected from the collection of CARs. The T-cells are transfected through an appropriate means. In this example, the transfection occurs through a non-viral means. In particular, the non-viral means utilizes a single sleeping beauty vector that encodes for at least the selected CAR, a T-cell expansion cytokine, and a kill switch. The patient is treated within 48 hours from the initial T-cell isolation.


Once the above round of treatment is complete the patient is followed in accordance with monitoring protocols. Following further evaluation the patient demonstrates that there is antigen escape. The patient is then dosed with a new CAR from the CAR collection following collection of new T-cells. Further as the patient has already received each portion of the follow-on therapy except the newly selected CAR, the risk of toxicities and or adverse events is reduced. Further the knowledge of previous T-cell expansion for this patient is informed by the levels of expansion from the initial CAR selection. This further allows for a potential change in the dosage size, be it greater or less than the initial dose.


Example 13

The clinician develops a treatment plan for a patient that involves use of an initial CAR from the CAR collection. The treatment plan includes a dosing of different CARs from the collection in consecutive manner to prevent antigen escape.


Example 14

In one instance a patient is diagnosed with a particular cancer. In this example the patient is diagnosed with pancreatic cancer. The patient is identified to receive a particular chimeric antigen receptor therapy. The patient provides an initial blood sample for isolation of the desired cell type. In this example, the isolated cell type is a T-cell. In this example the isolated T-cell's are transfected with the desired initial CAR selected from the collection of CARs. The T-cells are transfected through a non-viral means utilizing a single sleeping beauty vector that encodes for at least the selected CAR, a T-cell expansion cytokine, and a kill switch. The patient is treated within 48 hours from the initial T-cell isolation.


Once the above round of treatment is complete the patient is followed in accordance with monitoring protocols. In this instance, the patient through reevaluation is noted to suffer antigen escape. In this instance the patient requires dosing with a new CAR. However, due to the condition of the patient a sufficient sample of autologous T-cell may not be obtained. As a result the method allows for the inclusion of allogenic T-cells to be incorporated to the therapy. The CAR from the library is transduced into the allogenic T-cells and the patient is dosed in a timely fashion. Further as the patient has already received each portion of the follow-on therapy, the risk of toxicities and or adverse events is reduced.


Example 15

In one instance a patient is not diagnosed with a particular cancer, but rather a specific antigen has been identified as causing undesired proliferation of cells or expansion of an infective agent. The initial CAR is selected based on antigen being presented in the patient. The patient is unable to provide an initial blood sample for isolation of the desired cell type due to the current health of the patient. As such, allogenic T-cells are transfected with the desired initial CAR selected from the collection of CARs. The T-cells are transfected through an appropriate means utilizing a single attsite vector that encodes for at least the selected CAR, a T-cell expansion cytokine, and a termination switch.


Once the above round of treatment is complete the patient is followed in accordance with monitoring protocols. Following further evaluation the patient demonstrates that there is antigen escape. The patient is then dosed with a new CAR using the same source of allogenic T-cells. Further as the patient has already received each portion of the follow-on therapy except the newly selected CAR, the risk of toxicities and or adverse events is reduced. Further the knowledge of previous T-cell expansion for this patient is informed by the levels of expansion from the initial CAR selection. This further allows for a potential change in the dosage size, be it greater or less than the initial dose.


Example 16

The clinician develops a treatment plan for a patient that involves use of an initial CAR from the CAR collection. The treatment plan includes a dosing of different CARs from the collection in consecutive manner to prevent antigen escape. This treatment plan may include the use of both autologous and allogenic cells.


Example 17

In one instance a patient is diagnosed with a specific antigen has been identified as causing undesired proliferation of cells or expansion of an infective agent. The initial CAR is selected based on antigen being presented in the patient. The patient is unable to provide an initial blood sample for isolation of the desired cell type due to the current health of the patient. As such, allogenic cells are transfected with the desired initial CAR selected from the collection of CARs. The cells are transfected through an appropriate means utilizing a single sleeping beauty vector that encodes for at least the selected CAR and a termination switch.


Once the above round of treatment is complete the patient is followed in accordance with monitoring protocols. Following further evaluation the patient demonstrates that there is antigen escape. The patient is then dosed with a new CAR using a source of allogenic T-cells or if the patient show sufficient improvement autologous T-cells.


Example 18

In one instance a patient is not diagnosed with a particular cancer, but rather a specific antigen has been identified as causing undesired proliferation of cells or expansion of an infective agent. The initial CAR is selected based on antigen being presented in the patient. The patient provides an initial blood sample for isolation of T-cells. The T-cells are transfected with the desired initial CAR selected from the collection of CARs utilizing a single sleeping beauty vector that encodes for the selected CAR, a T-cell expansion cytokine, and a kill switch. The patient is treated within 48 hours from the initial T-cell isolation.


Once the above round of treatment is complete the patient is followed in accordance with monitoring protocols. Following further evaluation the patient demonstrates that there is antigen escape. The patient is then dosed with the same CAR and a new CAR from the CAR collection.


Example 19

In one instance a patient is diagnosed with a particular cancer. In this example the patient is diagnosed with AML. The patient is identified to receive a particular chimeric antigen receptor therapy. The patient provides an initial blood sample for isolation of the desired cell type. The cells are transfected with the desired two different CARs selected from the collection of CARs. The cells are transfected through a non-viral means utilizing a single vector that encodes for both selected CARs and a kill switch.


Once the above round of treatment is complete the patient is followed in accordance with monitoring protocols. In this instance, the patient through reevaluation is noted to suffer antigen escape. In this instance the patient requires dosing with a new CAR. However, due to the condition of the patient a sufficient sample of autologous cell may not be obtained. As a result the method allows for the inclusion of allogenic T-cells to be incorporated to the therapy. The CAR from the library is transduced into the allogenic cells and the patient is dosed in a timely fashion.












Nucleic Acid Sequences Encoding Backbone miRNA Sequences










Synthetic
DNA encoding 5′
DNA encoding stem
DNA encoding 3′


miRNA
backbone sequence
loop
backbone sequence





miR204
AGGAGGGTGGGGGTGG
GAGAATATATGAAGG
GTTCAATTGTCATCACTG



AGGCAAGCAGAGGACTT
(SEQ ID NO: 2)
GCATCTTTTTTGATCATTG



CCTGATCGCGTACCCATG

CACCATCATCAAATGCAT



GCTACAGTCTTTCTTCATG

TGGGATAACCATGAC



TGACTCGTGGAC

(SEQ ID NO: 3)



(SEQ ID NO: 1)







miR206
GATGCTACAAGTGGCCCA
TATGGATTACTTTGCTA
TTTCGGCAAGTGCCTCCT



CTTCTGAGATGCGGGCTG
(SEQ ID NO: 5)
CGCTGGCCCCAGGGTACC



CTTCTGGATGACACTGCT

ACCCGGAGCACAGGTTTG



TCCCGAGG

GTGACCTT



(SEQ ID NO: 4)

(SEQ ID NO: 6)





miR17
TTAGCAGGAAAAAAGAG
TGATATGTGCAT
GCATTATGGTGACAGCTG



AACATCACCTTGTAAAAC
(SEQ ID NO: 8)
CCTCGGGAAGCCAAGTTG



TGAAGATTGTGACCAGTC

GGCTTTAAAGTGCAGGG



AGAATAATGT

CCTGCTGATGT



(SEQ ID NO: 7)

(SEQ ID NO: 9)





miR150
AGGGACTGGGCCCACGG
CTGGGCTCAG
CAGGGACCTGGGGACCC



GGAGGCAGCGTCCCCGA
(SEQ ID NO: 11)
CGGCACCGGCAGGCCCC



GGCAGCAGCGGCAGCGG

AAGGGGTGAGGTGAGCG



CGGCTCCTCTCCCCATGG

GGCATTGGGACCTCCCCT



CCCTG

CCCTGTACTC



(SEQ ID NO: 10)

(SEQ ID NO: 12)





miR150
AGGGACTGGGCCCACGG
TGCTGGGCTCAGACC
GGACCTGGGGACCCCGG



GGAGGCAGCGTCCCCGA
(SEQ ID NO: 14)
CACCGGCAGGCCCCAAG



GGCAGCAGCGGCAGCGG

GGGTGAGGTGAGCGGGC



CGGCTCCTCTCCCCATGG

ATTGGGACCTCCCCTCCC



CC

TGTACTC



(SEQ ID NO: 13)

(SEQ ID NO: 15)





miR16
CTTCTGAAGAAAATATAT
TTAAGATTCTAAAATTATC
AAGTAAGGTTGACCATAC



TTCTTTTTATTCATAGCTC
T
TCTACAGTTGTGTTTTAAT



TTATGATAGCAATGTCAG
(SEQ ID NO: 17)
GTATATTAATGTTACTAAT



CAGTGCCT

GTGTTTT



(SEQ ID NO: 16)

(SEQ ID NO: 18)





miR30a
CAGGTTAACCCAACAGAA
CTGTGAAGCCACAGATG
TGCCTACTGCCTCGGACT



GGCTAAAGAAGGTATATT
GG
TCAAGGGGCTACTTTAGG



GCTGTTGACAGTGAGCG
(SEQ ID NO: 20)
AGCAATTATCTTGTTTA



AC

(SEQ ID NO: 21)



(SEQ ID NO: 19)







miR126
CCGGGGTCCTGTCTGCAT
CTGTGACACTTCAAAC
CCGTCCACGGCACCGCAT



CCAGCGCAGCATTCTGGA
(SEQ ID NO: 23)
CGAAAACGCCGCTGAGA



AGACGCCACGCCTCCGCT

CCTCAGCCTTGACCTCCCT



GGCGACGG

CAGCGTGG



(SEQ ID NO: 22)

(SEQ ID NO: 24)





miR122
CAATGGTGGAATGTGGA
TGTCTAAACTAT
TAGCTACTGCTAGGCAAT



GGTGAAGTTAACACCTTC
(SEQ ID NO: 26)
CCTTCCCTCGATAAATGTC



GTGGCTACAGAGTTTCCT

TTGGCATCGTTTGCTTTG



TAGCAGAGCTG

AGCAAGAAGGTTCATCTG



(SEQ ID NO: 25)

ATATCAGTCTTCTCAATCT





(SEQ ID NO: 27)





miR214
ACAGGCTGATTGTATCTG
GCAGAACATCCGCTCACC
CACATGACAACCCAGCCT



TCTATGAGCAAAGGAAAC
TGT
GAATGACAACCAGCCATT



CTGAAGGAACCAAGGGC
(SEQ ID NO: 29)
GAAAGAAAGCAGCCCTC



CTGGCTGGACAGAGTTGT

ACACCATAGCATCTA



CATGTG

(SEQ ID NO: 30)



(SEQ ID NO: 28)







miR214
ACAGGCTGATTGTATCTG
AGAACATCCGCTCACCT
CACATGACAACCCAGCCT



TCTATGAGCAAAGGAAAC
(SEQ ID NO: 32)
GAATGACAACCAGCCATT



CTGAAGGAACCAAGGGC

GAAAGAAAGCAGCCCTC



CTGGCTGGACAGAGTTGT

ACACCATAGCATCTA



CATGTGTC

(SEQ ID NO: 33)



(SEQ ID NO: 31)







miR29b1
GGGTTTATTGTAAGAGAG
GATTTAAATAGTGATTGT
CTTGGGGGAGACCAGCT



CATTATGAAGAAAAAAAT
C
GCGCTGCACTACCAACAG



AGATCATAAAGCTTCTTC
(SEQ ID NO: 35)
CAAAAGAAGTGAATGGG



AGG

ACAGCT



(SEQ ID NO: 34)

(SEQ ID NO: 36)





miR29b1
GGGTTTATTGTAAGAGAG
TTTAAATAGTGATTG
GTTCTTGGGGGAGACCA



CATTATGAAGAAAAAAAT
(SEQ ID NO: 38)
GCTGCGCTGCACTACCAA



AGATCATAAAGCTTCTTC

CAGCAAAAGAAGTGAAT



AGGAA

GGGACAGCT



(SEQ ID NO: 37)

(SEQ ID NO: 39)





miR133a1
TTTACCAATGAAAAGCAT
TCGCCTCTTCAATGGA
TAGCTATGCATTGATTAC



TTAACTGTTTTGGATTCCA
(SEQ ID NO: 41)
TACGGGACAACCAACGTT



AACTAGCAGCACTACAAT

TTCATTTGTGAATATCAAT



GCTTTGCTA

TACTTGCCA



(SEQ ID NO: 40)

(SEQ ID NO: 42)





miR26a
TGAAGCCACAGGAGCCA
GTGCAGGTCCCAATG
CGGGGACGCGGGCCTGG



AGAGCAGGAGGACCAAG
(SEQ ID NO: 44)
ACGCCGGCATCCGGGCTC



GCCCTGGCGAAGGCCGT

AGGACCCCCCTCTCTGCC



GGCCTCG

AGAGGC



(SEQ ID NO: 43)

(SEQ ID NO: 45)





miR412
GTCTTGGAGGCTGGGGC
GTTTCT
GCCGTCCGTATCCGCTGC



ACCTCGGGGAAGGACGC
(SEQ ID NO: 47)
AGCCTGTGGGGCCTGCG



CGGCATCAGCACCATTCT

GGCCGGGGAGCCGATCG



GGGGTACGGGGATGGA

CGCTTCAGCTCAGCGCCT



(SEQ ID NO: 46)

(SEQ ID NO: 48)





miR-19
CCAATAATTCAAGCCAAG
TACAAGAAGAATGTAGT
TGGTGGCCTGCTATTTCC



CAAGTATATAGGTGTTTT
(SEQ ID NO: 50)
TTCAAATGAATGATTTTTA



AATAGTTTTTGTTTGCAGT

CTAATTTTGTGTACTTTTA



CCTCTG

TTGTG



(SEQ ID NO: 49)

(SEQ ID NO: 51)





miR-21
TGTCTGCTTGTTTTGCCTA
CTGTTGAATCTCATGG
TCTGACATTTTGGTATCTT



CCATCGTGACATCTCCAT
(SEQ ID NO: 53)
TCATCTGACCATCCATATC



GGCTGTACCACCTTGTCG

CAATGTTCTCATTTAAACA



GG

(SEQ ID NO: 54)



(SEQ ID NO: 52)







miR-142
GGAGTCAGGAGGCCTGG
AACAGCACTGGAGGG
GATGAGTGTACTGTGGG



GCAGCCTGAAGAGTACA
(SEQ ID NO: 56)
CTTCGGAGATCACGCCAC



CGCCGACGGACAGACAG

TGCTGCCGCCCGCTGCCC



ACAGTGCAGTCACC

GCCACCATCTTC



(SEQ ID NO: 55)

(SEQ ID NO: 57)





miR-494
CAGTTCTGTTTTGATTTTT
TCTTTATTTATGA
TTTTTTAGTATCAAATCCC



TTTGTTTGTTTTTTGATCA
(SEQ ID NO: 59)
ACCCTGGAGGCACTTCCT



GTGCTAATCTTCGATACT

GTTCCTGATGCAGCCTTC



CGAAGGA

AGGGAGG



(SEQ ID NO: 58)

(SEQ ID NO: 60)





miR-1915
CGGACCACGGTGTCCCCT
GTGCACCCGTG
GCGGCCCTAGCGACCTGC



TCTCTCCAGCTGGGGGTC
(SEQ ID NO: 62)
GGCGGCGCCGGGAAAGC



TCGGGTCCTGGCGCTGAG

CCTGCCTCTGCAGCGGGT



AGGCCGC

CCCAGGGGTC



(SEQ ID NO: 61)

(SEQ ID NO: 63)



















Nucleic acid sequences encoding mature miRNA sequences










miRNA


DNA encoding passenger


Target
Backbone
DNA encoding guide miRNA
miRNA





CTLA4
miR-204
AATATAGTCTTCTCCCTCGCTT
AAGCAGGGACAGGACTATATT




(SEQ ID NO: 64)
(SEQ ID NO: 65)





CTLA4
miR-26a
AATATAGTCTTCTCCCTCGCTT
GAGCGAGGGATAGGACTATACT




(SEQ ID NO: 66)
(SEQ ID NO: 67)





CTLA4
miR-30a
AATATAGTCTTCTCCCTCGCTG
CAGTGAGGGAAGACTATGTT




(SEQ ID NO: 68)
(SEQ ID NO: 69)





CTLA4
miR-206
AATATAGTCTTCTCCCTCGCTG
CAGCGAGGGAGAAGATTAC




(SEQ ID NO: 70)
CATT





(SEQ ID NO: 71)





PD1
miR-204
TTCAGGAATGGGTTCCAAGGAT
ATCCTGGAAGCTATTCCTGAC




(SEQ ID NO: 72)
(SEQ ID NO: 73)





PD1
miR-206
TATAATATAATAGAACCACAGG
CCTGTGGTTCTGTTATATCCA




(SEQ ID NO: 74)
TA





(SEQ ID NO: 75)





PD1
miR-30a
TTCAGGAATGGGTTCCAAGGAG
CTCCTTGGACCATTCCTGAA




(SEQ ID NO: 76)
(SEQ ID NO: 77)





PD1
miR-412
TTCAGGAATGGGTTCCAAGGAATT
AATGTCCTGAAGCCATTCAT




(SEQ ID NO: 78)
GGA





(SEQ ID NO: 79)





PD1
miR122
TTCAGGAATGGGTTCCAAGGAG
CTCCTTGGAACACATTCCTAC




(SEQ ID NO: 80)
A





(SEQ ID NO: 81)





PD1
miR-17
TTCAGGAATGGGTTCCAAGGAAG
CTTCTTGGTACACGTTCCTGC




(SEQ ID NO: 82)
TCA





(SEQ ID NO: 83)





PD1
miR-150
TATAATATAATAGAACCACAGG
ACGTGTGGTTTATCCTGTTGT




(SEQ ID NO: 74)
A





(SEQ ID NO: 85)





PD1
miR-486
TATAATATAATAGAACCACAGG
CCAGCTTGTGGTTCTATTATG




(SEQ ID NO: 74)
TTATA





(SEQ ID NO: 87)





TIGIT
miR-17
AGATCCACGTTACTCACCCTAG
CTAGGGTGTGTCATGTGGAT




(SEQ ID NO: 88)
GAA





(SEQ ID NO: 89)





TIGIT
miR-150
AGATCCACGTTACTCACCCGTG
ACCCGGGTGATAATATGGAT




(SEQ ID NO: 90)
CT





(SEQ ID NO: 91)





TIGIT
miR-204
AGATCCACGTTACTCACCCCCT
AGGGGTGAGAAGCGTGGAT




(SEQ ID NO: 92)
CC





(SEQ ID NO: 93)





TIGIT
miR29b1
AGATCCACGTTACTCACCCTTA
TAGGTGAGTTACGTGGATCT




(SEQ ID NO: 94)
GTT





(SEQ ID NO: 95)





TIGIT
miR214
AGATCCACGTTACTCACCCTGC
GTAGGGTGAGACTCGTGGA




(SEQ ID NO: 96)
TCT





(SEQ ID NO: 97)





TIGIT
miR-206
ACCACGATGACTGCTGTGCAGA
TCTGCGTAGCAGTCATCGCC




(SEQ ID NO: 98)
GGT





(SEQ ID NO: 99)





TIGIT
miR-204
ACCACGATGACTGCTGTGCAGA
TCTGATGGCCGTCATCGTGGC




(SEQ ID NO: 100)
(SEQ ID NO: 101)





TIGIT
miR-22
ACCACGATGACTGCTGTGCAGA
TCTGTACAGCAGCATCGATG




(SEQ ID NO: 102)
GT





(SEQ ID NO: 103)





TIGIT
miR-16
ACCACGATGACTGCTGTGCAGA
TGCGCAGCTGTTCATCGTGGT




(SEQ ID NO: 104)
(SEQ ID NO: 105)





TIGIT
miR-21
ACCACGATGACTGCTGTGCAGA
CTGCGAAGCAGCATCGTGGC




(SEQ ID NO: 106)
(SEQ ID NO: 107)





TIGIT
miR-494
ACCACGATGACTGCTGTGCAGA
TCTGCAACGCAGTCTCGCTGG




(SEQ ID NO: 108)
(SEQ ID NO: 109)





TIGIT
miR-142
ACCACGATGACTGCTGTGCAGA
GATGCACAGCAATCATCGTG




(SEQ ID NO: 110)
GT





(SEQ ID NO: 111)





TIGIT
miR-19
ACCACGATGACTGCTGTGCAGAT
ATCTGCACAGCAGTTCGTGG




(SEQ ID NO: 112)
(SEQ ID NO: 113)





TIGIT
miR-1915
ACCACGATGACTGCTGTGCAGA
TCTGCCAATAGCTGTAATCG




(SEQ ID NO: 114)
TGG





(SEQ ID NO: 115)





TIGIT
miR-206
TATCGTTCACGGTCAGCGACTG
CAGTCGTTGACTGTGGACTT




(SEQ ID NO: 116)
ATA





(SEQ ID NO: 117)





TIGIT
miR-204
TATCGTTCACGGTCAGCGACTG
CAGTGCTGAACGTGAACGATC




(SEQ ID NO: 118)
(SEQ ID NO: 119)





TIGIT
miR-22
TATCGTTCACGGTCAGCGACTG
TAGTCGCTGACTTGAACAGA




(SEQ ID NO: 120)
TA





(SEQ ID NO: 121)





TIGIT
miR-142
TATCGTTCACGGTCAGCGACT
TGTCGCTGACAGTGAACGATA




(SEQ ID NO: 122)
(SEQ ID NO: 123)





TIGIT
miR-16
TATCGTTCACGGTCAGCGACTG
GTCGCTGAGCGCTGAACGATG




(SEQ ID NO: 124)
(SEQ ID NO: 125)





TIGIT
miR-206
TAACTCAGGACATTGAAGTAGT
ACTACTTCAATGTCCTGACCT




(SEQ ID NO: 126)
TA





(SEQ ID NO: 127)





TIGIT
miR-204
TAACTCAGGACATTGAAGTAGT
ACTATTCAGAGTCCTGAGTTC




(SEQ ID NO: 128)
(SEQ ID NO: 129)





TIGIT
miR-21
TAACTCAGGACATTGAAGTAGT
CTACTCCAATGCCTGAGTTG




(SEQ ID NO: 130)
(SEQ ID NO: 131)





TIGIT
miR-494
TAACTCAGGACATTGAAGTAGT
ACTACTGTAATGTCTGACGTT




(SEQ ID NO: 132)
(SEQ ID NO: 133)





TIGIT
miR-19
TAACTCAGGACATTGAAGTAGTC
GACTACTTCAATGTTGAGTT




(SEQ ID NO: 134)
(SEQ ID NO: 135)





TIGIT
miR-1915
AGATCCACGTTACTCACCCTAG
CTAGGCAGTGAGCAAAGTG




(SEQ ID NO: 136)
GATC





(SEQ ID NO: 137)





TIGIT
miR-204
AGATCCACGTTACTCACCCTAG
CTAGGTGAGAAACGTGGATCG




(SEQ ID NO: 138)
(SEQ ID NO: 139)





TIGIT
miR-206
AGATCCACGTTACTCACCCTAG
CTAGGGTGAGTAACGTGGCC




(SEQ ID NO: 140)
TCT





(SEQ ID NO: 141)





TIGIT
miR-21
AGATCCACGTTACTCACCCTAG
TAGGGCGAGTACGTGGATCG




(SEQ ID NO: 142)
(SEQ ID NO: 143)





TIGIT
miR-22
AGATCCACGTTACTCACCCTAG
CTAGGGTGAGTACGTGGCAT




(SEQ ID NO: 144)
CT





(SEQ ID NO: 145)





TIM3
miR204
CATTATGCCTGGGATTTGGATC
GATCAGATCGCAGGCATAATT




(SEQ ID NO: 146)
(SEQ ID NO: 147)





TIM3
miR206
CATTATGCCTGGGATTTGGATC
GATCCGGATCCTAGGTATCC




(SEQ ID NO: 148)
ATG





(SEQ ID NO: 149)





TIM3
miR17
CATTATGCCTGGGATTTGGATCG
CGGCCAGAACCAAGGCATAA




(SEQ ID NO: 150)
GTA





(SEQ ID NO: 151)





TIM3
miR126
CATTATGCCTGGGATTTGGATC
ACTCCAAATCCCGTGCATAA




(SEQ ID NO: 152)
TCG





(SEQ ID NO: 153)





TIM3
miR122
CATTATGCCTGGGATTTGGATC
GATTCGAATCCAAGGCATAT




(SEQ ID NO: 154)
GG





(SEQ ID NO: 155)





TIM3
miR214
TAATTCACATCCCTTTCATCAG
ATGATGAAAGGACAGTGAA




(SEQ ID NO: 156)
TTA





(SEQ ID NO: 157)





LAG3
miR-30a
TAGTCGTTGGGTAAAGTCGCCA
TGGTGACTTCTCAACGATTA




(SEQ ID NO: 158)
(SEQ ID NO: 159)





LAG3
miR122
GTTGCTTTCCGCTAAGTGGTGA
TCACCACTTAGAGGAAAGCC




(SEQ ID NO: 160)
TC





(SEQ ID NO: 161)





GITR
miR206
TTTGCAGTGGCCTTCGTGGCCC
GGGCTATGAAGGCCATTGA




(SEQ ID NO: 162)
GAAA





(SEQ ID NO: 163)





GITR
miR29b1
AGCCTCCCGTCCTAAGACCCCAC
GTGGGTCTTAGGTCGGGAG




(SEQ ID NO: 164)
CTGCT





(SEQ ID NO: 165)





PIK3IP1
miR206
TGAACGACCAGTGTTTAACCGG
CCGGTTGAACATTGGTTGCC




(SEQ ID NO: 166)
TCA





(SEQ ID NO: 167)





PIK3IP1
miR126
TTCTCCTTGGAGTTCATCCGCG
ATGAACTTAGAGGAGACG




(SEQ ID NO: 168)
(SEQ ID NO: 169)





PIK3IP1
miR30a
TTCTCCTTGGAGTTCATCCGCG
CGCGGATGATCCAAGGAGGA




(SEQ ID NO: 170)
(SEQ ID NO: 171)



















Nucleic acid sequences encoding non-naturally occurring pri-miRNA


sequences









miRNA
Encoded pri-



Target
miRNA(s)
DNA Sequence





CTLA4
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACAATA




TAGTCTTCTCCCTCGCTTGAGAATATATGAAGGAAGCAGGGAC




AGGACTATATTGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 178)





PD1
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCA




GGAATGGGTTCCAAGGATGAGAATATATGAAGGATCCTGGAA




GCTATTCCTGACGTTCAATTGTCATCACTGGCATCTTTTTTGATC




ATTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 179)





PD1
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATAT




GGATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCA




AGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGT




TTGGTGACCTT




(SEQ ID NO: 180)





TIGIT
miR17
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATT




GTGACCAGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTG




ATATGTGCATCTAGGGTGTGTCATGTGGATGAAGCATTATGGT




GACAGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGGG




CCTGCTGATGT




(SEQ ID NO: 181)





TIGIT
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAG




CGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGAGATCCACGTTA




CTCACCCGTGCTGGGCTCAGACCCGGGTGATAATATGGATCTC




AGGGACCTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTGA




GGTGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 182)





TIGIT
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACAGAT




CCACGTTACTCACCCCCTGAGAATATATGAAGGAGGGGTGAGA




AGCGTGGATCCGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 183)





TIGIT
miR-206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGTCTGCGTAGCAGTCATCGCCGGTTA




TGGATTACTTTGCTAACCACGATGACTGCTGTGCAGATTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 184)





TIGIT
miR-204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACACCA




CGATGACTGCTGTGCAGAGAGAATATATGAAGGTCTGATGGCC




GTCATCGTGGCGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 185)





TIGIT
miR-22
CATTTTCCCTCCCTTTCCCTTAGGAGCCTGTTCCTCTCACGCCCTC




ACCTGGCTGAGCCGCATCTGTACAGCAGCATCGATGGTTATGTC




CTGACCCAGCTAACCACGATGACTGCTGTGCAGATGCCCTCTGC




CCCTGGCTTCGAGGAGGAGGAGGAGCTGCTTTCCCCATCATCT




GGAAGGTG




(SEQ ID NO: 186)





TIGIT
miR-16
cttcTGAAGAAAATATATTTCTTTTTATTCATAGCTCTTATGATAG




CAATGTCAGCAGTGCCTACCACGATGACTGCTGTGCAGATTAA




GATTCTAAAATTATCTTGCGCAGCTGTTCATCGTGGTAGTAAGG




TTGACCATACTCTACAGTTGTGTTTTAATGTATATTAATGTTACT




AATGTGTTTT




(SEQ ID NO: 187)





TIGIT
miR-16
cttcTGAAGAAAATATATTTCTTTTTATTCATAGCTCTTATGATAG




CAATGTCAGCAGTGCCTACCACGATGACTGCTGTGCAGATTAA




GATTCTAAAATTATCTTGCGCAGCTGTTCATCGTGGTAAGTAAG




GTTGACCATACTCTACAGTTGTGTTTTAATGTATATTAATGTTAC




TAATGTGTTTT




(SEQ ID NO: 188)





TIGIT
miR-21
TGTCTGCTTGTTTTGCCTACCATCGTGACATCTCCATGGCTGTAC




CACCTTGTCGGGACCACGATGACTGCTGTGCAGACTGTTGAATC




TCATGGCTGCGAAGCAGCATCGTGGCTCTGACATTTTGGTATCT




TTCATCTGACCATCCATATCCAATGTTCTCATTTAAACA




(SEQ ID NO: 189)





TIGIT
miR-494
CAGTTCTGTTTTGATTTTTTTTGTTTGTTTTTTGATCAGTGCTAAT




CTTCGATACTCGAAGGATCTGCAACGCAGTCTCGCTGGTCTTTA




TTTATGAACCACGATGACTGCTGTGCAGATTTTTTAGTATCAAA




TCCCACCCTGGAGGCACTTCCTGTTCCTGATGCAGCCTTCAGGG




AGG




(SEQ ID NO: 190)





TIGIT
miR-142
GGAGTCAGGAGGCCTGGGCAGCCTGAAGAGTACACGCCGACG




GACAGACAGACAGTGCAGTCACCACCACGATGACTGCTGTGCA




GAACAGCACTGGAGGGATGCACAGCAATCATCGTGGTGATGA




GTGTACTGTGGGCTTCGGAGATCACGCCACTGCTGCCGCCCGC




TGCCCGCCACCATCTTC




(SEQ ID NO: 191)





TIGIT
miR-19
CCAATAATTCAAGCCAAGCAAGTATATAGGTGTTTTAATAGTTT




TTGTTTGCAGTCCTCTGATCTGCACAGCAGTTCGTGGTACAAGA




AGAATGTAGTACCACGATGACTGCTGTGCAGATTGGTGGCCTG




CTATTTCCTTCAAATGAATGATTTTTACTAATTTTGTGTACTTTTA




TTGTG




(SEQ ID NO: 192)





TIGIT
miR-1915
CGGACCACGGTGTCCCCTTCTCTCCAGCTGGGGGTCTCGGGTCC




TGGCGCTGAGAGGCCGCACCACGATGACTGCTGTGCAGAGTGC




ACCCGTGTCTGCCAATAGCTGTAATCGTGGGCGGCCCTAGCGA




CCTGCGGCGGCGCCGGGAAAGCCCTGCCTCTGCAGCGGGTCCC




AGGGGTC




(SEQ ID NO: 193)





TIGIT
miR-206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCAGTCGTTGACTGTGGACTTATATA




TGGATTACTTTGCTATATCGTTCACGGTCAGCGACTGTTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 194)





TIGIT
miR-204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTATC




GTTCACGGTCAGCGACTGGAGAATATATGAAGGCAGTGCTGAA




CGTGAACGATCGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 195)





TIGIT
miR-22
CATTTTCCCTCCCTTTCCCTTAGGAGCCTGTTCCTCTCACGCCCTC




ACCTGGCTGAGCCGCATAGTCGCTGACTTGAACAGATATATGTC




CTGACCCAGCTATATCGTTCACGGTCAGCGACTGTGCCCTCTGC




CCCTGGCTTCGAGGAGGAGGAGGAGCTGCTTTCCCCATCATCT




GGAAGGTG




(SEQ ID NO: 196)





TIGIT
miR-142
GGAGTCAGGAGGCCTGGGCAGCCTGAAGAGTACACGCCGACG




GACAGACAGACAGTGCAGTCACCTATCGTTCACGGTCAGCGAC




TAACAGCACTGGAGGGTGTCGCTGACAGTGAACGATAGATGA




GTGTACTGTGGGCTTCGGAGATCACGCCACTGCTGCCGCCCGC




TGCCCGCCACCATCTTC




(SEQ ID NO: 197)





TIGIT
miR-16
cttcTGAAGAAAATATATTTCTTTTTATTCATAGCTCTTATGATAG




CAATGTCAGCAGTGCCTTATCGTTCACGGTCAGCGACTGTTAAG




ATTCTAAAATTATCTGTCGCTGAGCGCTGAACGATGAAGTAAG




GTTGACCATACTCTACAGTTGTGTTTTAATGTATATTAATGTTAC




TAATGTGTTTT




(SEQ ID NO: 198)





TIGIT
miR-206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGACTACTTCAATGTCCTGACCTTATAT




GGATTACTTTGCTATAACTCAGGACATTGAAGTAGTTTTCGGCA




AGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGT




TTGGTGACCTT




(SEQ ID NO: 199)





TIGIT
miR-204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTAAC




TCAGGACATTGAAGTAGTGAGAATATATGAAGGACTATTCAGA




GTCCTGAGTTCGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 200)





TIGIT
miR-21
TGTCTGCTTGTTTTGCCTACCATCGTGACATCTCCATGGCTGTAC




CACCTTGTCGGGTAACTCAGGACATTGAAGTAGTCTGTTGAATC




TCATGGCTACTCCAATGCCTGAGTTGTCTGACATTTTGGTATCTT




TCATCTGACCATCCATATCCAATGTTCTCATTTAAACA




(SEQ ID NO: 201)





TIGIT
miR-494
CAGTTCTGTTTTGATTTTTTTTGTTTGTTTTTTGATCAGTGCTAAT




CTTCGATACTCGAAGGAACTACTGTAATGTCTGACGTTTCTTTAT




TTATGATAACTCAGGACATTGAAGTAGTTTTTTTAGTATCAAATC




CCACCCTGGAGGCACTTCCTGTTCCTGATGCAGCCTTCAGGGAGG




(SEQ ID NO: 202)





TIGIT
miR-19
CCAATAATTCAAGCCAAGCAAGTATATAGGTGTTTTAATAGTTT




TTGTTTGCAGTCCTCTGGACTACTTCAATGTTGAGTTTACAAGA




AGAATGTAGTTAACTCAGGACATTGAAGTAGTCTGGTGGCCTG




CTATTTCCTTCAAATGAATGATTTTTACTAATTTTGTGTACTTTTA




TTGTG




(SEQ ID NO: 203)





TIGIT
miR-1915
CGGACCACGGTGTCCCCTTCTCTCCAGCTGGGGGTCTCGGGTCC




TGGCGCTGAGAGGCCGCAGATCCACGTTACTCACCCTAGGTGC




ACCCGTGCTAGGCAGTGAGCAAAGTGGATCGCGGCCCTAGCGA




CCTGCGGCGGCGCCGGGAAAGCCCTGCCTCTGCAGCGGGTCCC




AGGGGTC




(SEQ ID NO: 204)





TIGIT
miR-204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACAGAT




CCACGTTACTCACCCTAGGAGAATATATGAAGGCTAGGTGAGA




AACGTGGATCGGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 205)





TIGIT
miR-206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCTAGGGTGAGTAACGTGGCCTCTT




ATGGATTACTTTGCTAAGATCCACGTTACTCACCCTAGTTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 206)





TIGIT
miR-21
TGTCTGCTTGTTTTGCCTACCATCGTGACATCTCCATGGCTGTAC




CACCTTGTCGGGAGATCCACGTTACTCACCCTAGCTGTTGAATC




TCATGGTAGGGCGAGTACGTGGATCGTCTGACATTTTGGTATCT




TTCATCTGACCATCCATATCCAATGTTCTCATTTAAACA




(SEQ ID NO: 207)





TIGIT
miR-22
CATTTTCCCTCCCTTTCCCTTAGGAGCCTGTTCCTCTCACGCCCTC




ACCTGGCTGAGCCGCACTAGGGTGAGTACGTGGCATCTTATGT




CCTGACCCAGCTAAGATCCACGTTACTCACCCTAGTGCCCTCTG




CCCCTGGCTTCGAGGAGGAGGAGGAGCTGCTTTCCCCATCATC




TGGAAGGTG




(SEQ ID NO: 208)





TIM3
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACCATT




ATGCCTGGGATTTGGATCGAGAATATATGAAGGGATCAGATCG




CAGGCATAATTGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 209)





TIM3
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAG




CGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGCATTATGCCTGG




GATTTGGATCCTGGGCTCAGACCTCCAAATCCACACATAGTGCA




GGGACCTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTGAG




GTGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 210)





TIM3
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACCATTATGCCTGGGATTTGGATCCTGTGAAGC




CACAGATGGGGATCCAGATCAGGCATAGTGGCTGCCTACTGCC




TCGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 211)





TIM3
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGGATCCGGATCCTAGGTATCCATGTA




TGGATTACTTTGCTACATTATGCCTGGGATTTGGATCTTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 212)





TIM3
miR16
CTTCTGAAGAAAATATATTTCTTTTTATTCATAGCTCTTATGATA




GCAATGTCAGCAGTGCCTCATTATGCCTGGGATTTGGATCTTAA




GATTCTAAAATTATCTTCCGAATCACATGGCATAATGAAGTAAG




GTTGACCATACTCTACAGTTGTGTTTTAATGTATATTAATGTTAC




TAATGTGTTTT




(SEQ ID NO: 213)





TIM3
miR17
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATT




GTGACCAGTCAGAATAATGTCATTATGCCTGGGATTTGGATCGT




GATATGTGCATCGGCCAGAACCAAGGCATAAGTAGCATTATGG




TGACAGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGG




GCCTGCTGATGT




(SEQ ID NO: 214)





TIM3
miR126
CCGGGGTCCTGTCTGCATCCAGCGCAGCATTCTGGAAGACGCC




ACGCCTCCGCTGGCGACGGACTCCAAATCCCGTGCATAATCGCT




GTGACACTTCAAACCATTATGCCTGGGATTTGGATCCCGTCCAC




GGCACCGCATCGAAAACGCCGCTGAGACCTCAGCCTTGACCTC




CCTCAGCGTGG




(SEQ ID NO: 215)





TIM3
miR122
CAATGGTGGAATGTGGAGGTGAAGTTAACACCTTCGTGGCTAC




AGAGTTTCCTTAGCAGAGCTGCATTATGCCTGGGATTTGGATCT




GTCTAAACTATGATTCGAATCCAAGGCATATGGTAGCTACTGCT




AGGCAATCCTTCCCTCGATAAATGTCTTGGCATCGTTTGCTTTGA




GCAAGAAGGTTCATCTGATATCAGTCTTCTCAATCT




(SEQ ID NO: 216)





TIM3
miR214
ACAGGCTGATTGTATCTGTCTATGAGCAAAGGAAACCTGAAGG




AACCAAGGGCCTGGCTGGACAGAGTTGTCATGTGATGATGAAA




GGACAGTGAATTAGCAGAACATCCGCTCACCTGTTAATTCACAT




CCCTTTCATCAGCACATGACAACCCAGCCTGAATGACAACCAGC




CATTGAAAGAAAGCAGCCCTCACACCATAGCATCTA




(SEQ ID NO: 217)





TIM3
miR29b1
GGGTTTATTGTAAGAGAGCATTATGAAGAAAAAAATAGATCAT




AAAGCTTCTTCAGGACGATGGAAGGGTTGTGAACGTTAGATTT




AAATAGTGATTGTCTAATTCACATCCCTTTCATCAGTCTTGGGG




GAGACCAGCTGCGCTGCACTACCAACAGCAAAAGAAGTGAATG




GGACAGCT




(SEQ ID NO: 218)





TIM3
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTAATT




CACATCCCTTTCATCAGGAGAATATATGAAGGCTGAGGAAGAG




GTGTGAATTCGTTCAATTGTCATCACTGGCATCTTTTTTGATCAT




TGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 219)





TIM3
miR133a1
TTTACCAATGAAAAGCATTTAACTGTTTTGGATTCCAAACTAGC




AGCACTACAATGCTTTGCTAGTGATGAATAGGCTGCGAATTATC




GCCTCTTCAATGGATAATTCACATCCCTTTCATCAGTAGCTATGC




ATTGATTACTACGGGACAACCAACGTTTTCATTTGTGAATATCA




ATTACTTGCCA




(SEQ ID NO: 220)





LAG3
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACTAGTCGTTGGGTAAAGTCGCCACTGTGAAGC




CACAGATGGGTGGTGACTTCTCAACGATTAGCTGCCTACTGCCT




CGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 221)





LAG3
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGTGGCGATTTTACCCAATGCTCTATA




TGGATTACTTTGCTATAGTCGTTGGGTAAAGTCGCCATTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 222)





LAG3
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTAGT




CGTTGGGTAAAGTCGCCAGAGAATATATGAAGGTGGCACTTTC




CTCAACGACTCGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 223)





LAG3
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAG




CGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGTAGTCGTTGGG




TAAAGTCGCCACTGGGCTCAGACCGCGACTTACCACACGACTAC




AGGGACCTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTGA




GGTGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 224)





LAG3
miR17
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATT




GTGACCAGTCAGAATAATGTGTTGCTTTCCGCTAAGTGGTGAGT




GATATGTGCATCTCCCACTCAGAGGAAAGCACTAGCATTATGGT




GACAGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGGG




CCTGCTGATGT




(SEQ ID NO: 225)





LAG3
miR122
CAATGGTGGAATGTGGAGGTGAAGTTAACACCTTCGTGGCTAC




AGAGTTTCCTTAGCAGAGCTGGTTGCTTTCCGCTAAGTGGTGAT




GTCTAAACTATTCACCACTTAGAGGAAAGCCTCTAGCTACTGCT




AGGCAATCCTTCCCTCGATAAATGTCTTGGCATCGTTTGCTTTGA




GCAAGAAGGTTCATCTGATATCAGTCTTCTCAATCT




(SEQ ID NO: 226)





LAG3
miR126
CCGGGGTCCTGTCTGCATCCAGCGCAGCATTCTGGAAGACGCC




ACGCCTCCGCTGGCGACGGGAACCACTTAGCGCGAAGCAAGGC




TGTGACACTTCAAACGTTGCTTTCCGCTAAGTGGTGACCGTCCA




CGGCACCGCATCGAAAACGCCGCTGAGACCTCAGCCTTGACCT




CCCTCAGCGTGG




(SEQ ID NO: 227)





GITR
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACTTTGCAGTGGCCTTCGTGGCCCCTGTGAAGC




CACAGATGGGGGGTCACGAGCCACTGCGAAGCTGCCTACTGCC




TCGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 228)





GITR
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGGGGCTATGAAGGCCATTGAGAAAT




ATGGATTACTTTGCTATTTGCAGTGGCCTTCGTAGCCCTTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 229)





GITR
miR17
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATT




GTGACCAGTCAGAATAATGTTTTGCAGTGGCCTTCGTGGCCCGT




GATATGTGCATCGGTCACGTAGACTACTGCACTCGCATTATGGT




GACAGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGGG




CCTGCTGATGT




(SEQ ID NO: 230)





GITR
miR122
CAATGGTGGAATGTGGAGGTGAAGTTAACACCTTCGTGGCTAC




AGAGTTTCCTTAGCAGAGCTGTTTGCAGTGGCCTTCGTGGCCCT




GTCTAAACTATGGGTCACGAAGACCACTGCCCATAGCTACTGCT




AGGCAATCCTTCCCTCGATAAATGTCTTGGCATCGTTTGCTTTGA




GCAAGAAGGTTCATCTGATATCAGTCTTCTCAATCT




(SEQ ID NO: 231)





GITR
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAG




CGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGAGCCTCCCGTCC




TAAGACCCCACTGGGCTCAGACAGGGTCTTGGAAAGGAGGCTC




AGGGACCTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTGA




GGTGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 232)





GITR
miR29
GGGTTTATTGTAAGAGAGCATTATGAAGAAAAAAATAGATCAT




AAAGCTTCTTCAGGGTGGGTCTTAGGTCGGGAGCTGCTGATTT




AAATAGTGATTGTCAGCCTCCCGTCCTAAGACCCCACCTTGGGG




GAGACCAGCTGCGCTGCACTACCAACAGCAAAAGAAGTGAATG




GGACAGCT




(SEQ ID NO: 233)





GITR
miR181a1
TCTCCCATCCCCTTCAGATACTTACAGATACTGTAAAGTGAGTA




GAATTCTGAGTTTTGAGGTTGCTTCAGTGAGCCTCCCGTCCTAA




GACCCCACTTGGAATTAAAATCAAGTGGGTCTTGACGGGTGGC




ACCCTATGGCTAACCATCATCTACTCCATGGTGCTCAGAATTCG




CTGAAGACAGGAAACCAAAGGTGGACACACCAGG




(SEQ ID NO: 234)





TIGIT
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCCAGGGTGAGTAACGTGGCCTCTT




ATGGATTACTTTGCTAAGATCCACGTTACTCACCCTGGTTTCGG




CAAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAG




GTTTGGTGACCTT




(SEQ ID NO: 235)





TIGIT
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACAGATCCACGTTACTCACCCAAGCTGTGAAGC




CACAGATGGGCTTGGGTGAGACGTGGATCTGCTGCCTACTGCC




TCGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 236)





TIGIT
miR133a1
TTTACCAATGAAAAGCATTTAACTGTTTTGGATTCCAAACTAGC




AGCACTACAATGCTTTGCTAGAGGGGTGTATACCGCGGATCTTC




GCCTCTTCAATGGAAGATCCACGTTACTCACCCCTGTAGCTATG




CATTGATTACTACGGGACAACCAACGTTTTCATTTGTGAATATC




AATTACTTGCCA




(SEQ ID NO: 237)





TIGIT
miR122
CAATGGTGGAATGTGGAGGTGAAGTTAACACCTTCGTGGCTAC




AGAGTTTCCTTAGCAGAGCTGAGATCCACGTTACTCACCCTTGT




GTCTAAACTATCAAGGGTGAGTCACGTGGAGATTAGCTACTGC




TAGGCAATCCTTCCCTCGATAAATGTCTTGGCATCGTTTGCTTTG




AGCAAGAAGGTTCATCTGATATCAGTCTTCTCAATCT




(SEQ ID NO: 238)





TIGIT
miR29b1
GGGTTTATTGTAAGAGAGCATTATGAAGAAAAAAATAGATCAT




AAAGCTTCTTCAGGAAAGATCCACGTTACTCACCCTTAGATTTA




AATAGTGATTGTCTAGGTGAGTTACGTGGATCTGTTCTTGGGG




GAGACCAGCTGCGCTGCACTACCAACAGCAAAAGAAGTGAATG




GGACAGCT




(SEQ ID NO: 239)





TIGIT
miR214
ACAGGCTGATTGTATCTGTCTATGAGCAAAGGAAACCTGAAGG




AACCAAGGGCCTGGCTGGACAGAGTTGTCATGTGTCAGATCCA




CGTTACTCACCCTGCAGAACATCCGCTCACCTGTAGGGTGAGAC




TCGTGGATCTGTCACATGACAACCCAGCCTGAATGACAACCAGC




CATTGAAAGAAAGCAGCCCTCACACCATAGCATCTA




(SEQ ID NO: 240)





PD1
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACTTCAGGAATGGGTTCCAAGGAGCTGTGAAGC




CACAGATGGGCTCCTTGGACCATTCCTGAAGCTGCCTACTGCCT




CGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 241)





PD1
miR412
GTCTTGGAGGCTGGGGCACCTCGGGGAAGGACGCCGGCATCA




GCACCATTCTGGGGTACGGGGATGGATTCAGGAATGGGTTCCA




AGGAATTGTTTCTAATGTCCTGAAGCCATTCATGGAGCCGTCCG




TATCCGCTGCAGCCTGTGGGGCCTGCGGGCCGGGGAGCCGATC




GCGCTTCAGCTCAGCGCCT




(SEQ ID NO: 242)





PD1
miR17
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATT




GTGACCAGTCAGAATAATGTTTCAGGAATGGGTTCCAAGGAAG




TGATATGTGCATCTTCTTGGTACACGTTCCTGCTCACATTATGGT




GACAGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGGG




CCTGCTGATGT




(SEQ ID NO: 243)





PD1
miR122
CAATGGTGGAATGTGGAGGTGAAGTTAACACCTTCGTGGCTAC




AGAGTTTCCTTAGCAGAGCTGTTCAGGAATGGGTTCCAAGGAG




TGTCTAAACTATCTCCTTGGAACACATTCCTACATAGCTACTGCT




AGGCAATCCTTCCCTCGATAAATGTCTTGGCATCGTTTGCTTTGA




GCAAGAAGGTTCATCTGATATCAGTCTTCTCAATCT




(SEQ ID NO: 244)





PD1
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAG




CGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGTATAATATAATA




GAACCACAGGCTGGGCTCAGACGTGTGGTTTATCCTGTTGTAC




AGGGACCTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTGA




GGTGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 245)





PD1
miR486
TGTGGTGCTGGGGGCTTCAGCGGCCGGCTCTGATCTCCATCCTC




CCTGGGGCATATAATATAATAGAACCACAGGGCCCTTCATGCTG




CCCAGCTTGTGGTTCTATTATGTTATAACTCGGGGTGGGAGTCA




GCAGGAGGTGAGGGGGCATGGTGGCCCCAGTGCAGC




(SEQ ID NO: 246)





PD1
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCGTGCTGAACTGGTATCGAAATGT




ATGGATTACTTTGCTACATGCGGTACCAGTTTAGCACGTTTCGG




CAAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAG




GTTTGGTGACCTT




(SEQ ID NO: 247)





PD1
miR122
CAATGGTGGAATGTGGAGGTGAAGTTAACACCTTCGTGGCTAC




AGAGTTTCCTTAGCAGAGCTGCATGCGGTACCAGTTTAGCACGT




GTCTAAACTATCGTGCTAGACTTGTACTGTCAGTAGCTACTGCT




AGGCAATCCTTCCCTCGATAAATGTCTTGGCATCGTTTGCTTTGA




GCAAGAAGGTTCATCTGATATCAGTCTTCTCAATCT




(SEQ ID NO: 248)





PD1
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACCATGCGGTACCAGTTTAGCACGCTGTGAAGC




CACAGATGGGCGTGCTAGAGGTGTCGCATGGCTGCCTACTGCC




TCGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 249)





CTLA4
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCAGCGAGGGAGAAGATTACCATTT




ATGGATTACTTTGCTAAATATAGTCTTCTCCCTCGCTGTTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 250)





CTLA4
miR26a
TGAAGCCACAGGAGCCAAGAGCAGGAGGACCAAGGCCCTGGC




GAAGGCCGTGGCCTCGAATATAGTCTTCTCCCTCGCTTGTGCAG




GTCCCAATGGAGCGAGGGATAGGACTATACTCGGGGACGCGG




GCCTGGACGCCGGCATCCGGGCTCAGGACCCCCCTCTCTGCCA




GAGGC




(SEQ ID NO: 251)





CTLA4
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACAATATAGTCTTCTCCCTCGCTGCTGTGAAGCC




ACAGATGGGCAGTGAGGGAAGACTATGTTGCTGCCTACTGCCT




CGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 252)





PIK3IP1
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCCGGTTGAACATTGGTTGCCTCATA




TGGATTACTTTGCTATGAACGACCAGTGTTTAACCGGTTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 253)





PIK3IP1
miR126
CCGGGGTCCTGTCTGCATCCAGCGCAGCATTCTGGAAGACGCC




ACGCCTCCGCTGGCGACGGGACGGATGAACTTAGAGGAGACG




CTGTGACACTTCAAACTTCTCCTTGGAGTTCATCCGCGCCGTCCA




CGGCACCGCATCGAAAACGCCGCTGAGACCTCAGCCTTGACCT




CCCTCAGCGTGG




(SEQ ID NO: 254)





PIK3IP1
miR30a
CAGGTTAACCCAACAGAAGGCTAAAGAAGGTATATTGCTGTTG




ACAGTGAGCGACTTCTCCTTGGAGTTCATCCGCGCTGTGAAGCC




ACAGATGGGCGCGGATGATCCAAGGAGGAGCTGCCTACTGCCT




CGGACTTCAAGGGGCTACTTTAGGAGCAATTATCTTGTTTA




(SEQ ID NO: 255)





TCRa3′UTR
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAG




CGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGATACACATCAGA




ATCCTTACTGCTGGGCTCAGACCGTAAGGATCTCCTGTGTATCA




GGGACCTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTGAG




GCGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 256)





TCRa3′UTR
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGC




GTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACATAC




ACATCAGAATCCTTACTTGAGAATATATGAAGGAGGTAGGATA




CTGATGTGTACGTTCAATTGTCATCACTGGCATCTTTTTTGATCA




TTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 257)





TCRa3′UTR
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCGGTAAGGATTCTGATGTAATATTA




TGGATTACTTTGCTAATACACATCAGAATCCTTACTGTTTCGGCA




AGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGT




TTGGTGACCTT




(SEQ ID NO: 258)





TCRa3′UTR
miR26a
TGAAGCCACAGGAGCCAAGAGCAGGAGGACCAAGGCCCTGGC




GAAGGCCGTGGCCTCGATACACATCAGAATCCTTACTTGTGCAG




GTCCCAATGGAGTAAGGATCCTGATGTGTCTCGGGGACGCGGG




CCTGGACGCCGGCATCCGGGCTCAGGACCCCCCTCTCTGCCAG




AGGC




(SEQ ID NO: 259)





TCRa3′UTR
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAG




CGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGTTGTTGAAGGC




GTTTGCACATGCTGGGCTCAGACCTGTGCAATGCGATCAATAG




CAGGGACCTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTG




AGGCGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 260)





TCRa3′UTR
miR16
CTTCTGAAGAAAATATATTTCTTTTTATTCATAGCTCTTATGATA




GCAATGTCAGCAGTGCCTTTGTTGAAGGCGTTTGCACATGTTAA




GATTCTAAAATTATCTTGTGCGGAAGCACTTCAATAAAAGTAAG




GTTGACCATACTCTACAGTTGTGTTTTAATGTATATTAATGTTAC




TAATGTGTTTT




(SEQ ID NO: 261)





TCRa3′UTR
miR206
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGG




ATGACACTGCTTCCCGAGGCGTGTGTGAACGCCTTCACATAATA




TGGATTACTTTGCTATTGTTGAAGGCGTTTGCACATGTTTCGGC




AAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTT




(SEQ ID NO: 262)





TCRa3′UTR
miR26a
TGAAGCCACAGGAGCCAAGAGCAGGAGGACCAAGGCCCTGGC




GAAGGCCGTGGCCTCGTTGTTGAAGGCGTTTGCACATTGTGCA




GGTCCCAATGGGTGTGCAAAGGTCTTCAATCACGGGGACGCGG




GCCTGGACGCCGGCATCCGGGCTCAGGACCCCCCTCTCTGCCA




GAGGC




(SEQ ID NO: 263)



















Non-naturally occurring miRNA sequences (two or more pri-miRNAs)









miRNA
miRNA



Target
backbone
DNA Sequence





PD1
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC



miR206
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCC




AAGGATGAGAATATATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACGATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTT




CTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATG




GATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCT




CCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 267)





PD1 + TIGIT
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC



miR150
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCC




AAGGATGAGAATATATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACAGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGC




AGCAGCGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGAGATCCACGTTA




CTCACCCGTGCTGGGCTCAGACCCGGGTGATAATATGGATCTCAGGGAC




CTGGGGACCCCGGCACCGGCAGGCCCCAAGGGGTGAGGTGAGCGGGCA




TTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 268)





PD1 + TIGIT
miR206 +
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGGATGACA



miR150
CTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATGGATTACTTTGC




TATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCTCCTCGCTGGCC




CCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTTAGGGACTGGGCCC




ACGGGGAGGCAGCGTCCCCGAGGCAGCAGCGGCAGCGGCGGCTCCTCT




CCCCATGGCCCTGAGATCCACGTTACTCACCCGTGCTGGGCTCAGACCCG




GGTGATAATATGGATCTCAGGGACCTGGGGACCCCGGCACCGGCAGGC




CCCAAGGGGTGAGGTGAGCGGGCATTGGGACCTCCCCTCCCTGTACTC




(SEQ ID NO: 269)





TIGIT + PD1
miR17 +
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACC



miR204
AGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCT




AGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAG




CCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTAGGAGGGTGGGG




GTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCCATGGCTACAGTCT




TTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCCAAGGATGAGAATA




TATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATTGTCATCACTGGCA




TCTTTTTTGATCATTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 270)





TIGIT + PD1
miR17 +
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACC



miR206
AGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCT




AGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAG




CCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTGATGCTACAAGTG




GCCCACTTCTGAGATGCGGGCTGCTTCTGGATGACACTGCTTCCCGAGGC




CTGTGGTTCTGTTATATCCATATATGGATTACTTTGCTATATAATATAATA




GAACCACAGGTTTCGGCAAGTGCCTCCTCGCTGGCCCCAGGGTACCACCC




GGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 271)





TIGIT + PD1
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC



miR206
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACAGATCCACGTTACTCAC




CCCCTGAGAATATATGAAGGAGGGGTGAGAAGCGTGGATCCGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACGATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTT




CTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATG




GATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCT




CCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 272)





TIGIT + PD1
miR150 +
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAGCGGCA



miR204
GCGGCGGCTCCTCTCCCCATGGCCCTGAGATCCACGTTACTCACCCGTGC




TGGGCTCAGACCCGGGTGATAATATGGATCTCAGGGACCTGGGGACCCC




GGCACCGGCAGGCCCCAAGGGGTGAGGTGAGCGGGCATTGGGACCTCC




CCTCCCTGTACTCAGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCT




GATCGCGTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCA




GGAATGGGTTCCAAGGATGAGAATATATGAAGGATCCTGGAAGCTATTC




CTGACGTTCAATTGTCATCACTGGCATCTTTTTTGATCATTGCACCATCAT




CAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 273)





TIGIT + PD1
miR150 +
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGCAGCGGCA



miR206
GCGGCGGCTCCTCTCCCCATGGCCCTGAGATCCACGTTACTCACCCGTGC




TGGGCTCAGACCCGGGTGATAATATGGATCTCAGGGACCTGGGGACCCC




GGCACCGGCAGGCCCCAAGGGGTGAGGTGAGCGGGCATTGGGACCTCC




CCTCCCTGTACTCGATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGC




TTCTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATAT




GGATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCAAGTGCC




TCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 274)





TIGIT +
miR17 +
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACC


PD1 + PD1
miR204 +
AGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCT



miR206
AGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAG




CCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTAGGAGGGTGGGG




GTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCCATGGCTACAGTCT




TTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCCAAGGATGAGAATA




TATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATTGTCATCACTGGCA




TCTTTTTTGATCATTGCACCATCATCAAATGCATTGGGATAACCATGACGA




TGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGGATGACACTG




CTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATGGATTACTTTGCTAT




ATAATATAATAGAACCACAGGTTTCGGCAAGTGCCTCCTCGCTGGCCCCA




GGGTACCACCCGGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 275)





TIGIT +
miR17 +
TTAGGATGAGTTGAGATCCCAGTGATCTTCTCGCTAAGAGTTTCCTGCCT


PD1 + PD1
miR204 +
GGGCAAGGAGGAAATTAGCAGGAAAAAAGAGAACATCACCTTGTAAAA



miR206 extra
CTGAAGATTGTGACCAGTCAGAATAATGTAGATCCACGTTACTCACCCTA



spacing 1
GTGATATGTGCATCTAGGGTGTGTCATGTGGATGAAGCATTATGGTGAC




AGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGAT




GTAGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTAC




CCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCAGGAATGGGTT




CCAAGGATGAGAATATATGAAGGATCCTGGAAGCTATTCCTGACGTTCA




ATTGTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATT




GGGATAACCATGACGATGCTACAAGTGGCCCACTTCTGAGATGCGGGCT




GCTTCTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATAT




ATGGATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCAAGTG




CCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTT




CTTCCTCATCAGGGCTTTGTGCCAGCAAATGACTCCCTCACCAAGGAAGC




AAGAGCCTCTGAATCCCATCTGGGCTCTTCCTGAACACCCCTATCTCCCCC




TCT




(SEQ ID NO: 276)





TIGIT +
miR17 +
TTAGGGATTATGCTGAATTTGTATGGTTTATAGTTGTTAGAGTTTGAGGT


PD1 + PD1
miR204 +
GTTAATTCTAATTATCTATTTCAAATTTTAGCAGGAAAAAAGAGAACATC



miR206 extra
ACCTTGTAAAACTGAAGATTGTGACCAGTCAGAATAATGTAGATCCACGT



spacing 2
TACTCACCCTAGTGATATGTGCATCTAGGGTGTGTCATGTGGATGAAGCA




TTATGGTGACAGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGG




GCCTGCTGATGTAGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCT




GATCGCGTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCA




GGAATGGGTTCCAAGGATGAGAATATATGAAGGATCCTGGAAGCTATTC




CTGACGTTCAATTGTCATCACTGGCATCTTTTTTGATCATTGCACCATCAT




CAAATGCATTGGGATAACCATGACGATGCTACAAGTGGCCCACTTCTGA




GATGCGGGCTGCTTCTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGT




TATATCCATATATGGATTACTTTGCTATATAATATAATAGAACCACAGGTT




TCGGCAAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGT




TTGGTGACCTTCTTCCTCATCAGGGCTTTGTGCCAGCAAATGACTCCCTCA




CCAAGGAAGCAAGAGCCTCTGAATCCCATCTGGGCTCTTCCTGAACACCC




CTATCTCCCCCTCT




(SEQ ID NO: 277)





PD1 + PD1 +
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC


TIGIT
miR206 +
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCC



miR17
AAGGATGAGAATATATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACGATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTT




CTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATG




GATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCT




CCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTTTTA




GCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACCAGT




CAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCTAGG




GTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAGCCA




AGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGT




(SEQ ID NO: 278)





PD1 + PD1 +
miR204 +
TTAGGATGAGTTGAGATCCCAGTGATCTTCTCGCTAAGAGTTTCCTGCCT


TIGIT
miR206 +
GGGCAAGGAGGAAAAGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTT



miR17 extra
CCTGATCGCGTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTT



spacing 1
CAGGAATGGGTTCCAAGGATGAGAATATATGAAGGATCCTGGAAGCTAT




TCCTGACGTTCAATTGTCATCACTGGCATCTTTTTTGATCATTGCACCATC




ATCAAATGCATTGGGATAACCATGACGATGCTACAAGTGGCCCACTTCTG




AGATGCGGGCTGCTTCTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTG




TTATATCCATATATGGATTACTTTGCTATATAATATAATAGAACCACAGGT




TTCGGCAAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGG




TTTGGTGACCTTTTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGA




AGATTGTGACCAGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGA




TATGTGCATCTAGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCT




GCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTCT




TCCTCATCAGGGCTTTGTGCCAGCAAATGACTCCCTCACCAAGGAAGCAAG




AGCCTCTGAATCCCATCTGGGCTCTTCCTGAACACCCCTATCTCCCCCTCT




(SEQ ID NO: 279)





PD1 + PD1 +
miR204 +
TGGAGAGGAGGGTGGGGGTGGAGGCAAGCAGAGGACCTCCTGATCAT


TIGIT
miR206 +
GTACCCATAGGACAGGGTGATGGAAAGGAGGGTGGGGGTGGAGGCAA



miR17 extra
GCAGAGGACTTCCTGATCGCGTACCCATGGCTACAGTCTTTCTTCATGTG



spacing 2
ACTCGTGGACTTCAGGAATGGGTTCCAAGGATGAGAATATATGAAGGAT




CCTGGAAGCTATTCCTGACGTTCAATTGTCATCACTGGCATCTTTTTTGAT




CATTGCACCATCATCAAATGCATTGGGATAACCATGACGATGCTACAAGT




GGCCCACTTCTGAGATGCGGGCTGCTTCTGGATGACACTGCTTCCCGAG




GCCTGTGGTTCTGTTATATCCATATATGGATTACTTTGCTATATAATATAA




TAGAACCACAGGTTTCGGCAAGTGCCTCCTCGCTGGCCCCAGGGTACCAC




CCGGAGCACAGGTTTGGTGACCTTTTAGCAGGAAAAAAGAGAACATCAC




CTTGTAAAACTGAAGATTGTGACCAGTCAGAATAATGTAGATCCACGTTA




CTCACCCTAGTGATATGTGCATCTAGGGTGTGTCATGTGGATGAAGCATT




ATGGTGACAGCTGCCTCGGGAAGCCAAGTTGGGCTTTAAAGTGCAGGGC




CTGCTGATGTCTTCCTCATCAGGGCTTTGTGCCAGCAAATGACTCCCTCAC




CAAGGAAGCAAGAGCCTCTGAATCCCATCTGGGCTCTTCCTGAACACCCC




TATCTCCCCCTCT




(SEQ ID NO: 280)





PD1 + CTLA4
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC



miR26a
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCC




AAGGATGAGAATATATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACTGAAGCCACAGGAGCCAAGAGCAGGAGGACCAAGGCC




CTGGCGAAGGCCGTGGCCTCGAATATAGTCTTCTCCCTCGCTTGTGCAGG




TCCCAATGGAGCGAGGGATAGGACTATACTCGGGGACGCGGGCCTGGA




CGCCGGCATCCGGGCTCAGGACCCCCCTCTCTGCCAGAGGC




(SEQ ID NO: 281)





PD1 + PD1
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC



miR206
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCC




AAGGATGAGAATATATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACGATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTT




CTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATG




GATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCT




CCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 282)





PD1 + CTLA4
miR206 +
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGGATGACA



miR26a
CTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATGGATTACTTTGC




TATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCTCCTCGCTGGCC




CCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTTTGAAGCCACAGGA




GCCAAGAGCAGGAGGACCAAGGCCCTGGCGAAGGCCGTGGCCTCGAAT




ATAGTCTTCTCCCTCGCTTGTGCAGGTCCCAATGGAGCGAGGGATAGGA




CTATACTCGGGGACGCGGGCCTGGACGCCGGCATCCGGGCTCAGGACCC




CCCTCTCTGCCAGAGGC




(SEQ ID NO: 283)





PD1 + CTLA4
miR206 +
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCTGGATGACA



miR204
CTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATGGATTACTTTGC




TATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCTCCTCGCTGGCC




CCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTTAGGAGGGTGGGG




GTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCCATGGCTACAGTCT




TTCTTCATGTGACTCGTGGACAATATAGTCTTCTCCCTCGCTTGAGAATAT




ATGAAGGAAGCAGGGACAGGACTATATTGTTCAATTGTCATCACTGGCA




TCTTTTTTGATCATTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 284)





TIGIT +
miR17 +
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACC


CTLA4
miR26a
AGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCT




AGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAG




CCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTTGAAGCCACAGGA




GCCAAGAGCAGGAGGACCAAGGCCCTGGCGAAGGCCGTGGCCTCGAAT




ATAGTCTTCTCCCTCGCTTGTGCAGGTCCCAATGGAGCGAGGGATAGGA




CTATACTCGGGGACGCGGGCCTGGACGCCGGCATCCGGGCTCAGGACCC




CCCTCTCTGCCAGAGGC




(SEQ ID NO: 285)





TIGIT +
miR17 +
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACC


CTLA4
miR204
AGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCT




AGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAG




CCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTAGGAGGGTGGGG




GTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCCATGGCTACAGTCT




TTCTTCATGTGACTCGTGGACAATATAGTCTTCTCCCTCGCTTGAGAATAT




ATGAAGGAAGCAGGGACAGGACTATATTGTTCAATTGTCATCACTGGCA




TCTTTTTTGATCATTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 286)





TIGIT + PD1
miR17 +
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACC



miR204
AGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCT




AGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAG




CCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTAGGAGGGTGGGG




GTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCCATGGCTACAGTCT




TTCTTCATGTGACTCGTGGACTTCAGGAATGGGTTCCAAGGATGAGAATA




TATGAAGGATCCTGGAAGCTATTCCTGACGTTCAATTGTCATCACTGGCA




TCTTTTTTGATCATTGCACCATCATCAAATGCATTGGGATAACCATGAC




(SEQ ID NO: 287)





TIGIT + PD1
miR17 +
TTAGCAGGAAAAAAGAGAACATCACCTTGTAAAACTGAAGATTGTGACC



206
AGTCAGAATAATGTAGATCCACGTTACTCACCCTAGTGATATGTGCATCT




AGGGTGTGTCATGTGGATGAAGCATTATGGTGACAGCTGCCTCGGGAAG




CCAAGTTGGGCTTTAAAGTGCAGGGCCTGCTGATGTGATGCTACAAGTG




GCCCACTTCTGAGATGCGGGCTGCTTCTGGATGACACTGCTTCCCGAGGC




CTGTGGTTCTGTTATATCCATATATGGATTACTTTGCTATATAATATAATA




GAACCACAGGTTTCGGCAAGTGCCTCCTCGCTGGCCCCAGGGTACCACCC




GGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 288)





TIGIT +
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC


CTLA4
miR26a
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACAGATCCACGTTACTCAC




CCCCTGAGAATATATGAAGGAGGGGTGAGAAGCGTGGATCCGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACTGAAGCCACAGGAGCCAAGAGCAGGAGGACCAAGGCC




CTGGCGAAGGCCGTGGCCTCGAATATAGTCTTCTCCCTCGCTTGTGCAGG




TCCCAATGGAGCGAGGGATAGGACTATACTCGGGGACGCGGGCCTGGA




CGCCGGCATCCGGGCTCAGGACCCCCCTCTCTGCCAGAGGC




(SEQ ID NO: 289)





TIGIT + PD1
miR204 +
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATCGCGTACCC



miR206
ATGGCTACAGTCTTTCTTCATGTGACTCGTGGACAGATCCACGTTACTCAC




CCCCTGAGAATATATGAAGGAGGGGTGAGAAGCGTGGATCCGTTCAATT




GTCATCACTGGCATCTTTTTTGATCATTGCACCATCATCAAATGCATTGGG




ATAACCATGACGATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTT




CTGGATGACACTGCTTCCCGAGGCCTGTGGTTCTGTTATATCCATATATG




GATTACTTTGCTATATAATATAATAGAACCACAGGTTTCGGCAAGTGCCT




CCTCGCTGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTT




(SEQ ID NO: 290)









Splice donor and splice acceptor site sequences















5′ side of intron
AGGTAAGAGTCGATCG (SEQ ID NO: 291)


(SEQ ID NO: 291)






3′ side of intron
ACGCGTTACTAACTGGTACCTCTTTTTTTTTT


(SEQ ID NO: 292)
TTGATATCCTGCAGGC (SEQ ID NO: 292)









Additional miRNA Sequences














SEQ



ID


Mature miRNA_Length (Sequence)
NO:







Guide PD1_204_21nt (TTCAGGAATGGGTTCCAAGGA)
704





Guide PD1_204_23nt (TTCAGGAATGGGTTCCAAGGATG)
705





Passenger PD1_204_21nt (TCCTGGAAGCTATTCCTGACG)
706





Passenger PD1_204_22nt (TCCTGGAAGCTATTCCTGACG
707


T)






Passenger PD1_204_23nt (TCCTGGAAGCTATTCCTGACG
708


TT)






Guide PD1_206_21nt (TATAATATAATAGAACCACAG)
709





Guide PD1_206_23nt (TATAATATAATAGAACCACAGGA)
710





Passenger PD1_206_21nt (TGTGGTTCTGTTATATCCATA)
711





Passenger PD1_206_22nt (TGTGGTTCTGTTATATCCATA
712


T)






Passenger PD1_206_23nt (TGTGGTTCTGTTATATCCATA
713


TA)









Exemplary anti-CD33 VH and VL Sequences

















SEQ
Amino Acid
SEQ



Name
ID NO
Sequence
ID NO
Nucleotide Sequence







hM195 VL
293
DIQMTQSPSSLSASVG
301
GACATTCAGATGACCCAGTCTCCGAGCTCTCTGTC




DRVTITCRASESVDNY

CGCATCAGTAGGAGACAGGGTCACCATCACATGC




GISFMNWFQQKPGKA

AGAGCCAGCGAAAGTGTCGACAATTATGGCATTA




PKLLIYAASNQGSGVPS

GCTTTATGAACTGGTTCCAACAGAAACCCGGGAA




RFSGSGSGTDFTLTISSL

GGCTCCTAAGCTTCTGATTTACGCTGCATCCAACC




QPDDFATYYCQQSKEV

AAGGCTCCGGGGTACCCTCTCGCTTCTCAGGCAG




PWTFGQGTKVEIK

TGGATCTGGGACAGACTTCACTCTCACCATTTCAT






CTCTGCAGCCTGATGACTTCGCAACCTATTACTGT






CAGCAAAGTAAGGAGGTTCCGTGGACGTTCGGTC






AAGGGACCAAGGTGGAGATCAAA





hM195 VH
294
QVQLVQSGAEVKKPGS
302
CAGGTTCAGCTGGTGCAGTCTGGAGCTGAGGTG




SVKVSCKASGYTFTDY

AAGAAGCCTGGGAGCTCAGTGAAGGTTTCCTGCA




NMHWVRQAPGQGLE

AAGCTTCTGGCTACACCTTCACTGACTACAACATG




WIGYIYPYNGGTGYNQ

CACTGGGTGAGGCAGGCTCCTGGCCAAGGCCTG




KFKSKATITADESTNTA

GAATGGATTGGATATATTTATCCTTACAATGGTG




YMELSSLRSEDTAVYYC

GTACCGGCTACAACCAGAAGTTCAAGAGCAAGG




ARGRPAMDYWGQGT

CCACAATTACAGCAGACGAGAGTACTAACACAGC




LVTVSS

CTACATGGAACTCTCCAGCCTGAGGTCTGAGGAC






ACTGCAGTCTATTACTGCGCAAGAGGGCGCCCCG






CTATGGACTACTGGGGCCAAGGGACTCTGGTCAC






TGTCTCTTCA





M2H12 VH
295
QVQLQQSGPELVRPGT






FVKISCKASGYTFTNYDI






NWVNQRPGQGLEWI






GWIYPGDGSTKYNEKF






KAKATLTADKSSSTAYL






QLNNLTSENSAVYFCA






SGYEDAMDYWGQGT






SVTVSS







M2H12 VL
296
DIKMTQSPSSMYASLG






ERVIINCKASQDINSYLS






WFQQKPGKSPKTLIYR






ANRLVDGVPSRFSGSG






SGQDYSLTISSLEYEDM






GIYYCLQYDEFPLTFGA






GTKLELKR







DRB2 VH
297
EVKLQESGPELVKPGAS






VKMSCKASGYKFTDYV






VHWLKQKPGQGLEWI






GYINPYNDGTKYNEKF






KGKATLTSDKSSSTAY






MEVSSLTSEDSAVYYC






ARDYRYEVYGMDYWG






QGTSVTVSS







DRB2 VL
298
DIVLTQSPTIMSASPGE






RVTMTCTASSSVNYIH






WYQQKSGDSPLRWIF






DTSKVASGVPARFSGS






GSGTSYSLTISTMEAED






AATYYCQQWRSYPLTF






GDGTRLELKRADAAPT






VS







My9-6 VH
299
QVQLQQPGAEVVKPG






ASVKMSCKASGYTFTS






YYIHWIKQTPGQGLEW






VGVIYPGNDDISYNQK






FKGKATLTADKSSTTAY






MQLSSLTSEDSAVYYC






AREVRLRYFDVWGAG






TTVTVSS







My9-6 VL
300
NIMLTQSPSSLAVSAG






EKVTMSCKSSQSVFFSS






SQKNYLAWYQQIPGQ






SPKLLIYWASTRESGVP






DRFTGSGSGTDFTLTIS






SVQSEDLAIYYCHQYLS






SRTFGGGTKLEIKR







hM195
303
DIQMTQSPSSLSA
304
GACATTCAGATGACCCAGTCTCCGAGCTCTCTGTC


scFv

SVGDRVTITCRAS

CGCATCAGTAGGAGACAGGGTCACCATCACATGC




ESVDNYGISFMN

AGAGCCAGCGAAAGTGTCGACAATTATGGCATTA




WFQQKPGKAPKL

GCTTTATGAACTGGTTCCAACAGAAACCCGGGAA




LIYAASNQGSGVP

GGCTCCTAAGCTTCTGATTTACGCTGCATCCAACC




SRFSGSGSGTDFT

AAGGCTCCGGGGTACCCTCTCGCTTCTCAGGCAG




LTISSLQPDDFATY

TGGATCTGGGACAGACTTCACTCTCACCATTTCAT




YCQQSKEVPWTF

CTCTGCAGCCTGATGACTTCGCAACCTATTACTGT




GQGTKVEIKGGGG

CAGCAAAGTAAGGAGGTTCCGTGGACGTTCGGTC




SGGGGSGGGGSQ

AAGGGACCAAGGTGGAGATCAAAGGTGGCGGTG




VQLVQSGAEVKKP

GCTCGGGCGGTGGTGGGTCGGGTGGCGGCGGAT




GSSVKVSCKASGY

CTCAGGTTCAGCTGGTGCAGTCTGGAGCTGAGGT




TFTDYNMHWVRQ

GAAGAAGCCTGGGAGCTCAGTGAAGGTTTCCTGC




APGQGLEWIGYIY

AAAGCTTCTGGCTACACCTTCACTGACTACAACAT




PYNGGTGYNQKF

GCACTGGGTGAGGCAGGCTCCTGGCCAAGGCCT




KSKATITADESTNT

GGAATGGATTGGATATATTTATCCTTACAATGGT




AYMELSSLRSEDT

GGTACCGGCTACAACCAGAAGTTCAAGAGCAAG




AVYYCARGRPAM

GCCACAATTACAGCAGACGAGAGTACTAACACAG




DYWGQGTLVTVSS

CCTACATGGAACTCTCCAGCCTGAGGTCTGAGGA






CACTGCAGTCTATTACTGCGCAAGAGGGCGCCCC






GCTATGGACTACTGGGGCCAAGGGACTCTGGTCA






CTGTCTCTTCA









Exemplary anti-MUC1 VH and VL Sequences















SEQ ID



Name
NO
Amino Acid Sequence







Anti-MUC1
305
QVQLVQSGAEVKKPGSSVKVSCKASGYAFSNFWMNWVRQAPGQGLEWMGQI


VH1

YPGDGDTNYNGKFKGRVTITADKSTSTAYMELSSLRSEDTAVYYCARSYYRSAWFA




YWGQGTLVTVSS





Anti-MUC1
306
QVQLVQSGAEVKKPGASVKVSCKASGYAFSNFWMNWVRQAPGQGLEWMGQI


VH2

YPGDGDTNYNGKFKGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARSYYRSAWF




AYWGQGTLVTVSS





Anti-MUC1
307
QVQLVQSGAEVKKPGASVKVSCKASGYAFSNFWMNWVRQAPGQGLEWMGQI


VH3

YPGDGDTNYNGKFKGRVTMTRDTSISTAYMELSRLRSDDTAVYYCARSYYRSAWF




AYWGQGTLVTVSS





Anti-MUC1
308
QVQLVQSGAEVKKPGATVKISCKVSGYAFSNFWMNWVQQAPGKGLEWMGQIY


VH4

PGDGDTNYNGKFKGRVTITADTSTDTAYMELSSLRSEDTAVYYCARSYYRSAWFAY




WGQGTLVTVSR





Anti-MUC1
309
EVQLVQSGAEVKKPGESLKISCKGSGYAFSNFWMNWVRQMPGKGLEWMGQIY


VH5

PGDGDTNYNGKFKGQVTISADKSISTAYLQWSSLKASDTAMYYCARSYYRSAWFA




YWGQGTLVTVSL





Anti-MUC1
310
EIVLTQSPDFQSVTPKEKVTITCRASQSIGTSIHWYQQKPDQSPKLLIKYASESISGVP


VL1

SRFSGSGSGTDFTLTINSLEAEDAATYYCQQSNNWPLTFGQGTKVEIK





Anti-MUC1
311
EIVMTQSPATLSVSPGERATLSCRASQSIGTSIHWYQQKPGQAPRLLIYYASESISGI


VL2

PARFSGSGSGTEFTLTISSLQSEDFAVYYCQQSNNWPLTFGGGTKVEIK





Anti-MUC1
312
EIVLTQSPATLSLSPGERATLSCRASQSIGTSIHWYQQKPGQAPRLLIYYASESISGIP


VL3

ARFSGSGSGTDFTLTISSLEPEDFAVYYCQQSNNWPLTFGGGTKVEIK





Anti-MUC1
313
AIQLTQSPSSLSASVGDRVTITCRASQSIGTSIHWYQQKPGKAPKLLIYYASESISGVP


VL4

SRFSGSGSGTDFTLTISSLQPEDFATYYCQQSNNWPLTFGGGTKVEIK





Anti-MUC1
314
DIVMTQSPSLLSASTGDRVTISCRASQSIGTSIHWYQQKPGKAPELLIYYASESISGV


VL5

PSRFSGSGSGTDFTLTISCLQSEDFATYYCQQSNNWPLTFGQGTKVEIK









Exemplary anti-MUC16 Sequences

















SEQ
Amino Acid
SEQ



Name
ID NO
Sequence
ID NO
Nucleotide Sequence







Anti-
315
QVQLQESGPGLVKPS
316
CAGGTGCAACTGCAGGAATCAGGTCCAGGCTTG


MUC16

QTLSLTCTVSGYSIVSH

GTCAAGCCATCGCAGACTCTTAGTCTGACATGCA


VH1

YYWSWIRQHPGKGLE

CCGTGAGTGGCTATAGCATCGTGTCGCACTATTA




WIGYISSDGSNYYNPSL

TTGGTCTTGGATCAGGCAGCATCCAGGAAAGGG




KSLVTISVDTSKNQFSL

ACTGGAGTGGATCGGGTACATTAGCAGCGATGG




KLSSVTAADTAVYYCV

GAGCAACTATTACAACCCATCTCTGAAGTCCCTGG




RGVDYWGQGTMVTV

TAACTATTAGCGTGGATACAAGCAAAAATCAGTT




SS

TTCATTAAAGCTCTCTTCAGTGACCGCAGCTGATA






CCGCCGTCTATTATTGCGTGCGGGGGGTGGACTA






CTGGGGTCAGGGCACCATGGTTACTGTGTCATCA





Anti-
317
QVQLQESGPGLVKPSD
318
CAGGTACAGCTGCAGGAGAGTGGCCCTGGTTTA


MUC16

TLSLTCAVSGYSIVSHY

GTAAAGCCATCAGATACACTTTCACTTACCTGCGC


VH2

YWGWIRQPPGKGLE

CGTGTCTGGTTATTCTATCGTGAGCCACTATTACT




WIGYISSDGSNYYNPSL

GGGGATGGATCCGCCAGCCCCCTGGCAAAGGTCT




KSRVTMSVDTSKNQFS

TGAGTGGATTGGCTATATAAGTTCGGATGGCAGT




LKLSSVTAVDTAVYYCV

AACTATTACAATCCTTCTCTGAAGAGCCGTGTCAC




RGVDYWGQGTMVTV

TATGAGCGTGGACACTAGCAAAAACCAGTTCAGC




SS

CTGAAGCTGTCCTCCGTCACCGCCGTAGACACCG






CTGTCTACTATTGTGTTAGGGGGGTGGACTACTG






GGGCCAAGGCACCATGGTCACGGTGAGCAGC





Anti-
319
EVQLVESGGGLVQPG
320



MUC16

GSLRLSCAASGYSIVSH




VH3

YYMSWVRQAPGKGLE






WVSVISSDGSNYYADS






VKGRFTISRDNSKNTLY






LQMNSLRAEDTAVYYC






VRGVDYWGQGTLVTV






SS







Anti-
321
EVQLVESGGGLVQPG
322
GAGGTGCAGCTCGTCGAGTCCGGAGGCGGTCTG


MUC16

RSLRLSCAASGYSIVSH

GTGCAACCCGGCCGTTCTTTGCGGCTGAGTTGCG


VH4

YYMHWVRQAPGKGL

CTGCCAGTGGGTATAGCATCGTGAGTCACTATTA




EWVSAISSDGSNEYAD

CATGCATTGGGTTCGTCAAGCCCCTGGCAAGGGA




SVEGRFTISRDNAKNSL

CTAGAGTGGGTGTCCGCCATCTCCTCAGACGGTA




YLQMNSLRAEDTAVYY

GTAATGAGTACGCGGACAGCGTAGAGGGTAGAT




CVRGVDYWGQGTLVT

TCACCATTTCTCGGGACAATGCCAAAAATAGTCTA




VSS

TACCTCCAAATGAATTCCCTTAGGGCCGAAGACA






CTGCCGTGTACTACTGTGTTCGGGGCGTGGACTA






CTGGGGGCAGGGGACATTGGTGACTGTGAGCTC






A





Anti-
323
QVQLQESGPGLVKPS
324
CAGGTCCAACTGCAGGAATCTGGCCCCGGACTGG


MUC16

QTLSLTCTVSGYSIVSH

TTAAACCATCTCAGACACTCTCCCTGACCTGCACC


VH5

YYWSWIRQHPGKGLE

GTGTCTGGATACAGCATCGTTTCTCATTATTACTG




WIGYISSDGSNEYNPSL

GTCATGGATTAGGCAGCATCCCGGAAAAGGGCTT




KSLVTISVDTSKNQFSL

GAATGGATTGGCTACATCTCCTCCGACGGCTCCA




KLSSVTAADTAVYFCV

ATGAGTACAACCCATCACTTAAATCTCTGGTCACG




RGVDYWGQGTMVTV

ATAAGCGTAGACACATCTAAAAATCAGTTCTCATT




SS

AAAGCTCAGCTCTGTTACAGCTGCCGACACCGCT






GTGTACTTCTGTGTGCGAGGGGTTGACTACTGGG






GGCAGGGCACAATGGTGACAGTGTCTTCC





Anti-
325
QVQLVQSGAEVKKPG
326
CAGGTTCAACTGGTTCAGTCCGGAGCCGAGGTCA


MUC16

SSVKVSCKASGYSIVSH

AAAAGCCTGGATCCTCTGTGAAGGTGTCATGTAA


VH6

YYISWVRQAPGQGLE

GGCTTCTGGCTACAGCATCGTCTCACATTATTACA




WMGGISSDGSNNYA

TATCTTGGGTGCGACAGGCCCCCGGCCAGGGGCT




QKFQGRVTITADESTS

CGAGTGGATGGGAGGTATTTCCTCCGACGGGAG




TAYMELSSLRS

TAACAATTACGCTCAGAAATTTCAGGGCCGGGTG




EDTAVYYCVRGVDYW

ACCATTACCGCCGACGAAAGTACAAGCACCGCTT




GQGTLVTVSS

ATATGGAATTAAGCTCTTTAAGATCAGAGGACAC






GGCTGTGTACTACTGTGTAAGGGGCGTGGATTAC






TGGGGTCAGGGGACGCTCGTCACCGTCTCGAGC





Anti-
327
QVQLQESGPGLVKPSE
328
CAGGTCCAGCTCCAGGAATCCGGCCCAGGGTTGG


MUC16

TLSLTCTVSGYSIVSHYY

TGAAGCCTTCGGAGACCCTGTCTCTGACATGCAC


VH7

WSWIRQPPGKGLEWI

AGTCAGCGGCTATAGTATCGTCTCCCACTATTATT




GYISSDGSNNYNPSLKS

GGTCTTGGATTCGGCAACCTCCAGGCAAGGGGTT




RVTISVDTSKNQFSLKL

AGAATGGATTGGATACATCTCAAGCGATGGGTCC




SSVTAADTAVYYCVRG

AATAACTACAACCCAAGTCTCAAAAGTAGAGTGA




VDYWGQGTTVTVSS

CTATCTCTGTGGATACCAGTAAAAACCAGTTTTCA






CTCAAGTTGAGTTCCGTCACCGCCGCCGACACAG






CCGTTTACTACTGTGTTCGGGGAGTGGACTACTG






GGGCCAAGGTACCACGGTTACCGTGAGCAGC





Anti-
648
QVQLQESGPGLVKPSD
649
CAGGTGCAGCTGCAGGAGAGCGGCCCCGGCCTG


MUC16

TLSLTCAVSGYSIVSHY

GTGAAGCCCAGCGACACCCTGAGCCTGACCTGCG


VH8

YWHWIRQPPGKGLE

CCGTGAGCGGCTACAGCATCGTGAGCCACTACTA




WMGYISSDGSNDFNP

CTGGCACTGGATCAGACAGCCCCCCGGCAAGGG




SLKTRITISRDTSKNQFS

CCTGGAGTGGATGGGCTACATCAGCAGCGACGG




LKLSSVTAVDTAVYYCV

CAGCAACGACTTCAACCCCAGCCTGAAGACCAGA




RGVDYWGQGTLVTVSS

ATCACCATCAGCAGAGACACCAGCAAGAACCAGT






TCAGCCTGAAGCTGAGCAGCGTGACCGCCGTGG






ACACCGCCGTGTACTACTGCGTGAGAGGCGTGGA






CTACTGGGGCCAGGGCACCCTGGTGACCGTGAG






CAGC





Anti-
650
QVQLQESGPGLVKPS
651
CAGGTGCAGCTGCAGGAGAGCGGCCCCGGCCTG


MUC16

QTLSLTCAVYGYSIVSH

GTGAAGCCCAGCCAGACCCTGAGCCTGACCTGCG


VH9

YYWSWIRQPPGKGLE

CCGTGTACGGCTACAGCATCGTGAGCCACTACTA




WIGEISSDGSNNYNPS

CTGGAGCTGGATCAGACAGCCCCCCGGCAAGGG




LKSRVTISVDTSKNQFS

CCTGGAGTGGATCGGCGAGATCAGCAGCGACGG




LKLSSVTAADTAVYYCV

CAGCAACAACTACAACCCCAGCCTGAAGAGCAGA




RGVDYWGQGTLVTVSS

GTGACCATCAGCGTGGACACCAGCAAGAACCAGT






TCAGCCTGAAGCTGAGCAGCGTGACCGCCGCCGA






CACCGCCGTGTACTACTGCGTGAGAGGCGTGGAC






TACTGGGGCCAGGGCACCCTGGTGACCGTGAGC






AGC





Anti-
652
QVQLQESGPGLVKPSE
653
CAGGTGCAGCTGCAGGAGAGCGGCCCCGGCCTG


MUC16

TLSLTCAVSGYSIVSHY

GTGAAGCCCAGCGAGACCCTGAGCCTGACCTGCG


VH10

YWGWIRQPPGKGLE

CCGTGAGCGGCTACAGCATCGTGAGCCACTACTA




WIGSISSDGSNYYNPSL

CTGGGGCTGGATCAGACAGCCCCCCGGCAAGGG




KSRVTISVDTSKNQFSL

CCTGGAGTGGATCGGCAGCATCAGCAGCGACGG




KLSSVTAADTAVYYCV

CAGCAACTACTACAACCCCAGCCTGAAGAGCAGA




RGVDYWGQGTLVTVSS

GTGACCATCAGCGTGGACACCAGCAAGAACCAGT






TCAGCCTGAAGCTGAGCAGCGTGACCGCCGCCGA






CACCGCCGTGTACTACTGCGTGAGAGGCGTGGAC






TACTGGGGCCAGGGCACCCTGGTGACCGTGAGC






AGC





Anti-
654
QVQLVESGGGVVQPG
655
CAGGTGCAGCTGGTGGAGAGCGGCGGCGGCGT


MUC16

RSLRLSCAASGYSIVSH

GGTGCAGCCCGGCAGAAGCCTGAGACTGAGCTG


VH11

YYWNWVRQAPGKGL

CGCCGCCAGCGGCTACAGCATCGTGAGCCACTAC




EWVAYISSDGSNEYNP

TACTGGAACTGGGTGAGACAGGCCCCCGGCAAG




SLKNRFTISRDNSKNTL

GGCCTGGAGTGGGTGGCCTACATCAGCAGCGAC




YLQMNSLRAEDTAVYY

GGCAGCAACGAGTACAACCCCAGCCTGAAGAAC




CVRGVDYWGQGTTVT

AGATTCACCATCAGCAGAGACAACAGCAAGAACA




VSS

CCCTGTACCTGCAGATGAACAGCCTGAGAGCCGA






GGACACCGCCGTGTACTACTGCGTGAGAGGCGT






GGACTACTGGGGCCAGGGCACCACCGTGACCGT






GAGCAGC





Anti-
656
QVQLQESGPGLVKPS
657
CAGGTGCAGCTGCAGGAGAGCGGCCCCGGCCTG


MUC16

QTLSLTCTVSGYSIVSH

GTGAAGCCCAGCCAGACCCTGAGCCTGACCTGCA


VH12

YYWNWIRQHPGKGLE

CCGTGAGCGGCTACAGCATCGTGAGCCACTACTA




WIGYISSDGSNEYNPSL

CTGGAACTGGATCAGACAGCACCCCGGCAAGGG




KNLVTISVDTSKNQFSL

CCTGGAGTGGATCGGCTACATCAGCAGCGACGG




KLSSVTAADTAVYYCV

CAGCAACGAGTACAACCCCAGCCTGAAGAACCTG




RGVDYWGQGTMVTV

GTGACCATCAGCGTGGACACCAGCAAGAACCAGT




SS

TCAGCCTGAAGCTGAGCAGCGTGACCGCCGCCGA






CACCGCCGTGTACTACTGCGTGAGAGGCGTGGAC






TACTGGGGCCAGGGCACCATGGTGACCGTGAGC






AGC





Anti-
658
QVQLQESGPGLVKPSD
659
CAGGTGCAGCTGCAGGAGAGCGGCCCCGGCCTG


MUC16

TLSLTCAVSGYSIVSHY

GTGAAGCCCAGCGACACCCTGAGCCTGACCTGCG


VH13

YWNWIRQPPGKGLE

CCGTGAGCGGCTACAGCATCGTGAGCCACTACTA




WIGYISSDGSNEYNPSL

CTGGAACTGGATCAGACAGCCCCCCGGCAAGGG




KNRVTMSVDTSKNQF

CCTGGAGTGGATCGGCTACATCAGCAGCGACGG




SLKLSSVTAVDTAVYYC

CAGCAACGAGTACAACCCCAGCCTGAAGAACAG




VRGVDYWGQGTMVT

AGTGACCATGAGCGTGGACACCAGCAAGAACCA




VSS

GTTCAGCCTGAAGCTGAGCAGCGTGACCGCCGTG






GACACCGCCGTGTACTACTGCGTGAGAGGCGTG






GACTACTGGGGCCAGGGCACCATGGTGACCGTG






AGCAGC





Anti-
660
EVQLLESGGGLVQPGG 
661
GAGGTGCAGCTGCTGGAGAGCGGCGGCGGCCTG


MUC16

SLRLSCAASGYSIVSHY

GTGCAGCCCGGCGGCAGCCTGAGACTGAGCTGC


VH14

YWNWVRQAPGKGLE

GCCGCCAGCGGCTACAGCATCGTGAGCCACTACT




WVSYISSDGSNEYNPS

ACTGGAACTGGGTGAGACAGGCCCCCGGCAAGG




LKNRFTISRDNSKNTLY

GCCTGGAGTGGGTGAGCTACATCAGCAGCGACG




LQMNSLRAEDTAVYYC

GCAGCAACGAGTACAACCCCAGCCTGAAGAACA




VRGVDYWGQGTLVTV

GATTCACCATCAGCAGAGACAACAGCAAGAACAC




SS

CCTGTACCTGCAGATGAACAGCCTGAGAGCCGAG






GACACCGCCGTGTACTACTGCGTGAGAGGCGTG






GACTACTGGGGCCAGGGCACCCTGGTGACCGTG






AGCAGC





Anti-
672
VKLQESGGGFVKPGG
673
GTGAAGCTGCAAGAGTCCGGCGGAGGCTTTG


MUC16

SLKVSCAASGFTFSSY

TGAAGCCTGGCGGCTCTCTGAAAGTGTCCTGT


VH

AMSWVRLSPEMRLE

GCCGCCAGCGGCTTCACCTTTAGCAGCTACGCC




WVATISSAGGYIFYSD

ATGAGCTGGGTCCGACTGAGCCCTGAGATGAG




SVQGRFTISRDNAKN

ACTGGAATGGGTCGCCACCATCAGTAGCGCAGG




TLHLQMGSLRSGDTA

CGGCTACATCTTCTACAGCGACTCTGTGCAGGGC




MYYCARQGFGNYGDY

AGATTCACCATCAGCCGGGACAACGCCAAGAAC




YAMDYWGQGTTVTV

ACCCTGCACCTCCAGATGGGCAGTCTGAGAAGC




SS

GGCGATACCGCCATGTACTACTGCGCCAGACAA






GGCTTCGGCAACTACGGCGACTACTATGCCATG






GATTACTGGGGCCAGGGCACCACCGTGACAGT






CTCTTCT





Anti-
674
VKLEESGGGFVKPGG
675
GTGAAGCTGGAAGAGTCCGGCGGAGGCTTTG


MUC16

SLKISCAASGFTFRNY

TGAAGCCTGGCGGAAGCCTGAAGATCAGCTGTG


VH

AMSWVRLSPEMRL

CCGCCAGCGGCTTCACCTTCAGAAACTACGCC




EWVATISSAGGYIFY

ATGAGCTGGGTCCGACTGAGCCCCGAGATGAGA




SDSVQGRFTISRDNA

CTGGAATGGGTCGCCACAATCAGCAGCGCAGGC




KNTHLQMGSLRSGD

GGCTACATCTTCTACAGCGATAGCGTGCAGGGC




TAMYYCARQGFGNY

AGATTCACCATCAGCCGGGACAACGCCAAGAA




GDYYAMDYWGQGT

CACCCTGCACCTCCAGATGGGC




TVTVSS

AGTCTGAGATCTGGCGACACCGCCATGTACTACT






GGCCAGACAAGGCTTCGGCAACTACGGCGACTA






CTATGCCATGGATTACTGGGGCCAGGGCACCAC






CGTGACAGTCTCTTCT





Anti-
676
DVQLLESGPGLVRPS
677
GACGTGCAACTTCTGGAGAGCGGGCCAGGGCT


MUC16

QSLSLTCSVTGYSIVSH

AGTCAGGCCCTCCCAGTCGCTTTCACTGACTTG


VH

YYWNWIRQFPGNKLE

CAGTGTGACCGGTTACTCTATTGTGAGTCACTA




WMGYISSDGSNEYNP

CTATTGGAACTGGATTCGGCAGTTCCCAGGCA




SLKNRISISLDTSKNQF

ACAAACTGGAATGGATGGGGTACATATCTTCC




FLKFDFVTTADTATYF

GATGGCTCGAATGAATATAACCCATCATTGAAA




CVRGVDYWGQGTTLT

AATCGTATTTCCATCAGTCTGGATACGAGTAA




VSS

AAACCAGTTTTTCCTCAAATTCGATTTCGTGAC






TACAGCAGATACTGCCACATACTTCTGTGTAC






GAGGTGTCGATTATTGGGGACAGGGCACAACG






CTGACCGTAAGTTCT





Anti-
678
DVQLQESGPGLVNPS
679
GACGTTCAGCTGCAAGAGTCTGGCCCTGGCCT


MUC16

QSLSLTCTVTGYSITN

GGTCAATCCTAGCCAGAGCCTGAGCCTGACAT


VH

DYAWNWIRQFPGNK

GTACCGTGACCGGCTACAGCATCACCAACGAC




LEWMGYINYSGYTT

TACGCCTGGAACTGGATCAGACAGTTCCCCGG




YNPSLKSRISITRDTS

CAACAAGCTGGAATGGATGGGCTACATCAAC




KNQFFLHLNSVTTED

TACAGCGGCTACACCACCTACAATCCCAGCCTG




TATYYCARWDGGLTY

AAGTCCCGGATCTCCATCACCAGAGACACCAG




WGQGTLVTVSA

CAAGAACCAGTTCTTCCTGCACCTGAACAGC






GTGACCACCGAGGATACCGCCACCTACTACTG






CGCTAGATGGGATGGCGGCCTGACATATTGGG






GCCAGGGAACACTGGTCACCGTGTCTGCT





Anti-
680
EVQLVESGGGLVQPG
681
GAGGTGCAGCTGGTGGAGAGCGGCGGCGGC


MUC16

GSLRLSCAASGYSITND

CTGGTGCAGCCCGGCGGCAGCCTGAGGCTGAG


VH

YAWNWVRQAPGKG

CTGCGCCGCCAGCGGCTACAGCATCACCAACGA




LEWVGYINYSGYTTYN

CTACGCCTGGAACTGGGTGAGGCAGGCCCCCG




PSLKSRFTISRDNSKNT

GCAAGGGCCTGGAGTGGGTGGGCTACATCAA




LYLQMNSLRAEDTAV

CTACAGCGGCTACACCACCTACAACCCCAGCCTG




YYCARWDGGLTYWG

AAGAGCAGGTTCACCATCAGCAGGGACAACAGC




QGTLVTVSS

AAGAACACCCTGTACCTGCAGATGAACAGCCTG






AGGGCCGAGGACACCGCCGTGTACTACTGCGC






CAGGTGGGACGGCGGCCTGACCTACTGGGGCC






AGGGCACCCTGGTGACCGTGAGCAGC





Anti-
682
EVQLVESGGGLVQPG
683
GAGGTGCAGCTGGTGGAGAGCGGCGGCGGC


MUC16

GSLRLSCAASGYSITND

CTGGTGCAGCCCGGCGGCAGCCTGAGGCTGAG


VH

YAWNWVRQAPGKG

CTGCGCCGCCAGCGGCTACAGCATCACCAACGA




LEWVGYINYSGYTTYN

CTACGCCTGGAACTGGGTGAGGCAGGCCCCCG




PSLKSRFTISRDNSKNT

GCAAGGGCCTGGAGTGGGTGGGCTACATCAA




FYLQMNSLRAEDTAV

CTACAGCGGCTACACCACCTACAACCCCAGCCTG




YYCARWDGGLTYWG

AAGAGCAGGTTCACCATCAGCAGGGACAACAGC




QGTLVTVSS

AAGAACACCTTCTACCTGCAGATGAACAGCCTG






AGGGCCGAGGACACCGCCGTGTACTACTGCGC






CAGGTGGGACGGCGGCCTGACCTACTGGGGCC






AGGGCACCCTGGTGACCGTGAGCAGC





Anti-
684
EVQLQQSGAELVKPG
685
GAGGTTCAGCTGCAGCAGTCTGGCGCCGAACTT


MUC16

ASVKLSCTASGFNIKD

GTGAAACCTGGCGCCTCTGTGAAGCTGAGCTGT


VH

TYMHWVKQRPEQGL

ACCGCCAGCGGCTTCAACATCAAGGACACCTAC




EWIGRVDPANGNTK

ATGCACTGGGTCAAGCAGAGGCCTGAGCAGGG




YDPKFQGKATLTADT

CCTCGAATGGATCGGAAGAGTGGATCCCGCCA




SSNTAYLQLSSLTSEDT

ACGGCAACACCAAATACGACCCCAAGTTCCAGG




AVYFCVRDYYGHTYG

GCAAAGCCACACTGACCGCCGACACCTCTAGCA




FAFCDQGTTLTVSA

ACACAGCCTACCTGCAGCTGTCCAGCCTGACCTC






TGAAGATACCGCCGTGTACTTCTGCGTGCGGGA






CTACTACGGCCATACCTACGGCTTCGCCTTCTGC






GACCAAGGCACAACCCTGACAGTGTCTGCT





Anti-
686
EVQLVESGGGLVQPG
687
GAGGTGCAGCTGGTTGAATCTGGCGGAGGACT


MUC16

GSLRLSCAASGFNIKD

GGTTCAGCCTGGCGGATCTCTGAGACTGTCTTG


VH

TYMHWVRQAPGKGL

TGCCGCCAGCGGCTTCAACATCAAGGACACCTA




EWVGRVDPANGNTK

CATGCACTGGGTCCGACAGGCCCCTGGCAAAGG




YDPKFQGRFTISADTS

ACTTGAGTGGGTTGGAAGAGTGGACCCCGCCAA




KNTAYLQMNSLRAED

CGGCAACACCAAATACGACCCCAAGTTCCAGGG




TAVYYCVRDYYGHTY

CAGATTCACCATCAGCGCCGACACCAGCAAGAA




GFAFWGQGTLVTVSS

CACCGCCTACCTGCAGATGAACAGCCTGAGAG






CCGAGGACACCGCCGTGTACTATTGCGTGCG






GGATTACTACGGCCATACCTACGGCTTCGCCT






TTTGGGGCCAGGGCACACTGGTTACCGTTAGCT






CT





Anti-
329
DIQMTQSPSSLSASVG
330
GACATACAGATGACTCAGAGCCCCTCCTCACTCTC


MUC16

DRVTITCQASRDINNFL

GGCATCAGTCGGCGACAGGGTCACAATTACCTGT


VL1

NWYQQKPGKAPKLLIY

CAGGCTTCTCGCGACATTAATAACTTCCTGAATTG




RANNLETGVPSRFSGS

GTATCAGCAAAAGCCCGGGAAGGCCCCTAAGCT




GSGTDFTFTISSLQPED

GTTGATTTATAGAGCAAATAATCTCGAAACCGGC




IATYFCLQYGDLYTFGG

GTGCCCAGTAGGTTTAGCGGGTCCGGGAGCGGA




GTKVEIK

ACAGACTTCACATTCACCATTTCTAGTTTGCAGCC






CGAAGACATTGCTACATATTTTTGCCTGCAGTACG






GGGATCTCTACACTTTCGGGGGCGGAACAAAGGT






TGAGATAAAA





Anti-
331
DIQMTQSPSSLSASVG
332
GATATTCAAATGACGCAGTCACCCTCATCGCTCTC


MUC16

DRVTITCQASRDINNFL

TGCGTCAGTAGGGGATCGTGTCACGATAACCTGT


VL2

NWYQQKPGKAPKLLIY

CAAGCATCAAGGGACATCAACAACTTCCTCAACT




RANNLETGVPSRFSGS

GGTACCAACAGAAGCCTGGCAAGGCACCTAAACT




GSGTDFTFTISSLQPED

CCTGATCTACCGGGCTAACAACCTAGAAACCGGG




IATYYCLQYGDLYTFGG

GTTCCGAGCCGATTCAGTGGGTCTGGAAGCGGG




GTKVEIK

ACAGACTTTACGTTCACTATTAGTTCGCTACAGCC






CGAAGACATTGCGACATATTACTGTCTTCAGTATG






GGGATTTGTATACCTTTGGGGGAGGCACCAAGGT






AGAGATAAAG





Anti-
333
DIQMTQSPSSLSASVG
334
GACATCCAGATGACTCAGAGCCCGTCTTCTCTATC


MUC16

DRVTITCRASRDINNFL

CGCAAGTGTAGGCGATCGTGTCACCATCACATGC


VL3

GWYQQKPGKAPKRLI

CGGGCTTCCCGGGATATCAACAACTTCCTTGGGT




YRANSLQSGVPSRFSG

GGTATCAGCAGAAGCCCGGAAAAGCCCCCAAAC




SGSGTEFTLTISSLQPE

GGCTCATCTACAGAGCGAATTCCCTGCAGTCAGG




DFATYYCLQYGDLYTF

TGTCCCCAGTAGGTTCAGCGGATCAGGCTCGGGG




GQGTKVEIK

ACCGAATTCACTCTGACCATTAGCTCACTGCAGCC






TGAGGATTTCGCTACTTACTATTGCCTGCAATACG






GCGATCTGTACACTTTCGGGCAGGGCACCAAGGT






GGAAATAAAA





Anti-
335
EIVLTQSPGTLSLSPGE
336
GAAATCGTACTGACCCAGTCTCCCGGAACCCTGA


MUC16

RATLSCRASRDINNFLA

GTCTCTCACCCGGCGAGCGCGCAACACTGTCGTG


VL4

WYQQKPGQAPRLLIYR

TAGGGCCAGTAGGGACATAAATAACTTCCTAGCC




ANSRATGIPDRFSGSG

TGGTACCAACAAAAACCGGGTCAGGCTCCAAGAC




SGTDFTLTISRLEPEDF

TGTTGATCTATAGAGCTAACTCCAGGGCCACCGG




AVYYCLQYGDLYTFGQ

CATCCCAGACCGATTCTCAGGCTCCGGATCTGGA




GTKVEIK

ACCGACTTCACGCTCACCATTAGCCGACTAGAACC






TGAGGACTTTGCTGTATACTATTGCCTGCAGTACG






GCGACCTGTATACCTTTGGACAGGGTACCAAGGT






CGAGATCAAG





Anti-
337
EIVLTQSPATLSLSPGE
338
GAGATCGTACTTACGCAGAGCCCAGCAACTCTGT


MUC16

RATLSCRASRDINNFLA

CTCTGTCCCCCGGAGAACGGGCCACCCTGTCGTG


VL5

WYQQKPGQAPRLLIYR

CCGGGCCAGCCGTGATATTAATAATTTCCTGGCCT




ANNRATGIPARFSGSG

GGTATCAACAAAAACCGGGGCAGGCTCCTCGACT




PGTDFTLTISSLEPEDFA

GTTGATCTACCGGGCCAACAATAGAGCAACTGGT




VYYCLQYGDLYTFGGG

ATCCCTGCTCGCTTCTCCGGCAGTGGGCCAGGTA




TKVEIK

CAGACTTCACCCTGACTATTTCGTCACTCGAACCA






GAAGACTTTGCCGTGTATTATTGCTTACAATACGG






GGATCTGTACACTTTCGGAGGAGGAACTAAGGTC






GAAATTAAG





Anti-
339
EIVLTQSPDFQSVTPKE
340
GAGATCGTGCTGACCCAGAGCCCCGACTTCCAGA


MUC16

KVTITCRASRDINNFLH

GCGTGACCCCCAAGGAGAAGGTGACCATCACCTG


VL6

WYQQKPDQSPKLLIKR

CAGAGCCAGCAGAGACATCAACAACTTCCTGCAC




ANQSFSGVPSRFSGSG

TGGTACCAGCAGAAGCCCGACCAGAGCCCCAAG




SGTDFTLTINSLEAEDA

CTGCTGATCAAGAGAGCCAACCAGAGCTTCAGCG




ATYYCLQYGDLYTFGQ

GCGTGCCCAGCAGATTCAGCGGCAGCGGCAGCG




GTKVEIK

GCACCGACTTCACCCTGACCATCAACAGCCTGGA






GGCCGAGGACGCCGCCACCTACTACTGCCTGCAG






TACGGCGACCTGTACACCTTCGGCCAGGGCACCA






AGGTGGAGATCAAG





Anti-
341
DIQMTQSPSSLSASVG
342
GACATCCAGATGACCCAGAGCCCCAGCAGCCTGA


MUC16

DRVTITCRASRDINNFL

GCGCCAGCGTGGGCGACAGAGTGACCATCACCT


VL7

AWFQQKPGKAPKSLIY

GCAGAGCCAGCAGAGACATCAACAACTTCCTGGC




RANSLQSGVPSRFSGS

CTGGTTCCAGCAGAAGCCCGGCAAGGCCCCCAAG




GSGTDFTLTISSLQPED

AGCCTGATCTACAGAGCCAACAGCCTGCAGAGCG




FATYYCLQYGDLYTFG

GCGTGCCCAGCAGATTCAGCGGCAGCGGCAGCG




GGTKVEIK

GCACCGACTTCACCCTGACCATCAGCAGCCTGCA






GCCCGAGGACTTCGCCACCTACTACTGCCTGCAG






TACGGCGACCTGTACACCTTCGGCGGCGGCACCA






AGGTGGAGATCAAG





Anti-
662
DIQMTQSPSSLSASVG
663
GACATCCAGATGACCCAGAGCCCCAGCAGCCTGA


MUC16

DRVTITCRASRDINNFL

GCGCCAGCGTGGGCGACAGAGTGACCATCACCT


VL8

AWYQQKPGKAPKLLL

GCAGAGCCAGCAGAGACATCAACAACTTCCTGGC




YRANRLESGVPSRFSG

CTGGTACCAGCAGAAGCCCGGCAAGGCCCCCAA




SGSGTDYTLTISSLQPE

GCTGCTGCTGTACAGAGCCAACAGACTGGAGAG




DFATYYCLQYGDLYTF

CGGCGTGCCCAGCAGATTCAGCGGCAGCGGCAG




GGGTKVEIK

CGGCACCGACTACACCCTGACCATCAGCAGCCTG






CAGCCCGAGGACTTCGCCACCTACTACTGCCTGC






AGTACGGCGACCTGTACACCTTCGGCGGCGGCAC






CAAGGTGGAGATCAAG





Anti-
664
DIQMTQSPSSLSASVG
665
GACATCCAGATGACCCAGAGCCCCAGCAGCCTGA


MUC16

DRVTITCKASRDINNFL

GCGCCAGCGTGGGCGACAGAGTGACCATCACCT


VL9

SWYQQKPGKAPKLLIY

GCAAGGCCAGCAGAGACATCAACAACTTCCTGAG




RANRLVDGVPSRFSGS

CTGGTACCAGCAGAAGCCCGGCAAGGCCCCCAA




GSGTDFTFTISSLQPED

GCTGCTGATCTACAGAGCCAACAGACTGGTGGAC




IATYYCLQYGDLYTFGG

GGCGTGCCCAGCAGATTCAGCGGCAGCGGCAGC




GTKVEIK

GGCACCGACTTCACCTTCACCATCAGCAGCCTGCA






GCCCGAGGACATCGCCACCTACTACTGCCTGCAG






TACGGCGACCTGTACACCTTCGGCGGCGGCACCA






AGGTGGAGATCAAG





Anti-
666
DIQMTQSPSSLSASVG
667
GACATCCAGATGACCCAGAGCCCCAGCAGCCTGA


MUC16

DRVTITCKASRDINNFL

GCGCCAGCGTGGGCGACAGAGTGACCATCACCT


VL10

SWYQQKPGKAPKLLIY

GCAAGGCCAGCAGAGACATCAACAACTTCCTGAG





RANRLVDGVPSRFSGS


CTGGTACCAGCAGAAGCCCGGCAAGGCCCCCAA




GSGTDFTLTISSLQPED

GCTGCTGATCTACAGAGCCAACAGACTGGTGGAC




FATYYCLQYGDLYTFG

GGCGTGCCCAGCAGATTCAGCGGCAGCGGCAGC




QGTKVEIK

GGCACCGACTTCACCCTGACCATCAGCAGCCTGC






AGCCCGAGGACTTCGCCACCTACTACTGCCTGCA






GTACGGCGACCTGTACACCTTCGGCCAGGGCACC






AAGGTGGAGATCAAG





Anti-
668
EIVLTQSPGTLSLSPGE
669
GAGATCGTGCTGACCCAGAGCCCCGGCACCCTGA


MUC16

RATLSCKASRDINNFLS

GCCTGAGCCCCGGCGAGAGAGCCACCCTGAGCT


VL11

WYQQKPGQAPRLLIYR

GCAAGGCCAGCAGAGACATCAACAACTTCCTGAG




ANRLVDGIPDRFSGSG

CTGGTACCAGCAGAAGCCCGGCCAGGCCCCCAG




SGTDFTLTISRLEPEDF

ACTGCTGATCTACAGAGCCAACAGACTGGTGGAC




AVYYCLQYGDLYTFGQ

GGCATCCCCGACAGATTCAGCGGCAGCGGCAGC




GTKVEIK

GGCACCGACTTCACCCTGACCATCAGCAGACTGG






AGCCCGAGGACTTCGCCGTGTACTACTGCCTGCA






GTACGGCGACCTGTACACCTTCGGCCAGGGCACC






AAGGTGGAGATCAAG





Anti-
670
EIVLTQSPATLSLSPGE
671
GAGATCGTGCTGACCCAGAGCCCCGCCACCCTGA


MUC16

RATLSCKASRDINNFLS

GCCTGAGCCCCGGCGAGAGAGCCACCCTGAGCT


VL12

WYQQKPGQAPRLLIYR

GCAAGGCCAGCAGAGACATCAACAACTTCCTGAG




ANRLVDGIPARFSGSG

CTGGTACCAGCAGAAGCCCGGCCAGGCCCCCAG




SGTDFTLTISSLEPEDFA

ACTGCTGATCTACAGAGCCAACAGACTGGTGGAC




VYYCLQYGDLYTFGQG

GGCATCCCCGCCAGATTCAGCGGCAGCGGCAGC




TKVEIK

GGCACCGACTTCACCCTGACCATCAGCAGCCTGG






AGCCCGAGGACTTCGCCGTGTACTACTGCCTGCA






GTACGGCGACCTGTACACCTTCGGCCAGGGCACC






AAGGTGGAGATCAAG





Anti-
688
DIELTQSPSSLAVSAGE
689
GACATCGAGCTGACACAGAGCCCATCTAGCCTG


MUC16 VL

KVTMSCKSSQSLLNSR

GCTGTGTCTGCCGGCGAGAAAGTGACCATGAG




TRKNQLAWYQQKPG

CTGCAAGAGCAGCCAGAGCCTGCTGAACAGCC




QSPELLIYWASTRQSG

GGACCAGAAAGAATCAGCTGGCCTGGTATCAGC




VPDRFTGSGSGTDFTL

AGAAGCCCGGCCAATCTCCTGAGCTGCTGATCT




TISSVQAEDLAVYYCQ

ACTGGGCCAGCACAAGACAGAGCGGCGTGCC




QSYNLLTFGPGTKLEV

CGATAGATTCACAGGATCTGGCAGCGGCACCG




KR

ACTTCACCCTGACAATCAGTTCTGTGCAGGCCG






AGGACCTGGCCGTG






TACTACTGTCAGCAGAGCTACAACCTGCTGACCT






TCGGACCCGGCACCAAGCTGGAAGTGAAGAGA





Anti-
690
DIELTQSPSSLAVSAGE
691
GACATCGAGCTGACACAGAGCCCATCTAGCCTG


MUC16 VL

KVTMSCKSSQSLLNSR

GCTGTGTCTGCCGGCGAGAAAGTGACCATGA




TRKNQLAWYQQKTG

GCTGCAAGAGCAGCCAGAGCCTGCTGAACAGC




QSPELLIYWASTRQSG

CGGACCAGAAAGAATCAGCTGGCCTGGTATCA




VPDRFTGSGSGTDFTL

GCAGAAAACCGGACAGAGCCCCGAGCTGCTG




TISSVQAEDLAVYYCQ

ATCTACTGGGCCAGCACAAGACAGAGCGGCGTG




QSYNLLTFGPGTKLEIKR

CCCGATAGATTCACAGGATCTGGCAGCGGCACC






GACTTCACCCTGACAATCAGTTCTGTGCAGGCC






GAGGACCTGGCCGTGTACTACTGTCAGCAGAGC






TACAACCTGCTGACCTTCGGACCCGGCACCAAG






CTGGAAATCAAGAGA





Anti-
692
DIKMAQSPSSVNASL
693
GACATCAAGATGGCTCAGTCCCCTTCTAGCGT


MUC16 VL

GERVTITCKASRDINN

GAATGCTTCGCTAGGGGAGCGTGTGACCATCAC




FLSWFHQKPGKSTL

ATGTAAAGCATCACGCGACATAAATAATTTCCTT




IYRANRLVDGVPSRFS

TCCTGGTTTCATCAGAAACCGGGCAAGTCGCCTA




GSGSGQDYSFTISSLEY

AGACGCTGATTTACAGAGCAAATCGGTTGGTAG




EDVGIYYCLQYGDLYTF

ATGGAGTGCCAAGCAGATTCAGCGGGAGCGG




GGGTKLEIK

AAGTGGACAGGATTATAGCTTCACTATTTCATC






CCTGGAATACGAGGACGTAGGTATCTATTATTG






CCTCCAGTATGGCGATCTTTACACATTTGGTGGG






GGGACTAAGCTGGAGATTAAG





Anti-
694
DIQMTQSSSFLSVSLG
695
GACATCCAGATGACCCAGAGCAGCAGCTTCCTG


MUC16 VL

GRVTITCKASDLIHN

AGCGTGTCCCTTGGCGGCAGAGTGACCATCAC




WLAWYQQKPGNAPR

CTGTAAAGCCAGCGACCTGATCCACAACTGGCT




LLISGATSLETGVPSRF

GGCCTGGTATCAGCAGAAGCCTGGCAACGCTCC




SGSGSGNDYTLSIASLQ

CAGACTGCTGATTAGCGGCGCCACCTCTCTGGA




TEDAATYYCQQYWTT

AACAGGCGTGCCAAGCAGATTTTCCGGCAGCGG




PFTFGSGTKLEIK

CTCCGGCAACGACTACACACTGTCTATTGCCAG






CCTGCAGACCGAGGATGCCGCCACCTATTACTG






CCAGCAGTACTGGACCACACCTTTCACCTTTG






GCAGCGGCACCAAGCTGGAAATCAAG





Anti-
696
DIQMTQSPSSLSASV
697
GACATCCAGATGACCCAGAGCCCCAGCAGCCTGA


MUC16 VL

GDRVTITCKASDLIHN

GCGCCAGCGTGGGCGACAGGGTGACCATCACCT




WLAWYQQKPGKAPK

GCAAGGCCAGCGACCTGATCCACAACTGGCTGGC




LLISGATSLETGVPSRFS

CTGGTACCAGCAGAAGCCCGGCAAGGCCCCCAA




GSGSGTDFTLTISSLQP

GCTGCTGATCAGCGGCGCCACCAGCCTGGAGACC




EDFATYYCQQYWTTPF

GGCGTGCCCAGCAGGTTCAGCGGCAGCGGCAGC




TFGQGTKVEIKR

GGCACCGACTTCACCCTGACCATCAGCAGCCTG






CAGCCCGAGGACTTCGCCACCTACTACTGCCAG






CAGTACTGGACCACCCCCTTCACCTTCGGCCAG






GGCACCAAGGTGGAGATCAAGAGG





Anti-
698
DIVLTQSPAIMSASLG
699
GACATCGTGCTGACACAGAGCCCTGCCATCATG


MUC16 VL

ERVTMTCTASSSVSS

TCTGCCAGCCTCGGCGAGCGAGTGACCATGACA




SYLHWYQQKPGSSP

TGTACAGCCAGCAGCAGCGTGTCCAGCAGCTAC




KLWIYSTSNLASGVPG

CTGCATTGGTATCAGCAGAAGCCCGGCAGCAGC




RFSGSGSGTSYSLTIS

CCCAAGCTGTGGATCTACAGCACAAGCAATCTG




SMEAEDAATYYCH

GCCAGCGGCGTGCCAGGCAGATTTTCTGGTTC




QYHRSPYTFGGGTKV

TGGCAGCGGCACCAGCTACAGCCTGACAATCAG




EIKR

CAGCATGGAAGCCGAGGATGCCGCCACCTAC






TACTGCCACCAGTACCACAGAAGCCCCTACACC






TTTGGCGGAGGCACCAAGGTGGAAATCAAGC






GG





Anti-
700
DIQMTQSPSSLSASV
701
GACATCCAGATGACACAGAGCCCTAGCAGCCTG


MUC16 VL

GDRVTITCTASSSVSSS

TCTGCCAGCGTGGGAGACAGAGTGACCATCACC




YLHWYQQKPGKAPKL

TGTACAGCCAGCAGCAGCGTGTCCAGCAGCTAC




LIYSTSNLASGVPSRFS

CTGCATTGGTATCAGCAGAAGCCCGGCAAGGCC




GSGSGTDFTLTISSLQ

CCTAAGCTGCTGATCTACAGCACCAGCAATCTGG




PEDFATYYCHQYHRSP

CCAGCGGCGTGCCAAGCAGATTTTCTGGCTCT




YTFGQGTKVEIKR

GGCAGCGGCACCGACTTCACCCTGACCATATCT






AGCCTGCAGCCTGAGGACTTCGCCACCTACTAC






TGCCACCAGTACCACAGAAGCCCCTACACCTTT






GGCCAGGGCACCAAGGTGGAAATCAAGCGG





Anti-
343
DIKMAQSPSSVNA
344
GACATCAAGATGGCTCAGTCCCCTTCTAGCGTGA


MUC16

SLGERVTITCKASR

ATGCTTCGCTAGGGGAGCGTGTGACCATCACATG


scFv

DINNFLSWFHQKP

TAAAGCATCACGCGACATAAATAATTTCCTTTCCT




GKSPKTLIYRANRL

GGTTTCATCAGAAACCGGGCAAGTCGCCTAAGAC




VDGVPSRFSGSGS

GCTGATTTACAGAGCAAATCGGTTGGTAGATGGA




GQDYSFTISSLEYE

GTGCCAAGCAGATTCAGCGGGAGCGGAAGTGGA




DVGIYYCLQYGDL

CAGGATTATAGCTTCACTATTTCATCCCTGGAATA




YTFGGGTKLEIKG

CGAGGACGTAGGTATCTATTATTGCCTCCAGTAT




GGGSGGGGSGGG

GGCGATCTTTACACATTTGGTGGGGGGACTAAGC




GSDVQLLESGPGL

TGGAGATTAAG




VRPSQSLSLTCSVT

GGCGGAGGCGGAAGCGGAGGCGGAGGCTCCGG




GYSIVSHYYWNWI

CGGAGGCGGAAGCGACGTGCAACTTCTGGAGAG




RQFPGNKLEWMG

CGGGCCAGGGCTAGTCAGGCCCTCCCAGTCGCTT




YISSDGSNEYNPSL

TCACTGACTTGCAGTGTGACCGGTTACTCTATTGT




KNRISISLDTSKNQ

GAGTCACTACTATTGGAACTGGATTCGGCAGTTC




FFLKFDFVTTADT

CCAGGCAACAAACTGGAATGGATGGGGTACATA




ATYFCVRGVDYW

TCTTCCGATGGCTCGAATGAATATAACCCATCATT




GQGTTLTVSS

GAAAAATCGTATTTCCATCAGTCTGGATACGAGT






AAAAACCAGTTTTTCCTCAAATTCGATTTCGTGAC






TACAGCAGATACTGCCACATACTTCTGTGTACGA






GGTGTCGATTATTGGGGACAGGGCACAACGCTG






ACCGTAAGTTCT









Exemplary anti-ROR1 Variable Heavy (VH) and Variable Light (VL) Sequences


















Amino acid sequence

Nucleotide sequence







hROR1 VH_04
345
EVQLVESGGGLVQPGGSLRLSCAA
346
GAGGTGCAGCTCGTGGAAT




SGFTFSSYAMSWVRQAPGKGLEW

CCGGCGGTGGCCTGGTGCA




VSAISRGGTTYYADSVKGRFTISRD

GCCGGGCGGCAGTCTTCGA




NSKNTLYLQMNSLRAEDTAVYYCG

CTCTCCTGTGCGGCGTCAG




RYDYDGYYAMDYWGQGTLVTVSS

GCTTTACGTTCAGTTCTTAT






GCCATGAGCTGGGTGAGGC






AAGCTCCCGGTAAGGGACT






GGAGTGGGTCTCTGCTATC






AGCCGGGGAGGTACGACCT






ACTACGCTGACTCCGTAAAA






GGAAGATTTACCATAAGTC






GTGACAATTCCAAAAACACT






CTATACTTACAGATGAACTC






GCTCAGGGCCGAAGATACC






GCAGTCTACTATTGTGGGA






GATACGATTACGACGGCTA






CTATGCTATGGATTATTGGG






GTCAGGGTACGCTCGTGAC






GGTGTCCTCC





hROR1 VL_04
347
DIQMTQSPSSLSASVGDRVTITCQA
348
GATATTCAAATGACGCAAA




SPDINSYLNWYQQKPGKAPKLLIYR

GTCCCAGCAGCCTCTCCGCC




ANNLETGVPSRFSGSGSGTDFTLTI

TCCGTTGGAGACAGGGTGA




SSLQPEDIATYYCLQYDEFPYTFGQ

CTATTACATGCCAAGCCAGC




GTKLEIK

CCCGATATTAATAGCTACTT






AAATTGGTATCAGCAGAAA






CCTGGGAAGGCACCTAAAC






TTCTCATCTACCGCGCTAAC






AATCTGGAGACCGGCGTGC






CGTCTAGATTTTCCGGCTCT






GGATCAGGGACCGATTTTA






CTCTGACAATTAGTTCCCTG






CAACCCGAAGACATCGCCA






CTTATTATTGCCTGCAATAT






GATGAGTTTCCTTACACATT






TGGTCAGGGAACTAAACTA






GAGATTAAG





hROR1 VH_05
349
EVQLVESGGGLVQPGGSLRLSCAA
350
GAAGTGCAACTGGTCGAGT




SGFTFSSYAMSWVRQAPGKGLEW

CTGGGGGCGGCCTTGTGCA




VSSISRGGTTYYPDSVKGRFTISRDN

ACCTGGAGGCAGCCTTCGA




SKNTLYLQMNSLRAEDTAVYYCGR

CTCAGTTGCGCCGCGTCTG




YDYDGYYAMDYWGQGTLVTVSS

GTTTTACCTTCTCCTCTTACG






CGATGAGCTGGGTTCGCCA






GGCCCCCGGCAAGGGACTT






GAGTGGGTTAGTTCGATCT






CCCGCGGAGGCACCACATA






TTATCCTGACTCGGTTAAGG






GACGCTTCACTATCTCTAGG






GACAATTCAAAGAACACAC






TGTATCTCCAAATGAACTCC






TTGCGGGCCGAGGACACTG






CTGTGTATTATTGCGGACG






ATACGACTACGATGGGTAT






TACGCCATGGATTACTGGG






GGCAAGGTACACTGGTCAC






TGTGAGTTCG





hROR1 VL_05
351
DIQMTQSPSSLSASVGDRVTITCKA
352
GATATTCAGATGACCCAGTC




SPDINSYLSWYQQKPGKAPKLLIYR

ACCTTCGAGTCTGAGCGCA




ANRLVDGVPSRFSGSGSGTDFTLTI

TCCGTGGGCGACAGAGTGA




SSLQPEDIATYYCLQYDEFPYTFGQ

CCATTACCTGTAAGGCCAGC




GTKLEIK

CCGGACATTAACAGCTACCT






ATCGTGGTATCAGCAAAAG






CCTGGTAAGGCCCCTAAACT






CCTTATCTACAGGGCTAATA






GGTTGGTAGACGGGGTGCC






TAGCCGGTTCTCTGGTTCCG






GCAGCGGTACGGACTTTAC






TCTGACCATAAGCTCTCTGC






AACCAGAAGACATCGCAAC






ATACTACTGTTTACAATACG






ACGAATTTCCTTATACCTTT






GGCCAGGGGACCAAGTTAG






AGATCAAG





hROR1 VH_06
353
EVQLVESGGGLVQPGGSLRLSCAA
354
GAGGTTCAGCTGGTCGAGT




SGFTFSSYAIIWVRQAPGKGLEWV

CCGGGGGAGGCTTAGTGCA




ARISRGGTTRYADSVKGRFTISADT

GCCAGGAGGCAGTCTGCGG




SKETAYLQMNSLRAEDTAVYYCGR

CTCTCTTGCGCTGCAAGTGG




YDYDGYYAMDYWGQGTLVTVSS

CTTCACATTCAGTTCATACG






CAATCATCTGGGTTCGACA






GGCTCCTGGTAAGGGCCTC






GAATGGGTCGCAAGGATAT






CACGAGGTGGAACCACTAG






ATACGCAGACTCTGTTAAG






GGCAGGTTCACAATTAGCG






CGGATACCTCCAAGGAGAC






TGCTTATTTACAGATGAACT






CTCTGAGAGCCGAGGACAC






TGCTGTTTACTACTGCGGCC






GATACGATTACGACGGATA






TTACGCAATGGATTACTGG






GGCCAGGGCACGCTGGTGA






CAGTTTCATCG





hROR1 VL_06
355
DIQMTQSPSSLSASVGDRVTITCKA
356
GATATCCAGATGACTCAGA




SPDINSYLSWYQQKPGKAPKLLIYR

GTCCCAGTAGCCTGTCGGC




ANRLVDGVPSRFSGSGSGTDFTLTI

AAGCGTCGGAGATCGGGTC




SSLQPEDIATYYCLQYDEFPYTFGQ

ACAATTACCTGCAAAGCTA




GTKLEIK

GTCCTGATATTAATTCTTAC






TTGTCCTGGTATCAGCAGA






AGCCTGGTAAGGCCCCTAA






GTTGCTCATCTATCGGGCTA






ACCGGCTGGTGGACGGTGT






TCCCTCTAGATTCTCAGGGA






GTGGAAGCGGCACTGACTT






CACCCTGACTATATCGAGCC






TTCAGCCAGAGGACATTGC






CACATACTACTGTCTGCAAT






ATGATGAATTTCCATATACA






TTCGGACAAGGTACAAAGT






TAGAAATTAAG





hROR1 VH_07
357
EVQLVESGGGLVQPGGSLRLSCAA
358
GAAGTCCAACTGGTGGAGT




SGFTFSSYAIIWVRQAPGKGLEWV

CTGGCGGGGGCTTGGTGCA




ARISRGGTTRYADSVKGRFTISADT

GCCCGGTGGCTCCCTTAGG




SKETAYLQMNSLRAEDTAVYYCGR

CTGTCTTGCGCTGCCAGCG




YDYDGYYAMDYWGQGTLVTVSS

GGTTCACATTCAGCTCCTAT






GCGATTATATGGGTCCGAC






AGGCACCCGGCAAGGGATT






GGAGTGGGTGGCTCGCATC






AGCAGAGGCGGCACTACTC






GTTACGCCGACTCCGTGAA






AGGCAGATTCACCATCAGT






GCAGACACATCCAAGGAAA






CCGCATATCTTCAGATGAAT






AGCCTGCGAGCGGAGGATA






CCGCCGTCTATTATTGCGGA






CGCTATGATTACGACGGTT






ATTATGCTATGGACTACTGG






GGCCAGGGCACACTTGTGA






CCGTCAGTAGC





hROR1 VL_07
359
DIQMTQSPSSLSASVGDRVTITCRA
360
GACATTCAAATGACGCAAA




SPDINSYVAWYQQKPGKAPKLLIYR

GCCCTAGTAGCTTGTCAGCT




ANFLESGVPSRFSGSRSGTDFTLTIS

TCTGTGGGGGACCGTGTCA




SLQPEDFATYYCLQYDEFPYTFGQG

CAATCACTTGTCGGGCCTCT




TKVEIK

CCAGATATAAACTCCTACGT






TGCTTGGTATCAGCAGAAG






CCCGGAAAGGCTCCGAAAT






TGTTGATTTATCGCGCTAAT






TTCTTAGAGTCAGGAGTGC






CCAGCCGGTTCTCAGGGTC






TCGCTCTGGAACCGACTTCA






CACTCACTATTTCTAGCCTA






CAGCCTGAGGATTTTGCAA






CTTACTACTGTCTACAGTAC






GACGAGTTTCCGTACACTTT






CGGACAGGGGACCAAGGT






GGAGATCAAG





hROR1 VH_08
361
QVQLVQSGGGLVKPGGSLRLSCAA
362
CAAGTACAGCTCGTGCAGA




SGFTFSSYAMSWVRQIPGKGLEW

GCGGCGGTGGCCTGGTGAA




VSSISRGGTTYYPDSVKGRFTISRDN

GCCAGGAGGTAGTCTTAGA




VKNTLYLQMSSLRAEDTAVYYCGR

CTGAGCTGTGCGGCTTCTG




YDYDGYYAMDYWGQGTMVTVSS

GTTTCACGTTCAGCAGTTAT






GCTATGTCCTGGGTTAGGC






AAATCCCCGGCAAAGGATT






GGAGTGGGTTAGCAGTATC






TCAAGGGGGGGAACCACAT






ATTATCCTGACTCTGTCAAA






GGACGGTTTACAATCAGCC






GCGATAACGTTAAAAATAC






CCTCTACCTCCAGATGTCTT






CGCTCCGCGCTGAAGATAC






AGCGGTTTACTACTGTGGC






AGATACGACTACGACGGTT






ATTACGCCATGGACTACTG






GGGACAGGGAACTATGGTC






ACAGTTAGCTCT





hROR1 VL_08
363
DIKMTQSPSSLSASVGDRVTITCKA
364
GACATCAAAATGACGCAGT




SPDINSYLSWYQQKPGKAPKTLIYR

CACCTAGTAGCCTCTCCGCC




ANRLVDGVPSRFSGSGSGTDFTLTI

TCGGTTGGCGATCGGGTAA




SSLQYEDMAIYYCLQYDEFPYTFGD

CCATTACCTGCAAAGCATCT




GTKVEIK

CCAGACATAAATAGTTATCT






TAGTTGGTATCAACAGAAA






CCTGGCAAAGCTCCTAAGA






CCCTCATCTACCGCGCTAAC






CGCCTCGTGGATGGTGTTC






CAAGTCGGTTCTCAGGAAG






CGGCAGTGGCACAGACTTT






ACACTGACAATTAGTTCCCT






CCAGTATGAGGATATGGCC






ATATATTACTGCCTTCAGTA






TGATGAGTTTCCATACACAT






TCGGAGACGGTACAAAGGT






GGAGATCAAG





hROR1 VH_09
365
QVSLRESGGGLVQPGRSLRLSCTAS
366
CAAGTGAGCCTCCGGGAGA




GFTFSSYAMTWVRQAPGKGLEWV

GTGGGGGCGGTCTGGTCCA




ASISRGGTTHFADSVKGRFTISRDN

ACCAGGACGGTCACTGCGG




SNNTLYLQMDNVRDEDTAIYYCGR

CTGTCATGCACTGCCAGCG




YDYDGYYAMDYWGRGTLVTVSS

GCTTCACATTTAGCTCTTAC






GCCATGACTTGGGTCCGCC






AAGCTCCCGGTAAGGGACT






GGAGTGGGTGGCCAGCATT






AGCAGGGGTGGTACAACCC






ACTTCGCGGATTCAGTTAA






GGGGAGATTCACTATCTCC






AGGGATAATTCCAACAACA






CGCTGTACCTTCAGATGGAT






AACGTGAGAGACGAGGATA






CCGCGATATACTACTGTGG






CCGCTATGACTACGATGGTT






ATTATGCTATGGATTACTGG






GGGCGGGGCACCCTGGTG






ACTGTGTCCTCG





hROR1 VL_09
367
DIVMTQSPSSLSASVGDRVTITCRA
368
GATATCGTGATGACACAGT




SPDINSYLAWYQQKPGKAPKLLIYR

CACCTAGCTCCCTGAGCGCA




ANSLQSGVPSRFSGSGSGTEFTLTIS

AGCGTGGGGGATAGGGTT




SLQPEDFATYYCLQYDEFPYTFGQG

ACCATAACTTGCAGGGCCA




TKLEMK

GTCCCGACATCAATAGTTAT






TTGGCCTGGTATCAACAGA






AGCCTGGGAAGGCACCTAA






GTTGCTTATTTATAGGGCTA






ACTCGTTACAGAGCGGTGT






GCCAAGTCGGTTCTCAGGC






TCAGGGTCCGGGACCGAGT






TCACCCTGACCATCAGTAGC






TTGCAGCCAGAAGATTTTG






CCACCTACTACTGTCTTCAA






TACGATGAGTTTCCTTACAC






TTTTGGACAGGGCACCAAA






CTAGAGATGAAG





hROR1 VH_10
369
QVQLVESGGGVVQPGRSLRLSCAA
370
CAGGTTCAACTGGTAGAAT




SGFTFSSYAMNWVRQAPAKGLEW

CCGGCGGAGGTGTAGTGCA




VAIISRGGTQYYADSVKGRFTISRD

GCCTGGAAGGTCATTACGG




NSKNTLYLQMNGLRAEDTAVYYCG

TTAAGTTGCGCCGCCTCCG




RYDYDGYYAMDYWGQGTLVTVSS

GGTTCACATTTAGCAGCTAT






GCTATGAACTGGGTGCGCC






AGGCCCCTGCGAAAGGACT






CGAATGGGTTGCCATCATC






AGCCGAGGAGGCACACAGT






ATTATGCCGATTCTGTGAAG






GGTCGTTTTACTATTTCCAG






AGACAACAGTAAAAATACG






CTGTACCTGCAAATGAACG






GATTGAGGGCTGAGGATAC






CGCCGTGTACTACTGTGGA






CGCTACGACTATGATGGGT






ACTACGCGATGGACTATTG






GGGGCAAGGAACCCTTGTA






ACCGTTAGTTCA





hROR1 VL_10
371
EIVLTQSPDFQSVTPKEKVTITCRAS
372
GAGATCGTTTTGACACAGA




PDINSYLSWYQQKPDQSPKLLIKRA

GCCCCGATTTCCAGAGCGT




NQSFSGVPSRFSGSGSGTDFTLTIN

CACGCCCAAGGAGAAGGTC




SLEAEDAAAYYCLQYDEFPYTFGPG

ACCATCACCTGCCGAGCCA




TKVDIK

GCCCCGACATCAACAGTTAT






CTTTCATGGTATCAACAGAA






ACCTGATCAGAGCCCTAAG






CTGCTGATTAAGCGCGCCA






ACCAGAGCTTCTCAGGGGT






TCCTTCACGGTTTTCCGGGT






CAGGCAGCGGGACTGACTT






CACGTTGACCATTAACTCTT






TGGAGGCTGAGGATGCTGC






TGCCTATTACTGCCTTCAGT






ACGACGAGTTCCCCTATACA






TTTGGTCCTGGAACAAAAG






TGGATATAAAG





hROR1 VH_11
373
QVQLVQSGAEVKKPGASVKVSCKA
374
CAGGTGCAGCTCGTCCAGA




SGFTFSSYAMHWVRQAPGQGLE

GCGGAGCCGAAGTGAAGA




WMGNISRGGTTNYAEKFKNRVTM

AGCCGGGAGCATCAGTGAA




TRDTSISTAYMELSRLRSDDTAVYY

AGTTTCCTGCAAAGCAAGT




CGRYDYDGYYAMDYWGQGTLVT

GGCTTCACTTTCAGCAGTTA




VSS

CGCGATGCACTGGGTGCGG






CAGGCACCAGGTCAGGGAC






TGGAATGGATGGGGAACAT






CTCTCGCGGCGGAACAACC






AATTACGCAGAGAAGTTTA






AGAATCGCGTTACGATGAC






CAGAGACACTTCTATTAGTA






CAGCCTATATGGAGTTGTC






GCGTCTGAGAAGCGACGAT






ACCGCTGTCTACTATTGCGG






CCGGTACGATTATGACGGC






TACTATGCAATGGATTACTG






GGGACAGGGCACACTTGTG






ACAGTGTCTAGT





hROR1 VL_11
375
DIVMTQSPLSLPVTPGEPASISCRSS
376
GACATTGTGATGACTCAGT




PDINSYLEWYLQKPGQSPQLLIYRA

CTCCACTCAGCCTGCCTGTC




NDRFSGVPDRFSGSGSGTDFTLKIS

ACGCCCGGCGAACCCGCTT




RVEAEDVGVYYCLQYDEFPYTFGQ

CTATCTCTTGTAGGAGTAGC




GTKVEIK

CCTGATATCAACAGCTACCT






CGAATGGTATCTCCAGAAA






CCTGGTCAGAGCCCCCAGC






TCTTGATCTATAGAGCAAAC






GACAGGTTCTCTGGCGTGC






CTGATAGGTTTTCCGGTAGT






GGCAGCGGAACCGACTTCA






CACTTAAGATTTCAAGGGTC






GAGGCCGAGGACGTGGGG






GTGTATTACTGCTTACAGTA






CGATGAGTTTCCGTATACAT






TCGGGCAAGGCACAAAGGT






GGAAATTAAG





hROR1 VH_12
377
EVQLVESGGGLVQPGGSLRLSCTG
378
GAAGTGCAACTGGTCGAAA




SGFTFSSYAMHWLRQVPGEGLEW

GTGGAGGGGGACTAGTGC




VSGISRGGTIDYADSVKGRFTISRD

AGCCCGGAGGGTCACTGAG




DAKKTLSLQMNSLRAEDTAVYYCG

GCTATCATGCACCGGCTCTG




RYDYDGYYAMDYWGQGTMVTVSS

GTTTTACTTTTTCCAGCTAT






GCCATGCACTGGCTCAGAC






AGGTTCCGGGGGAAGGACT






GGAGTGGGTTAGCGGAATC






TCCAGAGGCGGAACTATTG






ACTACGCAGACAGCGTGAA






AGGTAGGTTTACCATCAGC






AGGGACGATGCTAAAAAGA






CCCTGTCACTTCAAATGAAT






AGCCTGAGAGCTGAGGATA






CGGCCGTGTATTACTGTGG






ACGCTATGACTACGATGGA






TATTACGCAATGGACTACTG






GGGCCAGGGAACAATGGT






GACCGTCTCAAGC





hROR1 VL_12
379
EIVLTQSPATLSVSPGERATLSCRAS
380
GAGATCGTCCTGACCCAGA




PDINSYLAWYQQKPGQAPRLLFSR

GCCCAGCTACTTTGTCAGTT




ANNRATGIPARFTGSGSGTDFTLTI

TCGCCAGGCGAGCGGGCCA




SSLEPEDFAIYYCLQYDEFPYTFGQG

CACTGAGCTGTAGGGCTTC




TKVEIK

TCCTGATATCAATTCTTACC






TGGCCTGGTATCAACAGAA






ACCGGGACAGGCCCCTCGC






CTGCTGTTCTCCCGCGCCAA






CAATAGGGCGACTGGCATA






CCAGCTCGGTTTACTGGGA






GTGGGTCAGGCACTGATTT






CACGCTTACAATCAGTAGCC






TGGAGCCCGAAGACTTCGC






CATCTACTACTGTTTACAAT






ACGATGAGTTCCCCTATACC






TTCGGCCAAGGGACCAAGG






TGGAGATCAAG





hROR1 VH_13
381
EVQLVESGGGVVQPGRSLRLSCAA
382
GAAGTGCAGCTAGTAGAAA




SGFTFSSYAMSWVRQAPGKGLEW

GTGGTGGTGGGGTCGTGCA




VASISRGGTQYYADSVKGRFTISRD

GCCAGGCCGCTCGCTCAGG




NSKNTLYLQMNGLRAEDTAVYYCG

CTGTCTTGCGCTGCGAGTG




RYDYDGYYAMDYWGQGTLVTVSS

GTTTCACATTCTCTTCATAC






GCCATGAGCTGGGTGAGAC






AGGCTCCCGGCAAGGGCCT






CGAATGGGTCGCATCTATA






AGCAGAGGCGGAACCCAGT






ACTACGCTGACAGTGTGAA






GGGTCGCTTTACAATCTCAC






GGGACAACAGTAAAAACAC






CCTCTATCTACAGATGAATG






GCTTGCGAGCTGAAGACAC






GGCTGTGTATTATTGCGGG






CGCTATGACTATGATGGTTA






CTACGCTATGGATTACTGG






GGCCAGGGCACCCTGGTTA






CTGTTTCATCA





hROR1 VL_13
383
EIVLTQSPDFQSVTPKEKVTITCRAS
384
GAAATAGTCCTGACCCAGA




PDINSYLPWYQQKPDQSPKLLIKRA

GCCCAGACTTCCAGTCCGT




NQSFSGVPSRFSGSGSGTDFTLTIN

GACCCCTAAGGAGAAGGTT




SLEAEDAAAYYCLQYDEFPYTFGPG

ACTATCACTTGCAGGGCAA




TKVDIK

GCCCTGACATAAATTCATAC






CTGCCATGGTATCAGCAGA






AGCCAGACCAGTCGCCGAA






GCTATTAATCAAACGCGCCA






ACCAGTCTTTTAGCGGCGTA






CCATCCCGATTCTCAGGTTC






GGGGTCCGGGACCGATTTC






ACACTCACGATAAACTCCCT






TGAGGCAGAGGATGCAGC






GGCTTACTACTGTTTACAGT






ACGACGAGTTTCCATATACG






TTCGGCCCCGGCACGAAGG






TAGATATCAAG





hROR1 VH_14
385
EVQLVESGGGLVQPGGSLRLSCAT
386
GAAGTGCAGCTGGTGGAGT




SGFTFSSYAMSWMRQAPGKGLE

CTGGCGGCGGTCTGGTGCA




WVASISRGGTTYYADSVKGRFTISV

GCCCGGCGGCTCTCTGCGC




DKSKNTLYLQMNSLRAEDTAVYYC

CTCTCCTGTGCCACCTCTGG




GRYDYDGYYAMDYWGQGTLVTVSS

TTTTACATTCTCCTCCTACGC






TATGTCCTGGATGCGGCAA






GCCCCCGGCAAGGGCCTAG






AGTGGGTCGCCTCAATCAG






CAGGGGCGGGACGACTTAT






TATGCCGATTCAGTTAAGG






GGAGATTCACAATTTCCGT






GGATAAATCCAAGAATACC






TTATACCTCCAGATGAACTC






TCTGCGGGCCGAAGATACG






GCCGTATATTATTGTGGGA






GGTATGACTACGACGGATA






TTACGCCATGGATTATTGG






GGGCAGGGGACACTTGTTA






CAGTGAGTTCC





hROR1 VL_14
387
DIQMTQSPSSLSASVGDRVTITCKA
388
GATATACAGATGACACAGA




SPDINSYLNWYQQKPGKAPKLLIYR

GCCCTTCAAGTTTATCTGCA




ANRLVDGVPSRFSGSGSGTDYTLTI

AGCGTCGGCGATCGTGTTA




SSLQPEDFATYYCLQYDEFPYTFGA

CAATAACTTGCAAGGCATCT




GTKVEIK

CCCGACATCAATTCCTACCT






CAACTGGTATCAGCAGAAG






CCTGGGAAGGCTCCTAAGC






TGCTTATTTACAGAGCAAAT






CGCCTGGTGGACGGCGTGC






CCAGTCGGTTTTCCGGGTCT






GGGAGCGGAACGGATTACA






CACTGACCATCTCAAGCCTG






CAACCCGAAGACTTCGCTAC






ATATTACTGCCTTCAGTATG






ATGAGTTCCCATATACCTTC






GGCGCTGGGACCAAGGTG






GAGATAAAG





hROR1
389
EVQLVESGGGLVQPGGSLRLSCASS
390
GAGGTCCAGCTCGTCGAAT


VH_14-1

GFTFSSYAMSWRRQAPGKGLEWV

CTGGCGGAGGTTTAGTGCA




AGISRGGTTSYADSVKGRFTISSDD

ACCAGGCGGGTCGCTCCGA




SKNTLYLQMNSLRAEDTAVYYCGR

TTAAGTTGTGCGTCCAGTG




YDYDGYYAMDYWGQGTLVTVSS

GCTTCACCTTCTCCAGCTAC






GCCATGTCGTGGAGGCGAC






AGGCTCCTGGCAAAGGCTT






GGAGTGGGTTGCTGGTATC






TCCCGAGGAGGCACCACTA






GTTACGCTGACAGTGTAAA






AGGACGTTTCACTATTTCCT






CTGACGACAGCAAGAACAC






ACTCTATCTGCAAATGAATA






GTCTCCGTGCTGAGGACAC






AGCCGTGTATTATTGCGGG






CGGTATGATTACGACGGCT






ACTACGCTATGGACTACTG






GGGCCAGGGAACTCTGGTC






ACTGTGAGCTCT





hROR1
391
DIQMTQSPSSLSASVGDRVTITCRA
392
GATATACAGATGACTCAAA


VL 14-1

SPDINSYLSWYQQKPGKAPKLLIYR

GTCCTAGCTCCTTGAGCGCC




ANTLESGVPSRFSGSGSGTDFTLTIS

TCAGTGGGAGATCGGGTCA




SLQPEDFATYYCLQYDEFPYTFGQG

CTATAACTTGTAGAGCCTCA




TKIEIK

CCAGATATAAACTCCTATCT






CTCTTGGTATCAGCAGAAG






CCCGGCAAAGCACCAAAGC






TCTTGATCTATAGAGCTAAT






ACGCTAGAGAGCGGAGTGC






CTTCACGGTTTTCTGGTTCC






GGGAGCGGAACCGACTTTA






CCCTTACAATTTCTAGCCTC






CAGCCAGAGGACTTCGCAA






CTTACTATTGTCTCCAGTAT






GATGAATTTCCTTACACCTT






CGGCCAAGGGACCAAGATC






GAGATAAAG





hROR1
393
EVQLVESGGGLVQPGGSLRLSCASS
394
GAGGTGCAGCTCGTTGAGT


VH_14-2

GFTFSSYAMSWVRQAPGKGLEWV

CCGGTGGGGGGCTGGTGC




AGISRGGTTSYADSVKGRFTISADT

AGCCTGGCGGGTCTCTCCG




SKNTLYLQMNSLRAEDTAVYYCGR

CCTCTCTTGTGCCTCCTCCG




YDYDGYYAMDYWGQGTLVTVSS

GCTTTACCTTCAGCAGCTAT






GCTATGTCATGGGTGCGGC






AGGCACCAGGCAAAGGTCT






GGAATGGGTCGCTGGGATC






AGTAGAGGCGGCACAACCT






CCTATGCCGACAGCGTTAA






GGGGAGGTTCACAATCTCG






GCTGATACAAGCAAGAACA






CTCTGTATCTCCAAATGAAC






AGTCTCCGGGCAGAGGACA






CCGCGGTCTATTACTGCGG






CCGGTACGACTACGACGGG






TACTACGCAATGGACTATTG






GGGACAGGGAACTCTGGTT






ACTGTCAGCTCT





hROR1
395
DIQMTQSPSSLSASVGDRVTITCRA
396
GATATCCAGATGACTCAAA


VL_14-2

SPDINSYLSWYQQKPGKAPKLLIYR

GCCCATCTTCTCTCAGCGCA




ANTLESGVPSRFSGSGSGTDFTLTIS

AGCGTGGGTGACCGAGTGA




SLQPEDFATYYCLQYDEFPYTFGTG

CCATCACCTGCCGGGCGTCT




TKLEIK

CCTGATATCAACTCATACCT






GTCCTGGTATCAGCAGAAG






CCCGGAAAGGCCCCTAAGC






TGCTGATCTACCGCGCAAAT






ACACTGGAGAGCGGGGTCC






CAAGCAGATTCAGTGGGTC






CGGCAGTGGTACGGACTTT






ACTCTGACCATCAGCTCCCT






GCAACCGGAGGACTTTGCT






ACTTATTACTGTCTCCAGTA






CGACGAGTTCCCATACACTT






TCGGAACAGGCACTAAGCT






GGAGATCAAA





hROR1
397
EVQLVESGGGLVQPGGSLRLSCAA
398
GAGGTTCAACTTGTGGAAT


VH_14-3

SGFTFSSYAMSWVRQAPGKGLEW

CCGGCGGCGGGTTAGTCCA




VASISRGGTTYYADSVKGRFTISRD

GCCCGGCGGGAGCTTGCGG




NSKNTLYLQMNSLRAEDTAVYYCG

CTGTCCTGCGCCGCCTCTGG




RYDYDGYYAMDYWGQGTLVTVSS

ATTCACTTTTAGCTCCTATG






CTATGTCTTGGGTAAGGCA






GGCCCCTGGTAAAGGACTA






GAGTGGGTGGCCTCGATCT






CCCGTGGTGGCACTACATA






CTACGCCGACTCCGTTAAAG






GCCGGTTTACCATCTCCCGT






GACAACTCTAAAAATACTTT






GTACCTGCAAATGAACTCCC






TGCGGGCAGAAGACACAGC






CGTGTACTATTGCGGGCGT






TACGATTACGACGGATATTA






CGCAATGGACTACTGGGGC






CAGGGCACACTGGTCACCG






TGAGCAGC





hROR1
399
DIQMTQSPSSLSASVGDRVTITCKA
400
GATATACAAATGACTCAGTC


VL_14-3

SPDINSYLNWYQQKPGKAPKLLIYR

CCCTAGTAGCCTTAGTGCTA




ANRLVDGVPSRFSGSGSGTDFTLTI

GTGTGGGAGACAGAGTGAC




SSLQPEDIATYYCLQYDEFPYTFGG

CATCACCTGCAAAGCATCTC




GTKVEIK

CTGATATCAATTCCTACCTT






AACTGGTATCAACAGAAGC






CTGGCAAAGCTCCAAAGCT






CCTGATTTATCGCGCGAACA






GATTGGTCGATGGGGTCCC






TTCCAGATTCAGCGGCTCA






GGGTCAGGGACCGATTTCA






CCCTCACAATTAGTTCACTT






CAGCCCGAGGACATCGCCA






CGTATTATTGCCTTCAGTAC






GATGAGTTCCCTTACACCTT






TGGCGGGGGAACTAAAGTC






GAAATTAAG





hROR1
401
EVQLVESGGGLVQPGGSLRLSCAA
402
GAAGTGCAGCTTGTGGAGT


VH_14-4

SGFTFSSYAMSWVRQAPGKGLEW

CAGGAGGAGGGCTAGTTCA




VASISRGGTTYYPDSVKGRFTISRD

GCCAGGCGGCTCTCTGAGA




NVRNILYLQMSSLRSEDTAMYYCG

CTATCTTGTGCTGCCTCCGG




RYDYDGYYAMDYWGQGTLVTVSS

CTTCACATTTAGCTCTTATG






CAATGTCCTGGGTCCGCCA






GGCCCCTGGTAAAGGCCTG






GAATGGGTTGCTTCTATCTC






TAGAGGCGGAACCACTTAC






TACCCTGATTCAGTGAAGG






GGAGATTCACAATTAGTAG






GGACAACGTGCGGAACATC






CTCTACCTACAGATGTCAAG






TTTACGCAGTGAGGACACT






GCGATGTATTACTGCGGTC






GATACGATTATGATGGATA






TTATGCAATGGATTATTGG






GGCCAGGGCACTCTGGTCA






CAGTATCTTCC





hROR1
403
DIQMTQSPSSLSASVGDRVTITCKA
404
GACATCCAGATGACCCAAT


VL_14-4

SPDINSYLNWYQQKPGKAPKLLIYR

CACCATCGAGTCTTAGTGCA




ANRLVDGVPSRFSGSGSGTDYTLTI

TCCGTTGGGGATAGAGTGA




SSLQPEDFATYYCLQYDEFPYTFGA

CAATCACTTGTAAGGCATCC




GTKVEIK

CCGGACATCAACTCATATCT






TAATTGGTATCAGCAAAAG






CCGGGCAAGGCCCCTAAGC






TCCTGATTTATAGGGCCAAC






CGCCTTGTGGATGGAGTCC






CCTCCCGCTTTAGTGGAAGC






GGCTCTGGCACAGACTACA






CCCTGACTATCAGCTCCTTG






CAGCCTGAGGATTTTGCTAC






CTACTACTGTCTTCAGTACG






ATGAATTTCCATACACTTTC






GGTGCTGGGACAAAAGTGG






AGATCAAA





hROR1
405
EVQLVESGGGLVQPGGSLRLSCAT
406
GAAGTCCAGCTGGTTGAGT


VH_14-5

SGFTFSSYAMSWMRKAPGKGLEY

CTGGCGGAGGCCTCGTGCA




VASISRGGTTYYADSVKGRFTISVDK

GCCCGGTGGTTCCTTGCGA




SKNTLYLQMNSLRAEDTAVYYCGR

CTGTCATGCGCTACCAGCG




YDYDGYYAMDYWGQGTLVTVSS

GGTTCACATTCAGCTCTTAT






GCAATGTCCTGGATGCGGA






AGGCACCGGGTAAGGGCCT






GGAGTATGTGGCCTCAATC






TCCCGAGGAGGCACCACAT






ACTATGCCGATTCTGTGAAA






GGCCGATTCACCATTTCTGT






GGATAAGTCTAAAAACACT






CTCTACCTCCAGATGAACTC






CCTACGTGCCGAAGACACA






GCCGTGTATTATTGCGGGC






GATACGATTATGACGGTTA






TTATGCGATGGATTACTGG






GGTCAAGGCACACTGGTAA






CAGTGTCTTCC





hROR1
407
DIQMTQSPSSLSASVGDRVTITCKA
408
GATATTCAGATGACACAATC


VL_14-5

SPDINSYLNWYQQKPGKAPKLLIYR

ACCTAGCTCACTGTCAGCGA




ANRLVDGVPSRFSGSGSGTDYTLTI

GCGTCGGTGACCGGGTTAC




SSLQPEDFATYYCLQYDEFPYTFGA

TATCACATGCAAAGCCTCAC




GTKVEIK

CCGATATCAATTCATACCTT






AACTGGTATCAACAAAAAC






CAGGAAAGGCTCCAAAGCT






GCTAATTTATCGGGCCAATC






GGTTGGTGGATGGCGTCCC






GTCGAGGTTTAGTGGCTCC






GGGAGCGGGACAGACTAC






ACTCTTACAATTTCTTCTCTC






CAGCCAGAGGACTTCGCAA






CCTACTACTGCTTGCAGTAC






GATGAATTTCCATATACCTT






CGGCGCAGGGACAAAAGT






GGAAATCAAA





hROR1 VH_15
409
EVQLVESGGGLVQPGGSLRLSCVTS
410
GAGGTGCAGCTTGTAGAAA




GFTFSSYAMSWVRQAPGKGLEWV

GCGGGGGGGGCCTGGTGC




ASISRGGTTYYSDSVKGRFTISRDNS

AACCTGGCGGGTCCCTGCG




KNTLYLQMNSLRAEDTAVYYCGRY

GCTTAGTTGCGTTACGAGC




DYDGYYAMDYWGQGTLVTVSS

GGATTTACATTTTCCAGTTA






TGCCATGTCTTGGGTGAGA






CAAGCCCCCGGTAAGGGTC






TGGAGTGGGTGGCAAGCAT






TAGCCGAGGCGGCACTACA






TACTACAGTGATAGTGTGA






AAGGCCGTTTCACAATCAGT






AGAGATAATTCTAAAAACA






CCCTGTACTTGCAGATGAAC






AGCCTGCGCGCCGAGGATA






CAGCCGTGTACTACTGTGG






AAGATACGACTACGATGGA






TATTATGCGATGGATTACTG






GGGACAGGGAACCCTTGTC






ACCGTTTCCTCT





hROR1 VL_15
411
DIVLTQSPATLSLSPGERATLSCKAS
412
GACATAGTGTTGACGCAGT




PDINSYMNWYQQKPGQAPRLLISR

CCCCTGCCACCCTGAGCCTG




ANRLVDGVPARFSGSGSGTDFTLTI

AGCCCCGGAGAGCGAGCAA




SSLEPEDFAVYYCLQYDEFPYTFGQ

CGTTAAGTTGCAAGGCCAG




GTKVEIK

TCCAGATATTAACTCATACA






TGAATTGGTATCAACAGAA






ACCAGGCCAGGCTCCTAGA






CTTCTCATATCTCGGGCAAA






TCGACTGGTGGATGGAGTA






CCCGCAAGATTCAGCGGCA






GCGGCAGCGGAACGGATTT






CACGCTCACCATCTCTTCCC






TTGAGCCTGAGGACTTTGC






AGTCTATTATTGCTTGCAGT






ATGATGAGTTCCCCTACACA






TTCGGGCAAGGCACAAAAG






TGGAAATTAAG





hROR1 VH_16
413
EVQLVESGGGLVQPGGSLRLSCAA
414
GAGGTGCAGCTGGTGGAG




SGFTFSSYAMSWVRQAPGKGLEW

AGCGGAGGGGGCCTTGTCC




VASISRGGTTYYDPKFQDRATISAD

AACCAGGAGGTAGCCTCAG




NSKNTAYLQMNSLRAEDTAVYYCG

GCTGTCTTGCGCTGCCTCAG




RYDYDGYYAMDYWGQGTLVTVSS

GATTTACTTTTTCATCCTAC






GCAATGAGCTGGGTGCGGC






AAGCCCCAGGGAAGGGATT






AGAATGGGTTGCCAGCATT






TCTAGGGGGGGGACGACCT






ACTACGATCCGAAGTTTCAG






GATCGCGCCACTATCTCAGC






CGATAACTCCAAGAATACT






GCCTACTTACAGATGAACA






GCCTGCGGGCCGAAGACAC






GGCCGTCTACTATTGCGGC






CGATATGATTACGACGGCT






ATTACGCCATGGATTACTG






GGGGCAAGGGACTCTGGTC






ACAGTGAGCTCT





hROR1 VL_16
415
DIQMTQSPSSLSASVGDRVTITCKA
416
GATATTCAGATGACCCAGTC




SPDINSYLNWYQQKPGKAPKVLIYR

GCCCAGCAGTCTCTCGGCCT




ANRLVDGVPSRFSGSGSGTDYTLTI

CAGTGGGCGACCGGGTCAC




SSLQPEDFATYYCLQYDEFPYTFGQ

TATCACTTGCAAAGCAAGTC




GTKVEIK

CTGATATAAACTCCTATCTT






AATTGGTATCAGCAGAAGC






CCGGCAAGGCACCTAAGGT






TCTGATATATCGCGCAAATC






GGCTCGTGGATGGAGTACC






CAGCCGATTTTCCGGCAGC






GGCTCAGGCACTGACTACA






CACTGACAATCAGCAGCTT






GCAGCCTGAAGATTTCGCC






ACATACTATTGTCTACAGTA






CGACGAGTTCCCTTATACAT






TCGGCCAGGGGACCAAGGT






CGAGATCAAG





hROR1 VH_17
417
EVQLVESGGGLVQPGGSLRLSCTG
418
GAGGTCCAACTCGTGGAGA




SGFTFSSYAMSWLRQVPGEGLEW

GCGGAGGGGGGCTAGTGC




VSSISRGGTTDYADSVKGRFTISRD

AACCAGGTGGCTCCCTCCG




DAKKTLSLQMNSLRAEDTAVYYCG

CTTGTCCTGTACGGGCTCG




RYDYDGYYAMDYWGQGTMVTVSS

GGGTTCACATTTTCATCCTA






TGCCATGAGCTGGCTGAGA






CAGGTGCCTGGCGAGGGCC






TGGAATGGGTGTCTAGTAT






CAGCAGAGGGGGTACAACT






GATTACGCAGATTCCGTCAA






GGGACGTTTTACCATCTCAA






GAGACGATGCCAAGAAGAC






ATTATCACTCCAAATGAACT






CACTGAGGGCCGAGGACAC






CGCTGTGTACTATTGTGGG






AGATACGACTACGACGGAT






ACTATGCCATGGACTATTG






GGGACAAGGCACGATGGT






GACGGTATCTAGC





hROR1 VL_17
419
EIVLTQSPATLSVSPGERATLSCKAS
420
GAGATAGTGCTAACCCAGT




PDINSYLAWYQQKPGQAPRLLFSR

CTCCCGCAACCCTGTCTGTG




ANRLVDGIPARFTGSGSGTDFTLTIS

TCCCCCGGAGAGCGCGCTA




SLEPEDFAIYYCLQYDEFPYTFGQGT

CTCTGAGCTGCAAAGCCAG




KVEIK

CCCGGACATTAATTCCTACC






TTGCCTGGTATCAGCAGAA






GCCTGGACAGGCCCCAAGA






TTGCTCTTTTCACGCGCCAA






CCGCCTGGTAGATGGTATT






CCAGCTAGGTTTACGGGCT






CAGGCAGCGGAACAGACTT






CACTCTCACTATTAGCTCAT






TGGAGCCTGAGGACTTTGC






AATTTACTATTGTCTTCAGT






ACGACGAGTTCCCATATACT






TTCGGCCAGGGCACAAAAG






TAGAGATCAAG





hROR1 VH_18
421
EVQLVESGGGLVQPGGSLRLSCSAS
422
GAGGTTCAACTCGTGGAGT




GFTFSSYAMSWVRQVPGKGLVWI

CTGGAGGCGGGCTAGTGCA




SSISRGGTTYYADSVRGRFIISRDNA

GCCTGGCGGCTCCCTGCGA




KNTLYLEMNNLRGEDTAVYYCARY

CTGTCTTGCAGCGCATCAG




DYDGYYAMDYWGQGTLVTVSS

GCTTTACATTCAGTTCTTAT






GCCATGAGCTGGGTGAGGC






AGGTGCCCGGCAAGGGTCT






GGTGTGGATCAGCTCAATC






TCCAGGGGCGGGACTACAT






ATTACGCCGATTCGGTCAG






GGGTCGTTTTATCATTAGCA






GGGATAATGCCAAGAACAC






CTTGTATTTGGAGATGAAC






AACCTAAGAGGCGAAGACA






CCGCTGTGTACTATTGTGCC






CGTTACGACTACGATGGGT






ACTACGCCATGGACTATTG






GGGCCAGGGAACCTTGGTG






ACTGTGTCAAGT





hROR1 VL_18
423
DIQLTQSPDSLAVSLGERATINCKAS
424
GACATACAGTTGACTCAGTC




PDINSYLSWYQQRPGQPPRLLIHR

ACCGGATTCGCTGGCAGTT




ANRLVDGVPDRFSGSGFGTDFTLTI

TCGCTGGGTGAGAGAGCAA




TSLQAEDVAIYYCLQYDEFPYTFGQ

CCATCAACTGCAAAGCATCT




GTKLEIK

CCCGATATCAACTCTTATCT






GTCTTGGTATCAGCAGCGT






CCGGGACAACCCCCTAGGC






TGCTTATTCACCGAGCCAAC






AGGCTGGTGGACGGGGTG






CCAGACCGCTTCTCGGGAT






CAGGATTTGGAACCGATTTT






ACCCTAACAATTACTAGTCT






CCAAGCGGAAGACGTGGCG






ATCTATTATTGTCTACAATA






TGACGAGTTCCCCTACACCT






TCGGCCAGGGCACGAAGTT






GGAGATCAAG





hROR1 VH_19
425
EVQLVESGGGLVQPGGSLRLSCAA
426
GAGGTCCAGCTCGTCGAAT




SGFTFSSYAMSWVRQAPGKGLEW

CCGGTGGAGGGCTAGTTCA




VASISRGGTTYYADSVKGRFTISADT

GCCAGGCGGCTCATTGCGT




SKNTAYLQMNSLRAEDTAVYYCAR

TTGTCTTGTGCCGCCTCCGG




YDYDGYYAMDYWGQGTLVTVSS

TTTCACATTCTCTTCTTACGC






TATGTCCTGGGTCCGACAA






GCCCCAGGAAAAGGCTTGG






AATGGGTGGCCAGTATCAG






TAGAGGTGGGACTACATAT






TATGCCGACTCCGTGAAGG






GCAGATTCACCATCTCAGCT






GACACCAGTAAGAACACTG






CCTACCTACAGATGAACAG






CCTTCGGGCCGAGGACACC






GCTGTGTATTACTGTGCCCG






GTACGATTATGATGGATATT






ATGCTATGGACTATTGGGG






TCAGGGGACCTTGGTGACC






GTCTCTAGC





hROR1 VL_19
427
DIQMTQSPSSLSASVGDRVTITCKA
428
GACATTCAGATGACTCAATC




SPDINSYLSWYQQKPGKAPKLLIYR

GCCGAGTTCTCTTAGCGCTT




ANRLVDGVPSRFSGSGSGTDFTLTI

CTGTTGGGGACCGGGTGAC




SSLQPEDFATYYCLQYDEFPYTFGQ

AATCACATGCAAGGCCTCTC




GTKVEIK

CCGATATAAACTCCTATCTA






AGCTGGTATCAGCAGAAGC






CAGGGAAGGCCCCCAAGTT






GTTAATCTATCGCGCCAACA






GACTGGTGGATGGGGTGCC






CTCTCGATTCTCCGGGAGTG






GCAGTGGGACTGATTTTAC






ACTGACCATTTCCTCATTGC






AGCCCGAAGACTTCGCTAC






CTATTACTGCTTGCAGTACG






ATGAGTTCCCATATACATTC






GGTCAGGGGACTAAAGTGG






AGATAAAA





hROR1 VH_20
429
EVQLLESGGGLVQPGGSLRLSCAAS
430
GAGGTACAGCTGCTGGAAT




GFTFSSYAMSWVRQAPGKGLEWV

CTGGTGGGGGGCTGGTCCA




SSISRGGTTYYADSVKGRFTISRDNS

GCCAGGGGGGTCACTACGA




KNTLYLQMNSLRAEDTAVYYCARY

CTGAGCTGCGCTGCCTCCG




DYDGYYAMDYWGQGTLVTVSS

GTTTTACATTCAGCAGCTAT






GCAATGTCATGGGTCAGAC






AGGCACCAGGTAAAGGCCT






CGAATGGGTATCCTCCATCT






CACGTGGTGGGACCACTTA






CTATGCCGATAGTGTGAAG






GGCAGGTTCACGATCTCAA






GAGATAATTCAAAGAATAC






ACTCTATCTACAAATGAACA






GTTTAAGGGCCGAGGACAC






CGCTGTTTACTATTGTGCCA






GATATGACTACGACGGTTA






TTATGCTATGGATTACTGGG






GACAAGGAACGCTGGTAAC






TGTTAGCTCT





hROR1 VL_20
431
DIQMTQSPSSLSASVGDRVTITCKA
432
GACATCCAAATGACCCAGT




SPDINSYLSWYQQKPGEAPKLLIYR

CGCCTTCCTCCTTGTCTGCA




ANRLVDGVPSRFSGSGSGTDFTLTI

TCTGTCGGAGATCGGGTGA




SSLQPEDFATYYCLQYDEFPYTFGQ

CGATCACTTGCAAAGCGAG




GTKVEIK

TCCAGACATCAACTCATATC






TGTCCTGGTATCAGCAGAA






GCCGGGAGAGGCACCTAAG






CTCCTGATCTACAGAGCAAA






CAGATTAGTGGATGGTGTG






CCCTCACGGTTTTCTGGCTC






CGGGTCCGGCACCGATTTC






ACCTTGACCATCTCATCCCT






ACAGCCCGAGGATTTCGCT






ACTTACTATTGCTTACAGTA






TGATGAGTTTCCATACACCT






TCGGTCAAGGCACCAAGGT






TGAGATTAAG





hROR1 VH_21
433
EVQLLETGGGLVKPGGSLRLSCAAS
434
GAAGTTCAACTGCTTGAGA




GFTFSSYAMSWIRQAPGKGLEWV

CCGGAGGCGGCCTGGTAAA




ASISRGGTTYYGDSVKGRFTISRDH

ACCTGGGGGCTCACTGAGG




AKNSLYLQMNSLRVEDTAVYYCVR

CTGAGTTGTGCCGCTTCTGG




YDYDGYYAMDYWGLGTLVTVSS

GTTCACCTTTTCATCCTATG






CGATGTCATGGATACGGCA






GGCTCCTGGGAAGGGGCTT






GAGTGGGTTGCATCAATTT






CACGAGGTGGGACAACTTA






TTATGGGGATTCCGTTAAA






GGTAGATTTACGATCTCTAG






AGACCATGCCAAAAATTCTC






TCTATCTCCAGATGAATAGT






CTTAGGGTGGAGGACACCG






CTGTGTACTACTGTGTCCGG






TACGACTATGATGGGTACT






ATGCTATGGACTATTGGGG






GCTCGGCACTCTGGTCACT






GTTAGCTCT





hROR1 VL_21
435
AIRMTQSPSFLSASVGDRVTITCKA
436
GCCATCCGCATGACACAATC




SPDINSYLSWYQQRPGKAPKLLIYR

TCCCTCCTTCCTTTCTGCCAG




ANRLVDGVPSRFSGGGSGTDFTLTI

TGTCGGGGACAGAGTGACT




SSLQPEDIATYYCLQYDEFPYTFGQ

ATCACATGCAAAGCCAGCC




GTKLEIK

CAGATATTAATTCGTACCTG






TCTTGGTATCAGCAGAGGC






CCGGCAAGGCACCAAAGCT






GTTGATATATCGGGCCAAC






CGCTTAGTGGACGGTGTCC






CCTCTCGATTCAGCGGAGG






CGGTAGCGGGACGGACTTT






ACACTGACCATCTCCAGTCT






CCAACCCGAGGATATTGCC






ACTTACTATTGTCTTCAGTA






TGACGAGTTCCCCTACACAT






TTGGACAGGGCACCAAGCT






AGAAATTAAG





hROR1 VH_22
437
EVQLVESGGGLVQPGGSLRLSCAA
438
GAGGTTCAGCTGGTGGAGT




SGFTFSSYAMSWVRQAPGKGLEW

CTGGTGGGGGGCTCGTACA




VASISRGGTTYYAESLEGRFTISRDD

GCCGGGTGGCTCCCTAAGG




SKNSLYLQMNSLKTEDTAVYYCARY

CTGAGTTGCGCTGCCTCAG




DYDGYYAMDYWGQGTLVTVSS

GCTTTACCTTCTCAAGCTAC






GCGATGTCCTGGGTGAGAC






AGGCCCCTGGCAAAGGACT






GGAGTGGGTGGCAAGCATT






AGCCGGGGCGGAACTACCT






ATTACGCTGAGTCGTTAGA






GGGGCGGTTTACTATCTCC






AGAGACGATTCAAAGAACT






CGTTATACTTGCAGATGAAC






AGCCTCAAGACCGAGGACA






CCGCCGTGTACTACTGCGCC






CGGTACGACTATGACGGGT






ACTATGCTATGGATTATTGG






GGACAAGGCACCCTCGTGA






CCGTCTCTAGC





hROR1 VL_22
439
DIQMTQSPSSLSASVGDRVTITCKA
440
GACATCCAGATGACACAGT




SPDINSYLSWYQQKPGKAPKTLIYR

CCCCTTCTTCACTTTCCGCTT




ANRLVDGVPSRFSGSGSGTDFTLTI

CTGTGGGCGACAGGGTGAC




SSLQPEDFATYYCLQYDEFPYTFGQ

GATCACGTGTAAGGCCTCG




GTKLEIK

CCAGACATTAATTCGTACTT






ATCGTGGTATCAGCAGAAA






CCGGGTAAAGCTCCGAAGA






CTCTGATCTATAGAGCAAAT






AGGCTCGTAGACGGTGTCC






CATCTAGATTTAGTGGGAG






CGGCAGCGGAACCGACTTC






ACTCTCACCATCTCATCCCT






GCAACCGGAGGATTTCGCT






ACTTACTATTGCTTGCAGTA






TGACGAGTTTCCATATACGT






TTGGTCAGGGAACCAAATT






AGAGATCAAA





hROR1 VH_23
441
QVTLRESGPALVKPTQTLTLTCAAS
442
CAGGTAACACTCCGAGAGA




GFTFSSYAMSWIRQPPGKALEWLA

GTGGGCCAGCTCTCGTGAA




SISRGGTTYYNPSLKDRLTISKDTSA

GCCCACGCAGACTTTAACAC




NQVVLKVTNMDPADTATYYCARY

TAACGTGTGCGGCAAGCGG




DYDGYYAMDYWGQGTTVTVSS

CTTTACATTTTCGAGCTACG






CGATGAGCTGGATAAGGCA






ACCTCCTGGGAAGGCGTTG






GAGTGGTTGGCCTCAATTA






GCCGGGGTGGCACCACTTA






CTACAATCCTAGTCTTAAGG






ACAGACTTACTATTTCAAAA






GATACGTCCGCCAACCAGG






TGGTACTGAAGGTCACAAA






TATGGACCCAGCTGACACT






GCTACTTACTACTGCGCCCG






GTACGATTACGATGGTTACT






ACGCTATGGATTACTGGGG






TCAAGGAACCACAGTGACC






GTCAGTTCA





hROR1 VL_23
443
DIQMTQSPSTLSASVGDRVTITCKA
444
GATATCCAGATGACGCAGT




SPDINSYLSWYQQKPGKAPKLLIYR

CCCCTTCAACCCTCAGTGCC




ANRLVDGVPSRFSGSGSGTAFTLTI

AGCGTTGGTGACCGGGTTA




SSLQPDDFATYYCLQYDEFPYTFGG

CTATCACCTGTAAGGCTAGT




GTKVEIK

CCCGATATAAATTCCTATTT






GTCTTGGTATCAGCAGAAG






CCAGGCAAGGCTCCTAAGC






TGCTCATCTACCGGGCTAAC






AGGTTAGTTGACGGTGTGC






CCTCCCGATTTTCCGGCAGT






GGCAGCGGGACCGCTTTCA






CTCTTACAATCTCATCTCTTC






AACCGGACGACTTCGCTAC






GTACTACTGCCTCCAATATG






ATGAGTTTCCATACACATTC






GGAGGAGGCACAAAAGTC






GAAATCAAG





hROR1 VH_24
445
EVQLVESGGGLVQPGGSLRLSCAA
446
GAAGTCCAGCTGGTGGAGT




SGFTFSSYAMSWVRQAPGKGLEW

CCGGCGGAGGCTTGGTTCA




VSAISRGGTTYYADSVKGRFTISADT

GCCCGGAGGATCTTTGCGA




SKETAYLQMNSLRAEDTAVYYCGR

CTGTCTTGCGCCGCCAGCG




YDYDGYYAMDYWGQGTLVTVSS

GTTTCACTTTCAGCAGCTAT






GCCATGAGTTGGGTTAGAC






AAGCTCCCGGCAAGGGGCT






GGAATGGGTTAGTGCTATT






AGCCGGGGAGGGACAACA






TATTACGCTGACTCTGTCAA






AGGCCGATTCACCATCTCTG






CTGACACGAGCAAAGAAAC






CGCCTACCTCCAAATGAACA






GCCTGCGAGCTGAGGACAC






TGCCGTCTACTATTGTGGTC






GATATGATTATGATGGGTA






CTATGCAATGGACTATTGG






GGGCAGGGCACACTGGTGA






CCGTGAGCTCT





hROR1 VL_24
447
DIQMTQSPSSLSASVGDRVTITCKA
448
GATATTCAGATGACGCAGA




SPDINSYLSWYQQKPGKAPKLLIYR

GTCCCTCCTCCCTATCTGCC




ANRLVDGVPSRFSGSGSGTDFTLTI

TCTGTTGGAGATCGAGTCA




SSLQPEDIATYYCLQYDEFPYTFGQ

CCATTACGTGTAAAGCGTCT




GTKLEIK

CCCGATATCAACAGCTACCT






CTCTTGGTATCAGCAGAAAC






CAGGGAAGGCCCCCAAGCT






GCTGATCTATAGAGCTAATC






GCTTAGTGGATGGAGTGCC






AAGCAGGTTCTCCGGGTCC






GGCAGTGGAACCGATTTCA






CCTTGACAATAAGTAGCTTG






CAACCTGAGGATATTGCAA






CATACTACTGTCTACAGTAC






GACGAGTTCCCCTACACCTT






CGGCCAAGGGACAAAGCTG






GAGATTAAG





hROR1 VH_25
449
EVQLVESGGGLVQPGGSLRLSCAA
450
GAAGTGCAGCTCGTGGAGA




SGFTFSSYAMSWVRQAPGKGLEW

GCGGCGGCGGTCTGGTACA




VSAISRGGTTYYADSVKGRFTISRD

GCCAGGGGGGTCACTGCGT




NSKNTLYLQMNSLRAEDTAVYYCG

CTCTCATGTGCTGCGAGTG




RYDYDGYYAMDYWGQGTLVTVSS

GCTTTACGTTCTCTTCCTAC






GCTATGTCCTGGGTCAGGC






AGGCACCGGGGAAGGGCTT






AGAGTGGGTTAGTGCAATC






TCTAGGGGCGGTACAACCT






ACTATGCCGACTCTGTCAAG






GGCAGGTTTACAATTTCAA






GAGATAATTCTAAGAATACT






CTTTACCTACAGATGAATAG






CTTGCGGGCGGAAGACACA






GCAGTCTATTATTGTGGCCG






CTATGACTACGACGGATACT






ATGCCATGGACTACTGGGG






CCAAGGCACTTTGGTCACG






GTGAGCTCT





hROR1 VL_25
451
DIQMTQSPSSLSASVGDRVTITCKA
452
GACATCCAGATGACCCAGA




SPDINSYLSWYQQKPGKAPKLLIYR

GCCCTAGTTCATTGTCTGCC




ANRLVDGVPSRFSGSGSGTDFTLTI

AGTGTGGGGGATAGGGTC




SSLQPEDIATYYCLQYDEFPYTFGQ

ACTATCACGTGTAAGGCTTC




GTKLEIK

CCCTGACATCAATTCATACC






TGTCATGGTATCAGCAGAA






GCCTGGAAAAGCCCCTAAA






CTGCTGATCTACCGCGCGA






ATAGGCTTGTGGACGGCGT






TCCAAGCCGCTTCTCTGGCT






CTGGATCAGGGACCGACTT






CACCCTCACGATCTCCAGCC






TCCAACCCGAGGATATCGC






CACCTATTATTGCCTTCAGT






ACGATGAGTTCCCCTATACA






TTCGGCCAGGGGACAAAGC






TGGAAATCAAA





hROR1 VH_26
453
EVQLVESGGGLVQPGGSLRLSCAA
454
GAGGTCCAGCTCGTCGAGT




SGFTFSSYAMSWVRQAPGKGLEW

CGGGTGGGGGCTTGGTGCA




VSAISRGGTTYYADSVKGRFTISADT

ACCCGGTGGCAGTTTGCGC




SKETAYLQMNSLRAEDTAVYYCGR

CTGAGCTGCGCCGCGAGCG




YDYDGYYAMDYWGQGTLVTVSS

GGTTCACTTTCAGTTCGTAT






GCCATGAGTTGGGTGCGAC






AAGCGCCCGGCAAAGGACT






GGAGTGGGTGTCAGCCATT






AGCCGGGGCGGTACTACCT






ACTATGCGGACTCGGTCAA






GGGAAGATTCACCATCAGC






GCTGATACCAGTAAGGAAA






CCGCTTATCTTCAGATGAAC






TCCCTGCGTGCCGAGGATA






CAGCAGTCTACTATTGCGG






GCGCTACGATTATGACGGA






TATTATGCCATGGATTACTG






GGGGCAGGGCACTCTGGTC






ACAGTCAGCTCT





hROR1 VL_26
455
DIQMTQSPSSLSASVGDRVTITCQA
456
GATATTCAGATGACGCAGT




SPDINSYLNWYQQKPGKAPKLLIYR

CTCCCTCTTCCCTGAGCGCC




ANNLETGVPSRFSGSGSGTDFTLTI

TCCGTCGGCGATAGAGTTA




SSLQPEDIATYYCLQYDEFPYTFGQ

CGATCACCTGTCAGGCCAG




GTKLEIK

CCCAGATATCAACTCCTATC






TGAATTGGTATCAGCAAAA






GCCTGGGAAGGCTCCCAAG






TTGCTGATCTACAGAGCCAA






TAACTTAGAGACTGGCGTG






CCGTCTCGGTTCAGCGGGT






CCGGCAGTGGAACCGACTT






TACACTGACCATTTCCAGCC






TCCAACCTGAGGATATCGCC






ACATATTATTGTCTCCAGTA






TGACGAGTTCCCTTACACAT






TTGGTCAAGGAACTAAACT






GGAAATCAAA





hROR1 VH_27
457
EVQLVESGGGLVQPGGSLRLSCAA
458
GAGGTGCAGCTGGTCGAAA




SGFTFSSYAMSWVRQAPGKGLEW

GTGGAGGCGGACTCGTGCA




VSAISRGGTTYYADSVKGRFTISADT

GCCCGGCGGTAGTCTGCGA




SKETAYLQMNSLRAEDTAVYYCGR

TTGAGCTGTGCCGCGTCCG




YDYDGYYAMDYWGQGTLVTVSS

GCTTTACTTTCTCATCTTACG






CTATGAGTTGGGTCCGCCA






GGCCCCAGGCAAAGGACTG






GAGTGGGTATCAGCCATCA






GTAGGGGGGGAACTACCTA






TTACGCAGATTCTGTGAAG






GGACGCTTCACCATCAGCG






CGGACACTAGCAAGGAGAC






TGCCTACCTGCAAATGAATA






GTCTGAGAGCCGAGGATAC






CGCCGTGTACTATTGTGGC






AGGTATGACTACGATGGCT






ATTATGCTATGGATTACTGG






GGCCAGGGGACGTTAGTGA






CAGTAAGCTCT





hROR1 VL_27
459
DIQMTQSPSSLSASVGDRVTITCRA
460
GATATTCAGATGACCCAATC




SPDINSYVAWYQQKPGKAPKLLIYR

CCCTTCTTCTCTGAGCGCTT




ANFLESGVPSRFSGSRSGTDFTLTIS

CTGTGGGCGATAGAGTTAC




SLQPEDFATYYCLQYDEFPYTFGQG

AATAACCTGTCGGGCGTCC




TKVEIK

CCAGACATTAACTCTTATGT






AGCATGGTATCAGCAAAAG






CCTGGAAAGGCACCAAAGT






TACTGATCTACCGGGCCAAT






TTTCTGGAGTCGGGCGTGC






CCTCACGATTTAGCGGTAG






CAGATCAGGCACAGACTTT






ACTCTGACCATTAGCTCTCT






GCAACCCGAGGACTTCGCC






ACCTACTACTGTTTGCAGTA






TGACGAGTTTCCATACACTT






TTGGTCAAGGAACCAAAGT






CGAAATCAAA





hROR1 VH_28
461
QIQLVQSGAEVKKPGASVKVSCAA
462
CAGATACAGCTGGTGCAGT




SGFTFSSYAMSWVRQAPGKSFKW

CTGGTGCCGAGGTTAAAAA




MGSISRGGTTYYSADFKGRFAITKD

GCCCGGAGCCTCGGTTAAA




TSASTAYMELSSLRSEDTAVYYCAR

GTGAGTTGTGCGGCAAGCG




YDYDGYYAMDYWGQGTLVTVSS

GATTCACGTTCAGTTCCTAC






GCTATGTCCTGGGTGCGGC






AGGCTCCTGGCAAGTCATTT






AAGTGGATGGGGTCGATCT






CACGGGGTGGAACCACCTA






TTACTCTGCCGACTTCAAGG






GGAGATTTGCGATTACAAA






AGATACAAGCGCCTCTACG






GCCTACATGGAGTTAAGTA






GCCTTAGAAGCGAAGACAC






GGCGGTGTACTACTGCGCC






AGATATGACTATGACGGCT






ACTACGCCATGGACTACTG






GGGCCAGGGCACACTGGTT






ACAGTCAGCTCT





hROR1 VL_28
463
DIVMTQSPDSLAVSLGERATISCKA
464
GATATCGTGATGACACAAA




SPDINSYLSWYQQKPGQPPKLLIYR

GCCCAGACAGTCTGGCAGT




ANRLVDGVPDRFSGSGSRTDFTLTI

GTCCCTCGGCGAGCGCGCT




SSLQAEDVAVYYCLQYDEFPYTFGQ

ACCATCTCATGCAAAGCTAG




GTKVEIK

TCCCGACATCAATTCCTATC






TGTCCTGGTATCAGCAAAA






ACCAGGCCAACCCCCCAAG






CTGCTTATCTATCGGGCTAA






CCGATTAGTCGATGGGGTG






CCAGATAGATTTTCAGGCTC






TGGTTCCCGGACAGATTTTA






CTCTCACGATCTCCTCACTA






CAGGCAGAAGATGTTGCAG






TGTATTACTGCCTGCAATAC






GACGAGTTCCCCTACACCTT






CGGCCAAGGCACGAAAGTG






GAGATCAAG





hROR scFv
465
EVQLVESGGGLVQPGGSLRLSCAA
466
GAAGTGCAACTGGTCGAGT




SGFTFSSYAMSWVRQAPGKGLEW

CTGGGGGCGGCCTTGTGCA




VSSISRGGTTYYPDSVKGRFTISRDN

ACCTGGAGGCAGCCTTCGA




SKNTLYLQMNSLRAEDTAVYYCGR

CTCAGTTGCGCCGCGTCTG




YDYDGYYAMDYWGQGTLVTVSSG

GTTTTACCTTCTCCTCTTACG




GGGSGGGGSGGGGSDIQMTQSPS

CGATGAGCTGGGTTCGCCA




SLSASVGDRVTITCKASPDINSYLN

GGCCCCCGGCAAGGGACTT




WYQQKPGKAPKLLIYRANRLVDGV

GAGTGGGTTAGTTCGATCT




PSRFSGSGSGTDYTLTISSLQPEDFA

CCCGCGGAGGCACCACATA




TYYCLQYDEFPYTFGAGTKVEIK

TTATCCTGACTCGGTTAAGG






GACGCTTCACTATCTCTAGG






GACAATTCAAAGAACACAC






TGTATCTCCAAATGAACTCC






TTGCGGGCCGAGGACACTG






CTGTGTATTATTGCGGACG






ATACGACTACGATGGGTAT






TACGCCATGGATTACTGGG






GGCAAGGTACACTGGTCAC






TGTGAGTTCG






GGGGGCGGCGGAAGTGGT






GGAGGGGGAAGTGGTGGA






GGAGGAAGCGATATACAGA






TGACACAGAGCCCTTCAAG






TTTATCTGCAAGCGTCGGC






GATCGTGTTACAATAACTTG






CAAGGCATCTCCCGACATCA






ATTCCTACCTCAACTGGTAT






CAGCAGAAGCCTGGGAAG






GCTCCTAAGCTGCTTATTTA






CAGAGCAAATCGCCTGGTG






GACGGCGTGCCCAGTCGGT






TTTCCGGGTCTGGGAGCGG






AACGGATTACACACTGACC






ATCTCAAGCCTGCAACCCGA






AGACTTCGCTACATATTACT






GCCTTCAGTATGATGAGTTC






CCATATACCTTCGGCGCTGG






GACCAAGGTGGAGATAAAG









Anti-ROR1 CDRs
















SEQ ID NO







IMGT Method




VH-CDR-1
GFTFSSYA
715





VH-CDR-2
ISRGGTT
716





VH-CDR-3
GRYDYDGYYAMDY
717





Kabat Method




VH-CDR-1

SYAMS

718





VH-CDR-2

AISRGGTTYYADSVKG

719





VH-CDR-3

YDYDGYYAMDY

720





IMGT Method




VL-CDR-1
PDINSY
721





VL-CDR-2
RAN
722





VL-CDR-3
LQYDEFPYT
723





Kabat Method




VL-CDR-1

RASPDINSYLS

724





VL-CDR-2

RANTLES

725





VL-CDR-3

LQYDEFPYT

723









Portions of ROR1-specific antigen binding domain
















Portion of VH
GFTFSSYAMSWVRQAPGKGLEWVSSISRGGTTYYPDSVKGRFT
SEQ ID NO: 726


Domain
ISRDNSKNTLYLQMNSLRAEDTAVYYCGRYDYDGYYAMDY






Portion of VL
ASPDINSYLNWYQQKPGKAPKLLIYRANRLVDGVPSRFSGSGS
SEQ ID NO: 727


Domain
GTDYTLTISSLQPEDFATYYCLQYDEFPYT






Portion of VH
SYAMSWVRQAPGKGLEWVSSISRGGTTYYPDSVKGRFTISRDN
SEQ ID NO: 728


Domain
SKNTLYLQMNSLRAEDTAVYYCGRYDYDGYYAMDY






Portion of VL
PDINSYLNWYQQKPGKAPKLLIYRANRLVDGVPSRFSGSGSGT
SEQ ID NO: 729


Domain
DYTLTISSLQPEDFATYYCLQYDEFPYT









Exemplary Spacer Sequences


















CD8α hinge
467
KPTTTPAPRPPTPAPTIASQPLSLRP
468
AAGCCCACCACCACCCCTGC




EACRPAAGGAVHTRGLDFACD

CCCTAGACCTCCAACCCCAG






CCCCTACAATCGCCAGCCAG






CCCCTGAGCCTGAGGCCCG






AAGCCTGTAGACCTGCCGC






TGGCGGAGCCGTGCACACC






AGAGGCCTGGATTTCGCCT






GCGAC





CD8α hinge-
469
KPTTTPAPRPPTPAPTIASQPLSLRP
470
AAGCCTACCACCACCCCCGC


homologous

EASRPAAGGAVHTRGLDFASD

ACCTCGTCCTCCAACCCCTG


stalk



CACCTACGATTGCCAGTCAG


extension



CCTCTTTCACTGCGGCCTGA


region



GGCCAGCAGACCAGCTGCC






GGCGGTGCCGTCCATACAA






GAGGACTGGACTTCGCGTC






CGAT





CD8α hinge-
469
KPTTTPAPRPPTPAPTIASQPLSLRP
471
AAACCTACTACCACTCCAGC


homologous

EASRPAAGGAVHTRGLDFASD

CCCAAGGCCCCCAACCCCA


stalk



GCACCGACTATCGCATCACA


extension



GCCTTTGTCACTGCGTCCTG


region



AAGCCAGCCGGCCAGCTGC






AGGGGGGGCCGTCCACACA






AGGGGACTCGACTTTGCGA






GTGAT





CD8α hinge-
469
KPTTTPAPRPPTPAPTIASQPLSLRP
472
AAACCTACTACAACTCCTGC


homologous

EASRPAAGGAVHTRGLDFASD

CCCCCGGCCTCCTACACCAG


stalk



CTCCTACTATCGCCTCCCAG


extension



CCACTCAGTCTCAGACCCGA


region



GGCTTCTAGGCCAGCGGCC






GGAGGCGCGGTCCACACCC






GCGGGCTGGACTTTGCATC






CGAT





CD8α hinge
473
KPTTTPAPRPPTPAPTIASOPLSLRP
474
AAGCCTACCACCACCCCCGC


and 2 CD8a-

EASRPAAGGAVHTRGLDFASDKPT

ACCTCGTCCTCCAACCCCTG


homologous

TTPAPRPPTPAPTIASQPLSLRPEAS

CACCTACGATTGCCAGTCAG


stalk

RPAAGGAVHTRGLDFASDKPTTTP

CCTCTTTCACTGCGGCCTGA


extension

APRPPTPAPTIASQPLSLRPEACRPA

GGCCAGCAGACCAGCTGCC


regions

AGGAVHTRGLDFACD

GGCGGTGCCGTCCATACAA






GAGGACTGGACTTCGCGTC






CGATAAACCTACTACCACTC






CAGCCCCAAGGCCCCCAAC






CCCAGCACCGACTATCGCAT






CACAGCCTTTGTCACTGCGT






CCTGAAGCCAGCCGGCCAG






CTGCAGGGGGGGCCGTCCA






CACAAGGGGACTCGACTTT






GCGAGTGATAAGCCCACCA






CCACCCCTGCCCCTAGACCT






CCAACCCCAGCCCCTACAAT






CGCCAGCCAGCCCCTGAGC






CTGAGGCCCGAAGCCTGTA






GACCTGCCGCTGGCGGAGC






CGTGCACACCAGAGGCCTG






GATTTCGCCTGCGAC









Exemplary Transmembrane Domain Sequences


















CD8a
475
IYIWAPLAGTCGVLLLSLVITLYCNH
476
ATCTACATCTGGGCCCCTCT


Trans-

RN

GGCCGGCACCTGTGGCGTG


membrane



CTGCTGCTGAGCCTGGTCAT


Domain



CACCCTGTACTGCAACCACC






GGAAT






477
FWVLVVVGGVLACYSLLVTVAFIIF
478
TTTTGGGTGCTGGTGGTGG


CD28

WVRSKRS

TTGGTGGAGTCCTGGCTTG


Trans-



CTATAGCTTGCTAGTAACAG


membrane 



TGGCCTTTATTATTTTCTGG


Domain



GTGAGGAGTAAGAGGAGC









Exemplary Intracellular Signaling Domain Sequences


















CD3ζ signaling
479
RVKFSRSADAPAYQQGQNQLYNEL
480
CGGGTGAAGTTCAGCCGGA


domain

NLGRREEYDVLDKRRGRDPEMGG

GCGCCGACGCCCCTGCCTA




KPRRKNPQEGLYNELQKDKMAEA

CCAGCAGGGCCAGAACCAG




YSEIGMKGERRRGKGHDGLYQGLS

CTGTACAACGAGCTGAACC




TATKDTYDALHMQALPPR

TGGGCCGGAGGGAGGAGT






ACGACGTGCTGGACAAGCG






GAGAGGCCGGGACCCTGA






GATGGGCGGCAAGCCCCG






GAGAAAGAACCCTCAGGAG






GGCCTGTATAACGAACTGC






AGAAAGACAAGATGGCCGA






GGCCTACAGCGAGATCGGC






ATGAAGGGCGAGCGGCGG






AGGGGCAAGGGCCACGAC






GGCCTGTACCAGGGCCTGA






GCACCGCCACCAAGGATAC






CTACGACGCCCTGCACATGC






AGGCCCTGCCCCCCAGA





CD28 co-
481
RSKRSRGGHSDYMNMTPRRPGPT
482
AGGAGCAAGCGGAGCAGA


stimulatory

RKHYOPYAPPRDFAAYRS

GGCGGCCACAGCGACTACA


domain



TGAACATGACCCCCCGGAG






GCCTGGCCCCACCCGGAAG






CACTACCAGCCCTACGCCCC






TCCCAGGGACTTCGCCGCCT






ACCGGAGC





4-1BB co-
483
KRGRKKLLYIFKQPFMRPVQTTQEE
484
AAGAGAGGCCGGAAGAAA


stimulatory

DGCSCRFPEEEEGGCEL

CTGCTGTACATCTTCAAGCA


domain



GCCCTTCATGCGGCCCGTG






CAGACCACCCAGGAAGAGG






ACGGCTGCAGCTGCCGGTT






CCCCGAGGAAGAGGAAGG






CGGCTGCGAACTG





DNAX-
485
LCARPRRSPAQEDGKVYINMPGRG
486
CTGTGCGCACGCCCACGCC


activation



GCAGCCCCGCCCAAGAAGA


protein 10



TGGCAAAGTCTACATCAAC


(DAP10) co-



ATGCCAGGCAGGGGC


stimulatory






domain









DNAX-
487
YFLGRLVPRGRGAAEAATRKQRITE
488
TACTTCCTGGGCCGGCTGG


activation

TESPYQELQGQRSDVYSDLNTQRP

TCCCTCGGGGGCGAGGGGC


protein 12

YYK

TGCGGAGGCAGCGACCCG


(DAP12) co-



GAAACAGCGTATCACTGAG


stimulatory



ACCGAGTCGCCTTATCAGG






AGCTCCAGGGTCAGAGGTC






GGATGTCTACAGCGACCTC






AACACACAGAGGCCGTATT






ACAAA









Exemplary Signal Peptide Sequences


















GM-CSFRa signal
489
MLLLVTSLLLCELPHPAFLLIP
490
ATGCTGCTGCTGGTGACCA


peptide



GCCTGCTGCTGTGTGAGCT






GCCCCACCCCGCCTTTCTGC






TGATCCCC





Ig Kappa signal
491
MRLPAQLLGLLMLWVPGSSG
492
ATGAGGCTCCCTGCTCAGCT


peptide



CCTGGGGCTGCTAATGCTCT






GGGTCCCAGGATCCAGTGG






G





Immuno-globulin
493
MDWTWILFLVAAATRVHS
494
ATGGATTGGACCTGGATTCT


E signal peptide



GTTTCTGGTGGCCGCTGCCA






CAAGAGTGCACAGC





CD8α signal
495
MALPVTALLLPLALLLHAARP
496
ATGGCGCTGCCCGTGACCG


peptide



CCTTGCTCCTGCCGCTGGCC






TTGCTGCTCCACGCCGCCAG






GCCG





Mouse Ig VH
497
MGWSCIILFLVATATGVHS
498
ATGGGCTGGTCCTGCATCAT


region 3 signal



CCTGTTTCTGGTGGCTACCG


peptide



CCACCGGCGTGCACAGC





β2M signal
499
MSRSVALAVLALLSLSGLEA
500
ATGTCTCGCTCCGTGGCCTT


peptide



AGCTGTGCTCGCGCTACTCT





Azurocidin signal
501
MTRLTVLALLAGLLASSRA
502
CTCTTTCTGGCCTGGAGGCT


peptide



ATGACCCGGCTGACAGTCCT






GGCCCTGCTGGCTGGTCTG





Human Serum
503
MKWVTFISLLFLFSSAYS
504
CTGGCGTCCTCGAGGGCC


Albumin signal



ATGAAGTGGGTAACCTTTAT


peptide



TTCCCTTCTTTTTCTCTTTAG






CTCGGCTTATTCC





A2M receptor
505
MGKNKLLHPSLVLLLLVLLPTDA
506
ATGGGGAAGAACAAACTCC


associated



TTCATCCAAGTCTGGTTCTT


protein signal



CTCCTCTTGGTCCTCCTGCCC


peptide



ACAGACGCC





IGHV3-23 signal
507
MEFGLSWLFLVAILKGVQC
508
ATGGAGTTTGGGCTGAGCT


peptide



GGCTTTTTCTTGTGGCTATTT






TAAAAGGTGTCCAGTGT





IGKV1-D33
509
MDMRVPAQLLGLLLLWLSGARC
510
ATGGACATGAGGGTCCCTG


(HuL1) signal



CTCAGCTCCTGGGGCTCCTG


peptide



CTGCTCTGGCTCTCAGGTGC






CAGATGT





IGHV3-33(L14F)
511
MEFGLSWVFLVALFRGVQC
512
ATGGAGTTTGGGCTGAGCT


(HuH7) signal



GGGTTTTCCTCGTTGCTCTTT


peptide



TTAGAGGTGTCCAGTGT





TVB2 (T21A)
513
MGTSLLCWMALCLLGADHADA
514
ATGGGCACCAGCCTCCTCTG


signal peptide



CTGGATGGCCCTGTGTCTCC






TGGGGGCAGATCACGCAGA






TGCT





CD52 signal
515
MKRFLFLLLTISLLVMVQIQTGLS
516
ATGAAGCGCTTCCTCTTCCT


peptide



CCTACTCACCATCAGCCTCC






TGGTTATGGTACAGATACAA






ACTGGACTCTCA





Low-affinity
517
MGAGATGRAMDGPRLLLLLLLGVS
518
ATGGGGGCAGGTGCCACCG


nerve growth

LGGA

GCCGCGCCATGGACGGGCC


factor receptor



GCGCCTGCTGCTGTTGCTGC


(LNGFR,



TTCTGGGGGTGTCCCTTGGA


TNFRSF16) signal



GGTGCC


peptide









Exemplary Cytokine Sequences


















IL-15
519
NWVNVISDLKKIEDLIQSMHIDATL
520
AACTGGGTGAATGTGATCA




YTESDVHPSCKVTAMKCFLLELQVI

GCGACCTGAAGAAGATCGA




SLESGDASIHDTVENLIILANNSLSS

GGATCTGATCCAGAGCATG




NGNVTESGCKECEELEEKNIKEFLQ

CACATTGATGCCACCCTGTA




SFVHIVQMFINTS

CACAGAATCTGATGTGCAC






CCTAGCTGTAAAGTGACCG






CCATGAAGTGTTTTCTGCTG






GAGCTGCAGGTGATTTCTCT






GGAAAGCGGAGATGCCTCT






ATCCACGACACAGTGGAGA






ATCTGATCATCCTGGCCAAC






AATAGCCTGAGCAGCAATG






GCAATGTGACAGAGTCTGG






CTGTAAGGAGTGTGAGGAG






CTGGAGGAGAAGAACATCA






AGGAGTTTCTGCAGAGCTT






TGTGCACATCGTGCAGATG






TTCATCAATACAAGC





IL-15Rα
521
ITCPPPMSVEHADIWVKSYSLYSRE
522
ATTACATGCCCTCCTCCAAT




RYICNSGFKRKAGTSSLTECVLNKA

GTCTGTGGAGCACGCCGAT




TNVAHWTTPSLKCIRDPALVHQRP

ATTTGGGTGAAGTCCTACA




APPSTVTTAGVTPQPESLSPSGKEP

GCCTGTACAGCAGAGAGAG




AASSPSSNNTAATTAAIVPGSQLM

ATACATCTGCAACAGCGGC




PSKSPSTGTTEISSHESSHGTPSQTT

TTTAAGAGAAAGGCCGGCA




AKNWELTASASHQPPGVYPQGHS

CCTCTTCTCTGACAGAGTGC




DTTVAISTSTVLLCGLSAVSLLACYLK

GTGCTGAATAAGGCCACAA




SRQTPPLASVEMEAMEALPVTWG

ATGTGGCCCACTGGACAAC




TSSRDEDLENCSHHL

ACCTAGCCTGAAGTGCATTA






GAGATCCTGCCCTGGTCCA






CCAGAGGCCTGCCCCTCCAT






CTACAGTGACAACAGCCGG






AGTGACACCTCAGCCTGAA






TCTCTGAGCCCTTCTGGAAA






AGAACCTGCCGCCAGCTCTC






CTAGCTCTAATAATACCGCC






GCCACAACAGCCGCCATTG






TGCCTGGATCTCAGCTGAT






GCCTAGCAAGTCTCCTAGCA






CAGGCACAACAGAGATCAG






CAGCCACGAATCTTCTCACG






GAACACCTTCTCAGACCACC






GCCAAGAATTGGGAGCTGA






CAGCCTCTGCCTCTCACCAG






CCTCCAGGAGTGTATCCTCA






GGGCCACTCTGATACAACA






GTGGCCATCAGCACATCTAC






AGTGCTGCTGTGTGGACTG






TCTGCCGTGTCTCTGCTGGC






CTGTTACCTGAAGTCTAGAC






AGACACCTCCTCTGGCCTCT






GTGGAGATGGAGGCCATG






GAAGCCCTGCCTGTGACAT






GGGGAACAAGCAGCAGAG






ATGAGGACCTGGAGAATTG






TTCTCACCACCTG





mbIL15
523
NWVNVISDLKKIEDLIQSMHIDATL
524
AACTGGGTGAATGTGATCA




YTESDVHPSCKVTAMKCFLLELQVI

GCGACCTGAAGAAGATCGA




SLESGDASIHDTVENLIILANNSLSS

GGATCTGATCCAGAGCATG




NGNVTESGCKECEELEEKNIKEFLQ

CACATTGATGCCACCCTGTA




SFVHIVQMFINTSSGGGSGGGGSG

CACAGAATCTGATGTGCAC




GGGSGGGGSGGGSLQITCPPPMS

CCTAGCTGTAAAGTGACCG




VEHADIWVKSYSLYSRERYICNSGF

CCATGAAGTGTTTTCTGCTG




KRKAGTSSLTECVLNKATNVAHWT

GAGCTGCAGGTGATTTCTCT




TPSLKCIRDPALVHQRPAPPSTVTT

GGAAAGCGGAGATGCCTCT




AGVTPQPESLSPSGKEPAASSPSSN

ATCCACGACACAGTGGAGA




NTAATTAAIVPGSQLMPSKSPSTGT

ATCTGATCATCCTGGCCAAC




TEISSHESSHGTPSQTTAKNWELTA

AATAGCCTGAGCAGCAATG




SASHQPPGVYPQGHSDTTVAISTST

GCAATGTGACAGAGTCTGG




VLLCGLSAVSLLACYLKSRQTPPLAS

CTGTAAGGAGTGTGAGGAG




VEMEAMEALPVTWGTSSRDEDLE

CTGGAGGAGAAGAACATCA




NCSHHL

AGGAGTTTCTGCAGAGCTT






TGTGCACATCGTGCAGATG






TTCATCAATACAAGCTCTGG






CGGAGGATCTGGAGGAGG






CGGATCTGGAGGAGGAGG






CAGTGGAGGCGGAGGATCT






GGCGGAGGATCTCTGCAGA






TTACATGCCCTCCTCCAATG






TCTGTGGAGCACGCCGATA






TTTGGGTGAAGTCCTACAG






CCTGTACAGCAGAGAGAGA






TACATCTGCAACAGCGGCTT






TAAGAGAAAGGCCGGCACC






TCTTCTCTGACAGAGTGCGT






GCTGAATAAGGCCACAAAT






GTGGCCCACTGGACAACAC






CTAGCCTGAAGTGCATTAG






AGATCCTGCCCTGGTCCACC






AGAGGCCTGCCCCTCCATCT






ACAGTGACAACAGCCGGAG






TGACACCTCAGCCTGAATCT






CTGAGCCCTTCTGGAAAAG






AACCTGCCGCCAGCTCTCCT






AGCTCTAATAATACCGCCGC






CACAACAGCCGCCATTGTG






CCTGGATCTCAGCTGATGCC






TAGCAAGTCTCCTAGCACA






GGCACAACAGAGATCAGCA






GCCACGAATCTTCTCACGGA






ACACCTTCTCAGACCACCGC






CAAGAATTGGGAGCTGACA






GCCTCTGCCTCTCACCAGCC






TCCAGGAGTGTATCCTCAG






GGCCACTCTGATACAACAG






TGGCCATCAGCACATCTACA






GTGCTGCTGTGTGGACTGT






CTGCCGTGTCTCTGCTGGCC






TGTTACCTGAAGTCTAGACA






GACACCTCCTCTGGCCTCTG






TGGAGATGGAGGCCATGGA






AGCCCTGCCTGTGACATGG






GGAACAAGCAGCAGAGAT






GAGGACCTGGAGAATTGTT






CTCACCACCTG





mbIL15 + IgE
525
MDWTWILFLVAAATRVHSNWVN
526
ATGGATTGGACCTGGATTC


signal Peptide

VISDLKKIEDLIQSMHIDATLYTESD

TGTTTCTGGTGGCCGCTGCC




VHPSCKVTAMKCFLLELQVISLESG

ACAAGAGTGCACAGCAACT




DASIHDTVENLIILANNSLSSNGNVT

GGGTGAATGTGATCAGCGA




ESGCKECEELEEKNIKEFLQSFVHIV

CCTGAAGAAGATCGAGGAT




QMFINTSSGGGSGGGGSGGGGSG

CTGATCCAGAGCATGCACA




GGGSGGGSLQITCPPPMSVEHADI

TTGATGCCACCCTGTACACA




WVKSYSLYSRERYICNSGFKRKAGT

GAATCTGATGTGCACCCTA




SSLTECVLNKATNVAHWTTPSLKCI

GCTGTAAAGTGACCGCCAT




RDPALVHORPAPPSTVTTAGVTPQ

GAAGTGTTTTCTGCTGGAG




PESLSPSGKEPAASSPSSNNTAATT

CTGCAGGTGATTTCTCTGGA




AAIVPGSQLMPSKSPSTGTTEISSHE

AAGCGGAGATGCCTCTATC




SSHGTPSQTTAKNWELTASASHQP

CACGACACAGTGGAGAATC




PGVYPQGHSDTTVAISTSTVLLCGL

TGATCATCCTGGCCAACAAT




SAVSLLACYLKSRQTPPLASVEMEA

AGCCTGAGCAGCAATGGCA




MEALPVTWGTSSRDEDLENCSHHL

ATGTGACAGAGTCTGGCTG






TAAGGAGTGTGAGGAGCTG






GAGGAGAAGAACATCAAG






GAGTTTCTGCAGAGCTTTGT






GCACATCGTGCAGATGTTC






ATCAATACAAGCTCTGGCG






GAGGATCTGGAGGAGGCG






GATCTGGAGGAGGAGGCA






GTGGAGGCGGAGGATCTG






GCGGAGGATCTCTGCAGAT






TACATGCCCTCCTCCAATGT






CTGTGGAGCACGCCGATAT






TTGGGTGAAGTCCTACAGC






CTGTACAGCAGAGAGAGAT






ACATCTGCAACAGCGGCTTT






AAGAGAAAGGCCGGCACCT






CTTCTCTGACAGAGTGCGT






GCTGAATAAGGCCACAAAT






GTGGCCCACTGGACAACAC






CTAGCCTGAAGTGCATTAG






AGATCCTGCCCTGGTCCACC






AGAGGCCTGCCCCTCCATCT






ACAGTGACAACAGCCGGAG






TGACACCTCAGCCTGAATCT






CTGAGCCCTTCTGGAAAAG






AACCTGCCGCCAGCTCTCCT






AGCTCTAATAATACCGCCGC






CACAACAGCCGCCATTGTG






CCTGGATCTCAGCTGATGCC






TAGCAAGTCTCCTAGCACA






GGCACAACAGAGATCAGCA






GCCACGAATCTTCTCACGGA






ACACCTTCTCAGACCACCGC






CAAGAATTGGGAGCTGACA






GCCTCTGCCTCTCACCAGCC






TCCAGGAGTGTATCCTCAG






GGCCACTCTGATACAACAG






TGGCCATCAGCACATCTACA






GTGCTGCTGTGTGGACTGT






CTGCCGTGTCTCTGCTGGCC






TGTTACCTGAAGTCTAGACA






GACACCTCCTCTGGCCTCTG






TGGAGATGGAGGCCATGGA






AGCCCTGCCTGTGACATGG






GGAACAAGCAGCAGAGAT






GAGGACCTGGAGAATTGTT






CTCACCACCTG









Exemplary Linker Sequences

















SEQ

SEQ ID



Linker Name
ID NO
Amino Acids Sequence
NO
Polynucleotide Sequence







Whitlow Linker
527
GSTSGSGKPGSGEGSTKG
528
GGCAGCACCTCCGGCAGCGG






CAAGCCTGGCAGCGGCGAGG






GCAGCACCAAGGGC





Linker
529
SGGGSGGGGSGGGGSGGGGSGG
530
TCTGGCGGAGGATCTGGAGG




GSLQ

AGGCGGATCTGGAGGAGGAG






GCAGTGGAGGCGGAGGATCT






GGCGGAGGATCTCTGCAG





GSG linker
531
GSG
532
GGAAGCGGA





SGSG linker
533
SGSG
534
AGTGGCAGCGGC





(G4S)3 linker
535
GGGGSGGGGSGGGGS
536
GGTGGCGGTGGCTCGGGCGG






TGGTGGGTCGGGTGGCGGCG






GATCT





Furin cleavage
537
RAKR
538
CGTGCAAAGCGT


site/Furinlink1









Fmdv
539
RAKRAPVKQTLNFDLLKLAGDVESN
540
AGAGCCAAGAGGGCACCGGT




PGP

GAAACAGACTTTGAATTTTGA






CCTTCTGAAGTTGGCAGGAGA






CGTTGAGTCCAACCCTGGGCC






C





Thosea asigna
541
EGRGSLLTCGDVEENPGP
542
GAGGGCAGAGGAAGTCTGCT


virus 2A region



AACATGCGGTGACGTCGAGG


(T2A)



AGAATCCTGGACCT





Furin-GSG-T2A
543
RAKRGSGEGRGSLLTCGDVEENPG
544
AGAGCTAAGAGGGGAAGCGG




P

AGAGGGCAGAGGAAGTCTGC






TAACATGCGGTGACGTCGAGG






AGAATCCTGGACCT





Furin-SGSG-T2A
545
RAKRSGSGEGRGSLLTCGDVEENP
546
AGGGCCAAGAGGAGTGGCAG




GP

CGGCGAGGGCAGAGGAAGTC






TTCTAACATGCGGTGACGTGG






AGGAGAATCCCGGCCCT





Porcine
547
ATNFSLLKQAGDVEENPGP
548
GCAACGAACTTCTCTCTCCTAA


teschovirus-1 2A



AACAGGCTGGTGATGTGGAG


region (P2A)



GAGAATCCTGGTCCA





GSG-P2A
549
GSGATNFSLLKQAGDVEENPGP
550
GGAAGCGGAGCTACTAACTTC






AGCCTGCTGAAGCAGGCTGG






AGACGTGGAGGAGAACCCTG






GACCT





Equine rhinitis A
551
QCTNYALLKLAGDVESNPGP
552
CAGTGTACTAATTATGCTCTCT


virus 2A region



TGAAATTGGCTGGAGATGTTG


(E2A)



AGAGCAACCCTGGACCT





Foot-and-mouth
553
VKQTLNFDLLKLAGDVESNPGP
554
GTCAAACAGACCCTAAACTTT


disease virus 2A



GATCTGCTAAAACTGGCCGGG


region (F2A)



GATGTGGAAAGTAATCCCGGC






CCC





FP2A
555
RAKRAPVKQGSGATNFSLLKQAGD
556
CGTGCAAAGCGTGCACCGGTG




VEENPGP

AAACAGGGAAGCGGAGCTAC






TAACTTCAGCCTGCTGAAGCA






GGCTGGAGACGTGGAGGAGA






ACCCTGGACCT





Linker-GSG
557
APVKQGSG
558
GCACCGGTGAAACAGGGAAG






CGGA





Linker
559
GGGGSGGGSGGGGSGGGGS
560
GGTGGCGGTGGCTCGGGCGG






TGGTGGGTCGGGTGGCGGCG






GATCTGGTGGCGGTGGCTCG





Linker
561
APVKQ
562
GCACCGGTGAAACAG





Linker
563
A(EAAAK)nA (n = 2-5)
564









Exemplary Cell Tag Sequences

















SEQ ID

SEQ




NO
Amino Acid Sequence
ID NO
Polynucleotide Sequence







HER1 Domain
565
RKVCNGIGIGEFKDSLSINATNIKHF
566
CGCAAAGTGTGTAACGGAA


III

KNCTSISGDLHILPVAFRGDSFTHTP

TAGGTATTGGTGAATTTAA




PLDPQELD

AGACTCACTCTCCATAAATG




ILKTVKEITGFLLIQAWPENRTDLHA

CTACGAATATTAAACACTTC




FENLEIIRGRTKQHGQFSLAVVSLNI

AAAAACTGCACCTCCATCAG




TSLGLRSL

TGGCGATCTCCACATCCTGC




KEISDGDVIISGNKNLCYANTINWK

CGGTGGCATTTAGGGGTGA




KLFGTSGQKTKIISNRGENSCKATG

CTCCTTCACACATACTCCTC




Q

CTCTGGATCCACAGGAACT






GGATATTCTGAAAACCGTA






AAGGAAATCACAGGGTTTT






TGCTGATTCAGGCTTGGCCT






GAAAACAGGACGGACCTCC






ATGCCTTTGAGAACCTAGA






AATCATACGCGGCAGGACC






AAGCAACATGGTCAGTTTTC






TCTTGCAGTCGTCAGCCTGA






ACATAACATCCTTGGGATTA






CGCTCCCTCAAGGAGATAA






GTGATGGAGATGTGATAAT






TTCAGGAAACAAAAATTTGT






GCTATGCAAATACAATAAA






CTGGAAAAAACTGTTTGGG






ACCTCCGGTCAGAAAACCA






AAATTATAAGCAACAGAGG






TGAAAACAGCTGCAAGGCC






ACAGGCCAG





HER1
567
VCHALCSPEGCWGPEPRDCVS
568
GTCTGCCATGCCTTGTGCTC


truncated



CCCCGAGGGCTGCTGGGGC


Domain IV



CCGGAGCCCAGGGACTGCG






TCTCT





HER1t
569
RKVCNGIGIGEFKDSLSINATNIKHF
570
CGCAAAGTGTGTAACGGAA




KNCTSISGDLHILPVAFRGDSFTHTP

TAGGTATTGGTGAATTTAA




PLDPQELDILKTVKEITGFLLIQAWP

AGACTCACTCTCCATAAATG




ENRTDLHAFENLEIIRGRTKQHGQF

CTACGAATATTAAACACTTC




SLAVVSLNITSLGLRSLKEISDGDVIIS

AAAAACTGCACCTCCATCAG




GNKNLCYANTINWKKLFGTSGQKT

TGGCGATCTCCACATCCTGC




KIISNRGENSCKATGQVCHALCSPE

CGGTGGCATTTAGGGGTGA




GCWGPEPRDCVSCRNVSRGRECV

CTCCTTCACACATACTCCTC




DKCNLLEGEPREFVENSECIQCHPE

CTCTGGATCCACAGGAACT




CLPQAMNITCTGRGPDNCIQCAHY

GGATATTCTGAAAACCGTA




IDGPHCVKTCPAGVMGENNTLVW

AAGGAAATCACAGGGTTTT




KYADAGHVCHLCHPNCTYGCTGP

TGCTGATTCAGGCTTGGCCT




GLEGCPTNGPKIPSIATGMVGALLL

GAAAACAGGACGGACCTCC




LLVVALGIGLFM

ATGCCTTTGAGAACCTAGA






AATCATACGCGGCAGGACC






AAGCAACATGGTCAGTTTTC






TCTTGCAGTCGTCAGCCTGA






ACATAACATCCTTGGGATTA






CGCTCCCTCAAGGAGATAA






GTGATGGAGATGTGATAAT






TTCAGGAAACAAAAATTTGT






GCTATGCAAATACAATAAA






CTGGAAAAAACTGTTTGGG






ACCTCCGGTCAGAAAACCA






AAATTATAAGCAACAGAGG






TGAAAACAGCTGCAAGGCC






ACAGGCCAGGTCTGCCATG






CCTTGTGCTCCCCCGAGGG






CTGCTGGGGCCCGGAGCCC






AGGGACTGCGTCTCTTGCC






GGAATGTCAGCCGAGGCAG






GGAATGCGTGGACAAGTGC






AACCTTCTGGAGGGTGAGC






CAAGGGAGTTTGTGGAGAA






CTCTGAGTGCATACAGTGC






CACCCAGAGTGCCTGCCTCA






GGCCATGAACATCACCTGC






ACAGGACGGGGACCAGAC






AACTGTATCCAGTGTGCCCA






CTACATTGACGGCCCCCACT






GCGTCAAGACCTGCCCGGC






AGGAGTCATGGGAGAAAAC






AACACCCTGGTCTGGAAGT






ACGCAGACGCCGGCCATGT






GTGCCACCTGTGCCATCCAA






ACTGCACCTACGGATGCACT






GGGCCAGGTCTTGAAGGCT






GTCCAACGAATGGGCCTAA






GATCCCGTCCATCGCCACTG






GGATGGTGGGGGCCCTCCT






CTTGCTGCTGGTGGTGGCC






CTGGGGATCGGCCTCTTCAT






G





HER1t-1
571
RKVCNGIGIGEFKDSLSINATNIKHF
572
CGCAAAGTGTGTAACGGAA




KNCTSISGDLHILPVAFRGDSFTHTP

TAGGTATTGGTGAATTTAA




PLDPQELDILKTVKEITGFLLIQAWP

AGACTCACTCTCCATAAATG




ENRTDLHAFENLEIIRGRTKQHGQF

CTACGAATATTAAACACTTC




SLAVVSLNITSLGLRSLKEISDGDVIIS

AAAAACTGCACCTCCATCAG




GNKNLCYANTINWKKLFGTSGQKT

TGGCGATCTCCACATCCTGC




KIISNRGENSCKATGQVCHALCSPE

CGGTGGCATTTAGGGGTGA




GCWGPEPRDCVSGGGGSGGGSG

CTCCTTCACACATACTCCTC




GGGSGGGGSFWVLVVVGGVLACY

CTCTGGATCCACAGGAACT




SLLVTVAFIIFWVRSKRS

GGATATTCTGAAAACCGTA






AAGGAAATCACAGGGTTTT






TGCTGATTCAGGCTTGGCCT






GAAAACAGGACGGACCTCC






ATGCCTTTGAGAACCTAGA






AATCATACGCGGCAGGACC






AAGCAACATGGTCAGTTTTC






TCTTGCAGTCGTCAGCCTGA






ACATAACATCCTTGGGATTA






CGCTCCCTCAAGGAGATAA






GTGATGGAGATGTGATAAT






TTCAGGAAACAAAAATTTGT






GCTATGCAAATACAATAAA






CTGGAAAAAACTGTTTGGG






ACCTCCGGTCAGAAAACCA






AAATTATAAGCAACAGAGG






TGAAAACAGCTGCAAGGCC






ACAGGCCAGGTCTGCCATG






CCTTGTGCTCCCCCGAGGG






CTGCTGGGGCCCGGAGCCC






AGGGACTGCGTCTCTGGTG






GCGGTGGCTCGGGCGGTG






GTGGGTCGGGTGGCGGCG






GATCTGGTGGCGGTGGCTC






GTTTTGGGTGCTGGTGGTG






GTTGGTGGAGTCCTGGCTT






GCTATAGCTTGCTAGTAACA






GTGGCCTTTATTATTTTCTG






GGTGAGGAGTAAGAGGAG






CTAA





FL CD20
573
MTTPRNSVNGTFPAEPMKGPIAM
574
ATGACAACACCCAGAAATT




QSGPKPLFRRMSSLVGPTQSFFMR

CAGTAAATGGGACTTTCCC




ESKTLGAVQIMNGLFHIALGGLLMI

GGCAGAGCCAATGAAAGGC




PAGIYAPICVTVWYPLWGGIMYIIS

CCTATTGCTATGCAATCTGG




GSLLAATEKNSRKCLVKGKMIMNS

TCCAAAACCACTCTTCAGGA




LSLFAAISGMILSIMDILNIKISHFLK

GGATGTCTTCACTGGTGGG




MESLNFIRAHTPYINIYNCEPANPSE

CCCCACGCAAAGCTTCTTCA




KNSPSTQYCYSIQSLFLGILSVMLIFA

TGAGGGAATCTAAGACTTT




FFQELVIAGIVENEWKRTCSRPKSNI

GGGGGCTGTCCAGATTATG




VLLSAEEKKEQTIEIKEEVVGLTETSS

AATGGGCTCTTCCACATTGC




QPKNEEDIEIIPIQEEEEEETETNFPE

CCTGGGGGGTCTTCTGATG




PPQDQESSPIENDSSP

ATCCCAGCAGGGATCTATG






CACCCATCTGTGTGACTGTG






TGGTACCCTCTCTGGGGAG






GCATTATGTATATTATTTCC






GGATCACTCCTGGCAGCAA






CGGAGAAAAACTCCAGGAA






GTGTTTGGTCAAAGGAAAA






ATGATAATGAATTCATTGAG






CCTCTTTGCTGCCATTTCTG






GAATGATTCTTTCAATCATG






GACATACTTAATATTAAAAT






TTCCCATTTTTTAAAAATGG






AGAGTCTGAATTTTATTAGA






GCTCACACACCATATATTAA






CATATACAACTGTGAACCA






GCTAATCCCTCTGAGAAAA






ACTCCCCATCTACCCAATAC






TGTTACAGCATACAATCTCT






GTTCTTGGGCATTTTGTCAG






TGATGCTGATCTTTGCCTTC






TTCCAGGAACTTGTAATAGC






TGGCATCGTTGAGAATGAA






TGGAAAAGAACGTGCTCCA






GACCCAAATCTAACATAGTT






CTCCTGTCAGCAGAAGAAA






AAAAAGAACAGACTATTGA






AATAAAAGAAGAAGTGGTT






GGGCTAACTGAAACATCTTC






CCAACCAAAGAATGAAGAA






GACATTGAAATTATTCCAAT






CCAAGAAGAGGAAGAAGA






AGAAACAGAGACGAACTTT






CCAGAACCTCCCCAAGATCA






GGAATCCTCACCAATAGAA






AATGACAGCTCTCCT





CD20t-1
575
MTTPRNSVNGTFPAEPMKGPIAM
576
ATGACCACACCACGGAACT




QSGPKPLFRRMSSLVGPTQSFFMR

CTGTGAATGGCACCTTCCCA




ESKTLGAVQIMNGLFHIALGGLLMI

GCAGAGCCAATGAAGGGAC




PAGIYAPICVTVWYPLWGGIMYIIS

CAATCGCAATGCAGAGCGG




GSLLAATEKNSRKCLVKGKMIMNS

ACCCAAGCCTCTGTTTCGGA




LSLFAAISGMILSIMDILNIKISHFLK

GAATGAGCTCCCTGGTGGG




MESLNFIRAHTPYINIYNCEPANPSE

CCCAACCCAGTCCTTCTTTA




KNSPSTQYCYSIQSLFLGILSVMLIFA

TGAGAGAGTCTAAGACACT




FFQELVIAGIVENEWKRTCSRPKSNI

GGGCGCCGTGCAGATCATG




VLLSAEEKKEQTIEIKEEVVGLTETSS

AACGGACTGTTCCACATCGC




QPKNEEDIE

CCTGGGAGGACTGCTGATG






ATCCCAGCCGGCATCTACGC






CCCTATCTGCGTGACCGTGT






GGTACCCTCTGTGGGGCGG






CATCATGTATATCATCTCCG






GCTCTCTGCTGGCCGCCACA






GAGAAGAACAGCAGGAAG






TGTCTGGTGAAGGGCAAGA






TGATCATGAATAGCCTGTCC






CTGTTTGCCGCCATCTCTGG






CATGATCCTGAGCATCATG






GACATCCTGAACATCAAGA






TCAGCCACTTCCTGAAGATG






GAGAGCCTGAACTTCATCA






GAGCCCACACCCCTTACATC






AACATCTATAATTGCGAGCC






TGCCAACCCATCCGAGAAG






AATTCTCCAAGCACACAGTA






CTGTTATTCCATCCAGTCTC






TGTTCCTGGGCATCCTGTCT






GTGATGCTGATCTTTGCCTT






CTTTCAGGAGCTGGTCATC






GCCGGCATCGTGGAGAACG






AGTGGAAGAGGACCTGCAG






CCGCCCCAAGTCCAATATCG






TGCTGCTGTCCGCCGAGGA






GAAGAAGGAGCAGACAATC






GAGATCAAGGAGGAGGTG






GTGGGCCTGACCGAGACAT






CTAGCCAGCCTAAGAATGA






GGAGGATATCGAG









Exemplary Vector Sequences


















Human EF1A1


577
GCCGCAATAAAATATCTTTA


Promoter



TTTTCATTACATCTGTGTGTT






GGTTTTTTGTGTGAATCGTA






ACTAACATACGCTCTCCATC






AAAACAAAACGAAACAAAA






CAAACTAGCAAAATAGGCT






GTCCCCAGTGCAAGTGCAG






GTGCCAGAACATTTCTCTAT






CGAAGGATCTGCGATCGCT






CCGGTGCCCGTCAGTGGGC






AGAGCGCACATCGCCCACA






GTCCCCGAGAAGTTGGGGG






GAGGGGTCGGCAATTGAAC






CGGTGCCTAGAGAAGGTGG






CGCGGGGTAAACTGGGAAA






GTGATGTCGTGTACTGGCTC






CGCCTTTTTCCCGAGGGTGG






GGGAGAACCGTATATAAGT






GCAGTAGTCGCCGTGAACG






TTCTTTTTCGCAACGGGTTT






GCCGCCAGAACACAG





Human CMV


578
GTGATGCGGTTTTGGCAGT


immediate



ACATCAATGGGCGTGGATA


Early



GCGGTTTGACTCACGGGGA


Promoter



TTTCCAAGTCTCCACCCCATT






GACGTCAATGGGAGTTTGT






TTTGGCACCAAAATCAACGG






GACTTTCCAAAATGTCGTAA






CAACTCCGCCCCATTGACGC






AAATGGGCGGTAGGCGTGT






ACGGTGGGAGGTCTATATA






AGCAGAGCTC





ColE1 ORI


579
TGTGAGCAAAAGGCCAGCA






AAAGGCCAGGAACCGTAAA






AAGGCCGCGTTGCTGGCGT






TTTTCCATAGGCTCCGCCCC






CCTGACGAGCATCACAAAA






ATCGACGCTCAAGTCAGAG






GTGGCGAAACCCGACAGGA






CTATAAAGATACCAGGCGTT






TCCCCCTGGAAGCTCCCTCG






TGCGCTCTCCTGTTCCGACC






CTGCCGCTTACCGGATACCT






GTCCGCCTTTCTCCCTTCGG






GAAGCGTGGCGCTTTCTCAT






AGCTCACGCTGTAGGTATCT






CAGTTCGGTGTAGGTCGTTC






GCTCCAAGCTGGGCTGTGT






GCACGAACCCCCCGTTCAGC






CCGACCGCTGCGCCTTATCC






GGTAACTATCGTCTTGAGTC






CAACCCGGTAAGACACGAC






TTATCGCCACTGGCAGCAGC






CACTGGTAACAGGATTAGC






AGAGCGAGGTATGTAGGCG






GTGCTACAGAGTTCTTGAA






GTGGTGGCCTAACTACGGC






TACACTAGAAGAACAGTATT






TGGTATCTGCGCTCTGCTGA






AGCCAGTTACCTTCGGAAAA






AGAGTTGGTAGCTCTTGATC






CGG CAAACAAACCACCG CT






GGTAGCGGTGGTTTTTTTGT






TTGCAAGCAGCAGATTACG






CGCAGAAAAAAAGGATCTC






AAGAAGATCCTTTGATCTTT






TCTACGGGG





Left


580
CTACAGTTGA


Transposon



AGTCGGAAGT


Repeat



TTACATACAC TTAAGTTGGA


Region



GTCATTAAAA CTCGTTTTTC






AACTACTCCA CAAATTTCTT






GTTAACAAAC






AATAGTTTTG






GCAAGTCAGT






TAGGACATCT ACTTTGTGCA






TGACACAAGT CATTTTTCCA






ACAATTGTTT






ACAGACAGAT TATTTCACTT






ATAATTCACT GTATCACAAT






TCCAGTGGGT






CAGAAGTTTA






CATACACTAA






GTTGACTGTG CCTTTAAACA






GCTTGGAAAA






TTCCAGAAAA






TGATGTCATG GCTTTAG





Right


581
GTGGAAGGCT


Transposon



ACTCGAAATG


Repeat



TTTGACCCAA GTTAAACAAT


Region



TTAAAGGCAA






TGCTACCAAA TACTAATTGA






GTGTATGTTA ACTTCTGACC






CACTGGGAAT






GTGATGAAAG






AAATAAAAGC






TGAAATGAAT CATTCTCTCT






ACTATTATTC TGATATTTCA






CATTCTTAAA






ATAAAGTGGT






GATCCTAACT






GACCTTAAGA






CAGGGAATCT






TTACTCGGAT






TAAATGTCAG






GAATTGTGAA






AAAGTGAGTT






TAAATGTATT






TGGCTAAGGT






GTATGTAAAC TTCCGACTTC






AACTGTAGG









Exemplary control sequences















miRNA
SEQ
miRNA



Target
ID NO
backbone
DNA sequence of Pri-miRNA







Scrambled
582
miR206,
GATGCTACAAGTGGCCCACTTCTGAGATGCGGGCTGCTTCT


control 2

shorter
GGATGACACTGCTTCCCGAGGCATTTCGCCCTATCTGCAAGT




arms
ACTATGGATTACTTTGCTAGTGGTGTAGATAGGGTGAAATG





TTTCGGCAAGTGCCTCCTCGCTGGCCCCAGGGTACCACCCGG





AGCACAGGTTTGGTGACCTT





neg. ctl
583
miR150
AGGGACTGGGCCCACGGGGAGGCAGCGTCCCCGAGGCAGC



c.elegans



AGCGGCAGCGGCGGCTCCTCTCCCCATGGCCCTGTCACAACC


cel-miR-67


TCCTAGAAAGAGTACTGGGCTCAGACCCTCTTTCAGGCCGTT





GTGACAGGGACCTGGGGACCCCGGCACCGGCAGGCCCCAA





GGGGTGAGGTGAGCGGGCATTGGGACCTCCCCTCCCTGTAC





TC





neg. ctl
584
miR204
AGGAGGGTGGGGGTGGAGGCAAGCAGAGGACTTCCTGATC



c.elegans



GCGTACCCATGGCTACAGTCTTTCTTCATGTGACTCGTGGAC


cel-miR-67


TTGTACTACACAAAAGTACTGTGAGAATATATGAAGGACAG





GCTTTAGTGTAGTATGCGTTCAATTGTCATCACTGGCATCTTT





TTTGATCATTGCACCATCATCAAATGCATTGGGATAACCATG





AC





210 bp
585

GGTAAGTCATGACTCCCTACAATGGACATGATCATAGTAGGT


stuffer


ACTATAAGGCACCTAGCTATACCCTCCTATAGAGAGTTTGAG





TCTATTGTAGCAAGTCTTTCTTTTAGGGCCTAGGACTCTTCCC





ACTCTCTTCCATCGCCTAAGCTCTAACTCTTCCTTGTAGTAGA





AATAAGTCACTTCTAAGGCTGGGACCCTCCTAGACCCTAA





Scrambled
586
miR206
TTAGGATGAGTTGAGATCCCAGTGATCTTCTCGCTAAGAGTT


control 1


TCCTGCCTGGGCAAGGAGGAAAGATGCTACAAGTGGCCCAC





TTCTGAGATGCGGGCTGCTTCTGGATGACACTGCTTCCCGAG





GCATTTCGCCCTATCTGCAAGTACTATGGATTACTTTGCTAGT





GGTGTAGATAGGGTGAAATGTTTCGGCAAGTGCCTCCTCGC





TGGCCCCAGGGTACCACCCGGAGCACAGGTTTGGTGACCTT





CTTCCTCATCAGGGCTTTGTGCCAGCAAATGACTCCCTCACC





AAGGAAGCAAGAGCCTCTGAATCCCATCTGGGCTCTTCCTGA





ACACCCCTATCTCCCCCTCT
















TABLE 14







Additional Sequences












SEQ ID

SEQ



Name
NO
Amino Acid Sequence
ID NO
Nucleotide Sequence










CAR Sequences











Human ROR1
587
PLLALLAALLLAARGAAAQETELSVS
588
ATGCACCGGCCGCGCCGCC




AELVPTSSWNISSELNKDSYLTLDEP

GCGGGACGCGCCCGCCGCT




MNNITTSLGQTAELHCKVSGNPPP

CCTGGCGCTGCTGGCCGCG




TIRWFKNDAPVVQEPRRLSFRSTIY

CTGCTGCTGGCCGCACGCG




GSRLRIRNLDTTDTGYFQCVATNG

GGGCTGCTGCCCAAGAAAC




KEVVSSTGVLFVKFGPPPTASPGYS

AGAGCTGTCAGTCAGTGCT




DEYEEDGFCQPYRGIACARFIGNRT

GAATTAGTGCCTACCTCATC




VYMESLHMQGEIENQITAAFTMIG

ATGGAACATCTCAAGTGAA




TSSHLSDKCSQFAIPSLCHYAFPYCD

CTCAACAAAGATTCTTACCT




ETSSVPKPRDLCRDECEILENVLCQT

GACCCTCGATGAACCAATG




EYIFARSNPMILMRLKLPNCEDLPQ

AATAACATCACCACGTCTCT




PESPEAANCIRIGIPMADPINKNHK

GGGCCAGACAGCAGAACTG




CYNSTGVDYRGTVSVTKSGRQCQP

CACTGCAAAGTCTCTGGGA




WNSQYPHTHTFTALRFPELNGGHS

ATCCACCTCCCACCATCCGC




YCRNPGNQKEAPWCFTLDENFKS

TGGTTCAAAAATGATGCTCC




DLCDIPACDSKDSKEKNKMEILYILV

TGTGGTCCAGGAGCCCCGG




PSVAIPLAIALLFFFICVCRNNQKSSS

AGGCTCTCCTTTCGGTCCAC




APVQRQPKHVRGQNVEMSMLNA

CATCTATGGCTCTCGGCTGC




YKPKSKAKELPLSAVRFMEELGECA

GGATTAGAAACCTCGACAC




FGKIYKGHLYLPGMDHAQLVAIKTL

CACAGACACAGGCTACTTCC




KDYNNPQQWTEFQQEASLMAEL

AGTGCGTGGCAACAAACGG




HHPNIVCLLGAVTQEQPVCMLFEYI

CAAGGAGGTGGTTTCTTCC




NQGDLHEFLIMRSPHSDVGCSSDE

ACTGGAGTCTTGTTTGTCAA




DGTVKSSLDHGDFLHIAIQIAAGME

GTTTGGCCCCCCTCCCACTG




YLSSHFFVHKDLAARNILIGEQLHVK

CAAGTCCAGGATACTCAGA




ISDLGLSREIYSADYYRVQSKSLLPIR

TGAGTATGAAGAAGATGGA




WMPPEAIMYGKFSSDSDIWSFGV

TTCTGTCAGCCATACAGAG




VLWEIFSFGLQPYYGFSNQEVIEMV

GGATTGCATGTGCAAGATT




RKRQLLPCSEDCPPRMYSLMTECW

TATTGGCAACCGCACCGTCT




NEIPSRRPRFKDIHVRLRSWEGLSS

ATATGGAGTCTTTGCACATG




HTSSTTPSGGNATTQTTSLSASPVS

CAAGGGGAAATAGAAAATC




NLSNPRYPNYMFPSQGITPQGQIA

AGATCACAGCTGCCTTCACT




GFIGPPIPQNQRFIPINGYPIPPGYA

ATGATTGGCACTTCCAGTCA




AFPAAHYQPTGPPRVIQHCPPPKS

CTTATCTGATAAGTGTTCTC




RSPSSASGSTSTGHVTSLPSSGSNQ

AGTTCGCCATTCCTTCCCTG




EANIPLLPHMSIPNHPGGMGITVF

TGCCACTATGCCTTCCCGTA




GNKSQKPYKIDSKQASLLGDANIHG

CTGCGATGAAACTTCATCCG




HTESMISAEL

TCCCAAAGCCCCGTGACTTG






TGTCGCGATGAATGTGAAA






TCCTGGAGAATGTCCTGTGT






CAAACAGAGTACATTTTTGC






AAGATCAAATCCCATGATTC






TGATGAGGCTGAAACTGCC






AAACTGTGAAGATCTCCCCC






AGCCAGAGAGCCCAGAAGC






TGCGAACTGTATCCGGATT






GGAATTCCCATGGCAGATC






CTATAAATAAAAATCACAA






GTGTTATAACAGCACAGGT






GTGGACTACCGGGGGACCG






TCAGTGTGACCAAATCAGG






GCGCCAGTGCCAGCCATGG






AATTCCCAGTATCCCCACAC






ACACACTTTCACCGCCCTTC






GTTTCCCAGAGCTGAATGG






AGGCCATTCCTACTGCCGCA






ACCCAGGGAATCAAAAGGA






AGCTCCCTGGTGCTTCACCT






TGGATGAAAACTTTAAGTCT






GATCTGTGTGACATCCCAG






CGTGCGATTCAAAGGATTC






CAAGGAGAAGAATAAAATG






GAAATCCTGTACATACTAGT






GCCAAGTGTGGCCATTCCC






CTGGCCATTGCTTTACTCTT






CTTCTTCATTTGCGTCTGTC






GGAATAACCAGAAGTCATC






GTCGGCACCAGTCCAGAGG






CAACCAAAACACGTCAGAG






GTCAAAATGTAGAGATGTC






AATGCTGAATGCATATAAA






CCCAAGAGCAAGGCTAAAG






AGCTACCTCTTTCTGCTGTA






CGCTTTATGGAAGAATTGG






GTGAGTGTGCCTTTGGAAA






AATCTATAAAGGCCATCTCT






ATCTCCCAGGCATGGACCAT






GCTCAGCTGGTTGCTATCAA






GACCTTGAAAGACTATAAC






AACCCCCAGCAATGGACGG






AATTTCAACAAGAAGCCTCC






CTAATGGCAGAACTGCACC






ACCCCAATATTGTCTGCCTT






CTAGGTGCCGTCACTCAGG






AACAACCTGTGTGCATGCTT






TTTGAGTATATTAATCAGGG






GGATCTCCATGAGTTCCTCA






TCATGAGATCCCCACACTCT






GATGTTGGCTGCAGCAGTG






ATGAAGATGGGACTGTGAA






ATCCAGCCTGGACCACGGA






GATTTTCTGCACATTGCAAT






TCAGATTGCAGCTGGCATG






GAATACCTGTCTAGTCACTT






CTTTGTCCACAAGGACCTTG






CAGCTCGCAATATTTTAATC






GGAGAGCAACTTCATGTAA






AGATTTCAGACTTGGGGCT






TTCCAGAGAAATTTACTCCG






CTGATTACTACAGGGTCCA






GAGTAAGTCCTTGCTGCCC






ATTCGCTGGATGCCCCCTGA






AGCCATCATGTATGGCAAA






TTCTCTTCTGATTCAGATAT






CTGGTCCTTTGGGGTTGTCT






TGTGGGAGATTTTCAGTTTT






GGACTCCAGCCATATTATG






GATTCAGTAACCAGGAAGT






GATTGAGATGGTGAGAAAA






CGGCAGCTCTTACCATGCTC






TGAAGACTGCCCACCCAGA






ATGTACAGCCTCATGACAG






AGTGCTGGAATGAGATTCC






TTCTAGGAGACCAAGATTTA






AAGATATTCACGTCCGGCTT






CGGTCCTGGGAGGGACTCT






CAAGTCACACAAGCTCTACT






ACTCCTTCAGGGGGAAATG






CCACCACACAGACAACCTCC






CTCAGTGCCAGCCCAGTGA






GTAATCTCAGTAACCCCAGA






TATCCTAATTACATGTTCCC






GAGCCAGGGTATTACACCA






CAGGGCCAGATTGCTGGTT






TCATTGGCCCGCCAATACCT






CAGAACCAGCGATTCATTCC






CATCAATGGATACCCAATAC






CTCCTGGATATGCAGCGTTT






CCAGCTGCCCACTACCAGCC






AACAGGTCCTCCCAGAGTG






ATTCAGCACTGCCCACCTCC






CAAGAGTCGGTCCCCAAGC






AGTGCCAGTGGGTCGACTA






GCACTGGCCATGTGACTAG






CTTGCCCTCATCAGGATCCA






ATCAGGAAGCAAATATTCCT






TTACTACCACACATGTCAAT






TCCAAATCATCCTGGTGGAA






TGGGTATCACCGTTTTTGGC






AACAAATCTCAAAAACCCTA






CAAAATTGACTCAAAGCAA






GCATCTTTACTAGGAGACG






CCAATATTCATGGACACACC






GAATCTATGATTTCTGCAGA






ACTG





Human ROR1
589
MHRPRRRGTRPPLLALLAALLLAAR
590
ATGCACCGGCCGCGCCGCC


(1-437)

GAAAQETELSVSAELVPTSSWNISS

GCGGGACGCGCCCGCCGCT




ELNKDSYLTLDEPMNNITTSLGQTA

CCTGGCGCTGCTGGCCGCG




ELHCKVSGNPPPTIRWFKNDAPVV

CTGCTGCTGGCCGCACGCG




QEPRRLSFRSTIYGSRLRIRNLDTTD

GGGCTGCTGCCCAAGAAAC




TGYFQCVATNGKEVVSSTGVLFVK

AGAGCTGTCAGTCAGTGCT




FGPPPTASPGYSDEYEEDGFCQPYR

GAATTAGTGCCTACCTCATC




GIACARFIGNRTVYMESLHMQGEI

ATGGAACATCTCAAGTGAA




ENQITAAFTMIGTSSHLSDKCSQFA

CTCAACAAAGATTCTTACCT




IPSLCHYAFPYCDETSSVPKPRDLCR

GACCCTCGATGAACCAATG




DECEILENVLCQTEYIFARSNPMIL

AATAACATCACCACGTCTCT




MRLKLPNCEDLPQPESPEAANCIRI

GGGCCAGACAGCAGAACTG




GIPMADPINKNHKCYNSTGVDYRG

CACTGCAAAGTCTCTGGGA




TVSVTKSGRQCQPWNSQYPHTHT

ATCCACCTCCCACCATCCGC




FTALRFPELNGGHSYCRNPGNQKE

TGGTTCAAAAATGATGCTCC




APWCFTLDENFKSDLCDIPACDSKD

TGTGGTCCAGGAGCCCCGG




SKEKNKMEILYILVPSVAIPLAIALLF

AGGCTCTCCTTTCGGTCCAC




FFICVCRNNQKSSSA

CATCTATGGCTCTCGGCTGC






GGATTAGAAACCTCGACAC






CACAGACACAGGCTACTTCC






AGTGCGTGGCAACAAACGG






CAAGGAGGTGGTTTCTTCC






ACTGGAGTCTTGTTTGTCAA






GTTTGGCCCCCCTCCCACTG






CAAGTCCAGGATACTCAGA






TGAGTATGAAGAAGATGGA






TTCTGTCAGCCATACAGAG






GGATTGCATGTGCAAGATT






TATTGGCAACCGCACCGTCT






ATATGGAGTCTTTGCACATG






CAAGGGGAAATAGAAAATC






AGATCACAGCTGCCTTCACT






ATGATTGGCACTTCCAGTCA






CTTATCTGATAAGTGTTCTC






AGTTCGCCATTCCTTCCCTG






TGCCACTATGCCTTCCCGTA






CTGCGATGAAACTTCATCCG






TCCCAAAGCCCCGTGACTTG






TGTCGCGATGAATGTGAAA






TCCTGGAGAATGTCCTGTGT






CAAACAGAGTACATTTTTGC






AAGATCAAATCCCATGATTC






TGATGAGGCTGAAACTGCC






AAACTGTGAAGATCTCCCCC






AGCCAGAGAGCCCAGAAGC






TGCGAACTGTATCCGGATT






GGAATTCCCATGGCAGATC






CTATAAATAAAAATCACAA






GTGTTATAACAGCACAGGT






GTGGACTACCGGGGGACCG






TCAGTGTGACCAAATCAGG






GCGCCAGTGCCAGCCATGG






AATTCCCAGTATCCCCACAC






ACACACTTTCACCGCCCTTC






GTTTCCCAGAGCTGAATGG






AGGCCATTCCTACTGCCGCA






ACCCAGGGAATCAAAAGGA






AGCTCCCTGGTGCTTCACCT






TGGATGAAAACTTTAAGTCT






GATCTGTGTGACATCCCAG






CGTGCGATTCAAAGGATTC






CAAGGAGAAGAATAAAATG






GAAATCCTGTACATACTAGT






GCCAAGTGTGGCCATTCCC






CTGGCCATTGCTTTACTCTT






CTTCTTCATTTGCGTCTGTC






GGAATAACCAGAAGTCATC






GTCGGCA





Murine ROR1
591
DIKMTQSPSSMYASLGERVTITCKA
592
GACATCAAGATGACCCAGA


(VL-VH).

SPDINSYLSWFQQKPGKSPKTLIYR

GCCCCAGCTCTATGTACGCC


IgG4 Fc-

ANRLVDGVPSRFSGGGSGQDYSLT

AGCCTGGGCGAGCGCGTGA


CD28m-Z

INSLEYEDMGIYYCLQYDEFPYTFG

CCATCACATGCAAGGCCAG




GGTKLEMKGSTSGSGKPGSGEGST

CCCCGACATCAACAGCTACC




KGEVKLVESGGGLVKPGGSLKLSCA

TGTCCTGGTTCCAGCAGAA




ASGFTFSSYAMSWVRQIPEKRLEW

GCCCGGCAAGAGCCCCAAG




VASISRGGTTYYPDSVKGRFTISRD

ACCCTGATCTACCGGGCCA




NVRNILYLQMSSLRSEDTAMYYCG

ACCGGCTGGTGGACGGCGT




RYDYDGYYAMDYWGQGTSVTVSS

GCCAAGCAGATTTTCCGGC




ESKYGPPCPPCPAPEFEGGPSVFLF

GGAGGCAGCGGCCAGGAC




PPKPKDTLMISRTPEVTCVVVDVSQ

TACAGCCTGACCATCAACA




EDPEVQFNWYVDGVEVHNAKTKP

GCCTGGAATACGAGGACAT




REEQFQSTYRVVSVLTVLHQDWLN

GGGCATCTACTACTGCCTGC




GKEYKCKVSNKGLPSSIEKTISKAKG

AGTACGACGAGTTCCCCTAC




QPREPQVYTLPPSQEEMTKNQVSL

ACCTTCGGAGGCGGCACCA




TCLVKGFYPSDIAVEWESNGQPEN

AGCTGGAAATGAAGGGCA




NYKTTPPVLDSDGSFFLYSRLTVDKS

GCACCTCCGGCAGCGGCAA




QKSLSLSLGKMFWVLVVVGGVLAC

GCCTGGCAGCGGCGAGGG




YSLLVTVAFIIFWVRSKRSRGGHSD

CAGCACCAAGGGCGAAGTG




YMNMTPRRPGPTRKHYQPYAPPR

AAGCTGGTGGAAAGCGGC




DFAAYRSRVKFSRSADAPAYQQGQ

GGAGGCCTGGTGAAACCTG




NCILYNELNLGRREEYDVLDKRRGR

CTGCGCCGCCAGCGGCTTC




DPEMGGKPRRKNPQEGLYNELQK

ACCTTCAGCAGCTACGCCAT




DKMAEAYSEIGMKGERRRGKGHD

GAGCTGGGTCCGACAGATC




GLYQGLSTATKDTYDALHMQALPP

CCCGAGAAGCGGCTGGAAT




R

GGGTGGCCAGCATCAGCAG






GGGCGGCACCACCTACTAC






CCCGACAGCGTGAAGGGCC






GGTTCACCATCAGCCGGGA






CAACGTGCGGAACATCCTG






TACCTGCAGATGAGCAGCC






TGCGGAGCGAGGACACCGC






CATGTACTACTGCGGCAGA






TACGACTACGACGGCTACT






ACGCCATGGATTACTGGGG






CCAGGGCACCAGCGTGACC






GTGTCTAGCGAGAGCAAGT






ACGGCCCTCCCTGCCCCCCT






TGCCCTGCCCCCGAGTTCGA






GGGCGGACCCAGCGTGTTC






CTGTTCCCCCCCAAGCCCAA






GGACACCCTGATGATCAGC






CGGACCCCCGAGGTGACCT






GTGTGGTGGTGGACGTGTC






CCAGGAGGACCCCGAGGTC






CAGTTCAACTGGTACGTGG






ACGGCGTGGAGGTGCACAA






CGCCAAGACCAAGCCCCGG






GAGGAGCAGTTCCAGAGCA






CCTACCGGGTGGTGTCCGT






GCTGACCGTGCTGCACCAG






GACTGGCTGAACGGCAAGG






AATACAAGTGTAAGGTGTC






CAACAAGGGCCTGCCCAGC






AGCATCGAGAAAACCATCA






GCAAGGCCAAGGGCCAGCC






TCGGGAGCCCCAGGTGTAC






ACCCTGCCCCCTAGCCAAGA






GGAGATGACCAAGAATCAG






GTGTCCCTGACCTGCCTGGT






GAAGGGCTTCTACCCCAGC






GACATCGCCGTGGAGTGGG






AGAGCAACGGCCAGCCCGA






GAACAACTACAAGACCACC






CCCCCTGTGCTGGACAGCG






ACGGCAGCTTCTTCCTGTAC






AGCAGGCTGACCGTGGACA






AGAGCCGGTGGCAGGAGG






GCAACGTCTTTAGCTGCTCC






GTGATGCACGAGGCCCTGC






ACAACCACTACACCCAGAA






GAGCCTGTCCCTGAGCCTG






GGCAAGATGTTCTGGGTGC






TGGTCGTGGTGGGTGGCGT






GCTGGCCTGCTACAGCCTG






CTGGTGACAGTGGCCTTCA






TCATCTTTTGGGTGAGGAG






CAAGCGGAGCAGAGGCGG






CCACAGCGACTACATGAAC






ATGACCCCCCGGAGGCCTG






GCCCCACCCGGAAGCACTA






CCAGCCCTACGCCCCTCCCA






GGGACTTCGCCGCCTACCG






GAGCCGGGTGAAGTTCAGC






CGGAGCGCCGACGCCCCTG






CCTACCAGCAGGGCCAGAA






CCAGCTGTACAACGAGCTG






AACCTGGGCCGGAGGGAG






GAGTACGACGTGCTGGACA






AGCGGAGAGGCCGGGACC






CTGAGATGGGCGGCAAGCC






CCGGAGAAAGAACCCTCAG






GAGGGCCTGTATAACGAAC






TGCAGAAAGACAAGATGGC






CGAGGCCTACAGCGAGATC






GGCATGAAGGGCGAGCGG






CGGAGGGGCAAGGGCCAC






GACGGCCTGTACCAGGGCC






TGAGCACCGCCACCAAGGA






TACCTACGACGCCCTGCACA






TGCAGGCCCTGCCCCCCAG






A





Murine ROR1
593
DIKMTQSPSSMYASLGERVTITCKA
594
GACATCAAGATGACCCAGA


(VL-VH).

SPDINSYLSWFQQKPGKSPKTLIYR

GCCCCAGCTCTATGTACGCC


IgG4 Fcm-

ANRLVDGVPSRFSGGGSGQDYSLT

AGCCTGGGCGAGCGCGTGA


CD28m-Z

INSLEYEDMGIYYCLQYDEFPYTFG

CCATCACATGCAAGGCCAG




GGTKLEMKGSTSGSGKPGSGEGST

CCCCGACATCAACAGCTACC




KGEVKLVESGGGLVKPGGSLKLSCA

TGTCCTGGTTCCAGCAGAA




ASGFTFSSYAMSWVRQIPEKRLEW

GCCCGGCAAGAGCCCCAAG




VASISRGGTTYYPDSVKGRFTISRD

ACCCTGATCTACCGGGCCA




NVRNILYLQMSSLRSEDTAMYYCG

ACCGGCTGGTGGACGGCGT




RYDYDGYYAMDYWGQGTSVTVSS

GCCAAGCAGATTTTCCGGC




QGTSVTVSSESKYGPPCPPCPAPEF

GGAGGCAGCGGCCAGGAC




LGGPSVFLFPPKPKDTLMISRTPEVT

TACAGCCTGACCATCAACA




CVVVDVSQEDPEVQFNWYVDGVE

GCCTGGAATACGAGGACAT




VHNAKTKPREEQFNSTYRVVSVLT

GGGCATCTACTACTGCCTGC




VLHQDWLNGKEYKCKVSNKGLPSS

AGTACGACGAGTTCCCCTAC




IEKTISKAKGQPREPQVYTLPPSQEE

ACCTTCGGAGGCGGCACCA




MTKNQVSLTCLVKGFYPSDIAVEW

AGCTGGAAATGAAGGGCA




ESNGQPENNYKTTPPVLDSDGSFFL

GCACCAGCGGCAGCGGCAA




YSRLTVDKSRWQEGNVFSCSVMH

GCCTGGAAGCGGCGAGGG




EALHNHYTQKSLSLSLGKMFWVLV

CTCCACCAAGGGCGAAGTG




VVGGVLACYSLLVTVAFIIFWVRSK

AAGCTGGTGGAAAGCGGC




RSRGGHSDYMNMTPRRPGPTRKH

GGAGGCCTGGTGAAACCTG




YQPYAPPRDFAAYRSRVKFSRSAD

GCGGCAGCCTGAAGCTGAG




APAYQQGQNQLYNELNLGRREEY

CTGCGCCGCCAGCGGCTTC




DVLDKRRGRDPEMGGKPRRKNPQ

ACCTTCAGCAGCTACGCCAT




EGLYNELQKDKMAEAYSEIGMKGE

GAGCTGGGTCCGACAGATC




RRRGKGHDGLYQGLSTATKDTYDA

CCCGAGAAGCGGCTGGAAT




LHMQALPPR

GGGTGGCCAGCATCAGCAG






GGGCGGCACCACCTACTAC






CCCGACAGCGTGAAGGGCC






GGTTCACCATCAGCCGGGA






CAACGTGCGGAACATCCTG






TACCTGCAGATGAGCAGCC






TGCGGAGCGAGGACACCGC






CATGTACTACTGCGGCAGA






TACGACTACGACGGCTACT






ACGCCATGGATTACTGGGG






CCAGGGCACCAGCGTGACC






GTGTCTAGCCAGGGAACCT






CCGTGACAGTGTCCAGCGA






GTCCAAATATGGTCCCCCAT






GCCCACCATGCCCAGCACCT






GAGTTCCTGGGGGGACCAT






CAGTCTTCCTGTTCCCCCCA






AAACCCAAGGACACTCTCAT






GATCTCCCGGACCCCTGAG






GTCACGTGCGTGGTGGTGG






ACGTGAGCCAGGAAGACCC






CGAGGTCCAGTTCAACTGG






TACGTGGATGGCGTGGAGG






TGCATAATGCCAAGACAAA






GCCCCGGGAGGAGCAGTTC






AATAGCACCTACCGGGTGG






TGTCCGTGCTGACCGTGCT






GCACCAGGACTGGCTGAAC






GGCAAGGAATACAAGTGTA






AGGTGTCCAACAAGGGCCT






GCCCAGCAGCATCGAGAAA






ACCATCAGCAAGGCCAAGG






GCCAGCCTCGGGAGCCCCA






GGTGTACACCCTGCCCCCTA






GCCAAGAGGAGATGACCAA






GAATCAGGTGTCCCTGACC






TGCCTGGTGAAGGGCTTCT






ACCCCAGCGACATCGCCGT






GGAGTGGGAGAGCAACGG






CCAGCCCGAGAACAACTAC






AAGACCACCCCCCCTGTGCT






GGACAGCGACGGCAGCTTC






TTCCTGTACAGCAGGCTGA






CCGTGGACAAGAGCCGGTG






GCAGGAGGGCAACGTCTTT






AGCTGCTCCGTGATGCACG






AGGCCCTGCACAACCACTA






CACCCAGAAGAGCCTGTCC






CTGAGCCTGGGCAAGATGT






TCTGGGTGCTGGTCGTGGT






GGGTGGCGTGCTGGCCTGC






TACAGCCTGCTGGTGACAG






TGGCCTTCATCATCTTTTGG






GTGAGGAGCAAGCGGAGC






AGAGGCGGCCACAGCGACT






ACATGAACATGACCCCCCG






GAGGCCTGGCCCCACCCGG






AAGCACTACCAGCCCTACG






CCCCTCCCAGGGACTTCGCC






GCCTACCGGAGCCGGGTGA






AGTTCAGCCGGAGCGCCGA






CGCCCCTGCCTACCAGCAG






GGCCAGAACCAGCTGTACA






ACGAGCTGAACCTGGGCCG






GAGGGAGGAGTACGACGT






GCTGGACAAGCGGAGAGG






CCGGGACCCTGAGATGGGC






GGCAAGCCCCGGAGAAAG






AACCCTCAGGAGGGCCTGT






ATAACGAACTGCAGAAAGA






CAAGATGGCCGAGGCCTAC






AGCGAGATCGGCATGAAGG






GCGAGCGGCGGAGGGGCA






AGGGCCACGACGGCCTGTA






CCAGGGCCTGAGCACCGCC






ACCAAGGATACCTACGACG






CCCTGCACATGCAGGCCCT






GCCCCCCAGA





Murine ROR1
595
DIKMTQSPSSMYASLGERVTITCKA
596
GACATCAAGATGACCCAGA


(VL-VH).

SPDINSYLSWFQQKPGKSPKTLIYR

GCCCCAGCTCTATGTACGCC


CD8α.

ANRLVDGVPSRFSGGGSGQDYSLT

AGCCTGGGCGAGCGCGTGA


CD28z

INSLEYEDMGIYYCLQYDEFPYTFG

CCATCACATGCAAGGCCAG




GGTKLEMKGSTSGSGKPGSGEGST

CCCCGACATCAACAGCTACC




KGEVKLVESGGGLVKPGGSLKLSCA

TGTCCTGGTTCCAGCAGAA




ASGFTFSSYAMSWVRQIPEKRLEW

GCCCGGCAAGAGCCCCAAG




VASISRGGTTYYPDSVKGRFTISRD

ACCCTGATCTACCGGGCCA




NVRNILYLQMSSLRSEDTAMYYCG

ACCGGCTGGTGGACGGCGT




RYDYDGYYAMDYWGQGTSVTVSS

GCCAAGCAGATTTTCCGGC




KPTTTPAPRPPTPAPTIASQPLSLRP

GGAGGCAGCGGCCAGGAC




EACRPAAGGAVHTRGLDFACDIYI

TACAGCCTGACCATCAACA




WAPLAGTCGVLLLSLVITLYCNHRN

GCCTGGAATACGAGGACAT




RSKRSRGGHSDYMNMTPRRPGPT

GGGCATCTACTACTGCCTGC




RKHYQPYAPPRDFAAYRSRVKFSRS

AGTACGACGAGTTCCCCTAC




ADAPAYQQGQNQLYNELNLGRRE

ACCTTCGGAGGCGGCACCA




EYDVLDKRRGRDPEMGGKPRRKN

AGCTGGAAATGAAGGGCA




PQEGLYNELQKDKMAEAYSEIGMK

GCACCTCCGGCAGCGGCAA




GERRRGKGHDGLYQGLSTATKDTY

GCCTGGCAGCGGCGAGGG




DALHMQALPPR

CAGCACCAAGGGCGAAGTG






AAGCTGGTGGAAAGCGGC






GGAGGCCTGGTGAAACCTG






GCGGCAGCCTGAAGCTGAG






CTGCGCCGCCAGCGGCTTC






ACCTTCAGCAGCTACGCCAT






GAGCTGGGTCCGACAGATC






CCCGAGAAGCGGCTGGAAT






GGGTGGCCAGCATCAGCAG






GGGCGGCACCACCTACTAC






CCCGACAGCGTGAAGGGCC






GGTTCACCATCAGCCGGGA






CAACGTGCGGAACATCCTG






TACCTGCAGATGAGCAGCC






TGCGGAGCGAGGACACCGC






CATGTACTACTGCGGCAGA






TACGACTACGACGGCTACT






ACGCCATGGATTACTGGGG






CCAGGGCACCAGCGTGACC






GTGTCTAGCAAGCCCACCA






CCACCCCTGCCCCTAGACCT






CCAACCCCAGCCCCTACAAT






CGCCAGCCAGCCCCTGAGC






CTGAGGCCCGAAGCCTGTA






GACCTGCCGCTGGCGGAGC






CGTGCACACCAGAGGCCTG






GATTTCGCCTGCGACATCTA






CATCTGGGCCCCTCTGGCC






GGCACCTGTGGCGTGCTGC






TGCTGAGCCTGGTCATCACC






CTGTACTGCAACCACCGGA






ATAGGAGCAAGCGGAGCA






GAGGCGGCCACAGCGACTA






CATGAACATGACCCCCCGG






AGGCCTGGCCCCACCCGGA






AGCACTACCAGCCCTACGCC






CCTCCCAGGGACTTCGCCG






CCTACCGGAGCCGGGTGAA






GTTCAGCCGGAGCGCCGAC






GCCCCTGCCTACCAGCAGG






GCCAGAACCAGCTGTACAA






CGAGCTGAACCTGGGCCGG






AGGGAGGAGTACGACGTG






CTGGACAAGCGGAGAGGCC






GGGACCCTGAGATGGGCG






GCAAGCCCCGGAGAAAGAA






CCCTCAGGAGGGCCTGTAT






AACGAACTGCAGAAAGACA






AGATGGCCGAGGCCTACAG






CGAGATCGGCATGAAGGGC






GAGCGGCGGAGGGGCAAG






GGCCACGACGGCCTGTACC






AGGGCCTGAGCACCGCCAC






CAAGGATACCTACGACGCC






CTGCACATGCAGGCCCTGC






CCCCCAGA





Murine ROR1
597
DIKMTQSPSSMYASLGERVTITCKA
598
GACATCAAGATGACCCAGA


(VL-VH).

SPDINSYLSWFQQKPGKSPKTLIYR

GCCCCAGCTCTATGTACGCC


CD8α(2x).

ANRLVDGVPSRFSGGGSGQDYSLT

AGCCTGGGCGAGCGCGTGA


CD28z

INSLEYEDMGIYYCLQYDEFPYTFG

CCATCACATGCAAGGCCAG




GGTKLEMKGSTSGSGKPGSGEGST

CCCCGACATCAACAGCTACC




KGEVKLVESGGGLVKPGGSLKLSCA

TGTCCTGGTTCCAGCAGAA




ASGFTFSSYAMSWVRQIPEKRLEW

GCCCGGCAAGAGCCCCAAG




VASISRGGTTYYPDSVKGRFTISRD

ACCCTGATCTACCGGGCCA




NVRNILYLQMSSLRSEDTAMYYCG

ACCGGCTGGTGGACGGCGT




RYDYDGYYAMDYWGQGTSVTVSS

GCCAAGCAGATTTTCCGGC




KPTTTPAPRPPTPAPTIASQPLSLRP

GGAGGCAGCGGCCAGGAC




EASRPAAGGAVHTRGLDFASDKPT

TACAGCCTGACCATCAACA




TTPAPRPPTPAPTIASQPLSLRPEAC

GCCTGGAATACGAGGACAT




RPAAGGAVHTRGLDFACDIYIWAP

GGGCATCTACTACTGCCTGC




LAGTCGVLLLSLVITLYCNHRNRSKR

AGTACGACGAGTTCCCCTAC




SRGGHSDYMNMTPRRPGPTRKHY

ACCTTCGGAGGCGGCACCA




QPYAPPRDFAAYRSRVKFSRSADA

AGCTGGAAATGAAGGGCA




PAYQQGQNQLYNELNLGRREEYD

GCACCTCCGGCAGCGGCAA




VLDKRRGRDPEMGGKPRRKNPQE

GCCTGGCAGCGGCGAGGG




GLYNELQKDKMAEAYSEIGMKGER

CAGCACCAAGGGCGAAGTG




RRGKGHDGLYQGLSTATKDTYDAL

AAGCTGGTGGAAAGCGGC




HMQALPPR

GGAGGCCTGGTGAAACCTG






GCGGCAGCCTGAAGCTGAG






CTGCGCCGCCAGCGGCTTC






ACCTTCAGCAGCTACGCCAT






GAGCTGGGTCCGACAGATC






CCCGAGAAGCGGCTGGAAT






GGGTGGCCAGCATCAGCAG






GGGCGGCACCACCTACTAC






CCCGACAGCGTGAAGGGCC






GGTTCACCATCAGCCGGGA






CAACGTGCGGAACATCCTG






TACCTGCAGATGAGCAGCC






TGCGGAGCGAGGACACCGC






CATGTACTACTGCGGCAGA






TACGACTACGACGGCTACT






ACGCCATGGATTACTGGGG






CCAGGGCACCAGCGTGACC






GTGTCTAGCAAACCTACTAC






AACTCCTGCCCCCCGGCCTC






CTACACCAGCTCCTACTATC






GCCTCCCAGCCACTCAGTCT






CAGACCCGAGGCTTCTAGG






CCAGCGGCCGGAGGCGCG






GTCCACACCCGCGGGCTGG






ACTTTGCATCCGATAAGCCC






ACCACCACCCCTGCCCCTAG






ACCTCCAACCCCAGCCCCTA






CAATCGCCAGCCAGCCCCT






GAGCCTGAGGCCCGAAGCC






TGTAGACCTGCCGCTGGCG






GAGCCGTGCACACCAGAGG






CCTGGATTTCGCCTGCGACA






TCTACATCTGGGCCCCTCTG






GCCGGCACCTGTGGCGTGC






TGCTGCTGAGCCTGGTCATC






ACCCTGTACTGCAACCACCG






GAATAGGAGCAAGCGGAG






CAGAGGCGGCCACAGCGAC






TACATGAACATGACCCCCCG






GAGGCCTGGCCCCACCCGG






AAGCACTACCAGCCCTACG






CCCCTCCCAGGGACTTCGCC






GCCTACCGGAGCCGGGTGA






AGTTCAGCCGGAGCGCCGA






CGCCCCTGCCTACCAGCAG






GGCCAGAACCAGCTGTACA






ACGAGCTGAACCTGGGCCG






GAGGGAGGAGTACGACGT






GCTGGACAAGCGGAGAGG






CCGGGACCCTGAGATGGGC






GGCAAGCCCCGGAGAAAG






AACCCTCAGGAGGGCCTGT






ATAACGAACTGCAGAAAGA






CAAGATGGCCGAGGCCTAC






AGCGAGATCGGCATGAAGG






GCGAGCGGCGGAGGGGCA






AGGGCCACGACGGCCTGTA






CCAGGGCCTGAGCACCGCC






ACCAAGGATACCTACGACG






CCCTGCACATGCAGGCCCT






GCCCCCCAGA





Murine
599
DIKMTQSPSSMYASLGERVTITCKA
600
GACATCAAGATGACCCAGA


ROR1

SPDINSYLSWFQQKPGKSPKTLIYR

GCCCCAGCTCTATGTACGCC


(VL-VH).

ANRLVDGVPSRFSGGGSGQDYSLT

AGCCTGGGCGAGCGCGTGA


CD8a(3x).

INSLEYEDMGIYYCLQYDEFPYTFG

CCATCACATGCAAGGCCAG


CD28z

GGTKLEMKGSTSGSGKPGSGEGST

CCCCGACATCAACAGCTACC




KGEVKLVESGGGLVKPGGSLKLSCA

TGTCCTGGTTCCAGCAGAA




ASGFTFSSYAMSWVRQIPEKRLEW

GCCCGGCAAGAGCCCCAAG




VASISRGGTTYYPDSVKGRFTISRD

ACCCTGATCTACCGGGCCA




NVRNILYLQMSSLRSEDTAMYYCG

ACCGGCTGGTGGACGGCGT




RYDYDGYYAMDYWGQGTSVTVSS

GCCAAGCAGATTTTCCGGC




KPTTTPAPRPPTPAPTIASQPLSLRP

GGAGGCAGCGGCCAGGAC




EASRPAAGGAVHTRGLDFASDKPT

TACAGCCTGACCATCAACA




TTPAPRPPTPAPTIASQPLSLRPEAS

GCCTGGAATACGAGGACAT




RPAAGGAVHTRGLDFASDKPTTTP

GGGCATCTACTACTGCCTGC




APRPPTPAPTIASQPLSLRPEACRPA

AGTACGACGAGTTCCCCTAC




AGGAVHTRGLDFACDIYIWAPLAG

ACCTTCGGAGGCGGCACCA




TCGVLLLSLVITLYCNHRNRSKRSRG

AGCTGGAAATGAAGGGCA




GHSDYMNMTPRRPGPTRKHYQPY

GCACCTCCGGCAGCGGCAA




APPRDFAAYRSRVKFSRSADAPAY

GCCTGGCAGCGGCGAGGG




QQGQNQLYNELNLGRREEYDVLD

CAGCACCAAGGGCGAAGTG




KRRGRDPEMGGKPRRKNPQEGLY

AAGCTGGTGGAAAGCGGC




NELQKDKMAEAYSEIGMKGERRR

GGAGGCCTGGTGAAACCTG




GKGHDGLYQGLSTATKDTYDALH

GCGGCAGCCTGAAGCTGAG




MQALPPR

CTGCGCCGCCAGCGGCTTC






ACCTTCAGCAGCTACGCCAT






GAGCTGGGTCCGACAGATC






CCCGAGAAGCGGCTGGAAT






GGGTGGCCAGCATCAGCAG






GGGCGGCACCACCTACTAC






CCCGACAGCGTGAAGGGCC






GGTTCACCATCAGCCGGGA






CAACGTGCGGAACATCCTG






TACCTGCAGATGAGCAGCC






TGCGGAGCGAGGACACCGC






CATGTACTACTGCGGCAGA






TACGACTACGACGGCTACT






ACGCCATGGATTACTGGGG






CCAGGGCACCAGCGTGACC






GTGTCTAGCAAGCCTACCAC






CACCCCCGCACCTCGTCCTC






CAACCCCTGCACCTACGATT






GCCAGTCAGCCTCTTTCACT






GCGGCCTGAGGCCAGCAGA






CCAGCTGCCGGCGGTGCCG






TCCATACAAGAGGACTGGA






CTTCGCGTCCGATAAACCTA






CTACCACTCCAGCCCCAAGG






CCCCCAACCCCAGCACCGAC






TATCGCATCACAGCCTTTGT






CACTGCGTCCTGAAGCCAG






CCGGCCAGCTGCAGGGGG






GGCCGTCCACACAAGGGGA






CTCGACTTTGCGAGTGATA






AGCCCACCACCACCCCTGCC






CCTAGACCTCCAACCCCAGC






CCCTACAATCGCCAGCCAGC






CCCTGAGCCTGAGGCCCGA






AGCCTGTAGACCTGCCGCT






GGCGGAGCCGTGCACACCA






GAGGCCTGGATTTCGCCTG






CGACATCTACATCTGGGCCC






CTCTGGCCGGCACCTGTGG






CGTGCTGCTGCTGAGCCTG






GTCATCACCCTGTACTGCAA






CCACCGGAATAGGAGCAAG






CGGAGCAGAGGCGGCCAC






AGCGACTACATGAACATGA






CCCCCCGGAGGCCTGGCCC






CACCCGGAAGCACTACCAG






CCCTACGCCCCTCCCAGGGA






CTTCGCCGCCTACCGGAGC






CGGGTGAAGTTCAGCCGGA






GCGCCGACGCCCCTGCCTA






CCAGCAGGGCCAGAACCAG






CTGTACAACGAGCTGAACC






TGGGCCGGAGGGAGGAGT






ACGACGTGCTGGACAAGCG






GAGAGGCCGGGACCCTGA






GATGGGCGGCAAGCCCCG






GAGAAAGAACCCTCAGGAG






GGCCTGTATAACGAACTGC






AGAAAGACAAGATGGCCGA






GGCCTACAGCGAGATCGGC






ATGAAGGGCGAGCGGCGG






AGGGGCAAGGGCCACGAC






GGCCTGTACCAGGGCCTGA






GCACCGCCACCAAGGATAC






CTACGACGCCCTGCACATGC






AGGCCCTGCCCCCCAGA





Murine ROR1
601
DIKMTQSPSSMYASLGERVTITCKA
602
GACATCAAGATGACCCAGA


(VL-VH).

SPDINSYLSWFQQKPGKSPKTLIYR

GCCCCAGCTCTATGTACGCC


CD8a(4x).

ANRLVDGVPSRFSGGGSGQDYSLT

AGCCTGGGCGAGCGCGTGA


CD28z

INSLEYEDMGIYYCLQYDEFPYTFG

CCATCACATGCAAGGCCAG




GGTKLEMKGSTSGSGKPGSGEGST

CCCCGACATCAACAGCTACC




KGEVKLVESGGGLVKPGGSLKLSCA

TGTCCTGGTTCCAGCAGAA




ASGFTFSSYAMSWVRQIPEKRLEW

GCCCGGCAAGAGCCCCAAG




VASISRGGTTYYPDSVKGRFTISRD

ACCCTGATCTACCGGGCCA




NVRNILYLQMSSLRSEDTAMYYCG

ACCGGCTGGTGGACGGCGT




RYDYDGYYAMDYWGQGTSVTVSS

GCCAAGCAGATTTTCCGGC




KPTTTPAPRPPTPAPTIASQPLSLRP

GGAGGCAGCGGCCAGGAC




EASRPAAGGAVHTRGLDFASDKPT

TACAGCCTGACCATCAACA




TTPAPRPPTPAPTIASQPLSLRPEAS

GCCTGGAATACGAGGACAT




RPAAGGAVHTRGLDFASDKPTTTP

GGGCATCTACTACTGCCTGC




APRPPTPAPTIASQPLSLRPEASRPA

AGTACGACGAGTTCCCCTAC




AGGAVHTRGLDFASDKPTTTPAPR

ACCTTCGGAGGCGGCACCA




PPTPAPTIASQPLSLRPEACRPAAG

AGCTGGAAATGAAGGGCA




GAVHTRGLDFACDIYIWAPLAGTC

GCACCTCCGGCAGCGGCAA




GVLLLSLVITLYCNHRNRSKRSRGG

GCCTGGCAGCGGCGAGGG




HSDYMNMTPRRPGPTRKHYQPYA

CAGCACCAAGGGCGAAGTG




PPRDFAAYRSRVKFSRSADAPAYQ

AAGCTGGTGGAAAGCGGC




QGQNQLYNELNLGRREEYDVLDKR

GGAGGCCTGGTGAAACCTG




RGRDPEMGGKPRRKNPQEGLYNE

GCGGCAGCCTGAAGCTGAG




LQKDKMAEAYSEIGMKGERRRGK

CTGCGCCGCCAGCGGCTTC




GHDGLYQGLSTATKDTYDALHMQ

ACCTTCAGCAGCTACGCCAT




ALPPR

GAGCTGGGTCCGACAGATC






CCCGAGAAGCGGCTGGAAT






GGGTGGCCAGCATCAGCAG






GGGCGGCACCACCTACTAC






CCCGACAGCGTGAAGGGCC






GGTTCACCATCAGCCGGGA






CAACGTGCGGAACATCCTG






TACCTGCAGATGAGCAGCC






TGCGGAGCGAGGACACCGC






CATGTACTACTGCGGCAGA






TACGACTACGACGGCTACT






ACGCCATGGATTACTGGGG






CCAGGGCACCAGCGTGACC






GTGTCTAGCAAGCCTACCAC






CACCCCCGCACCTCGTCCTC






CAACCCCTGCACCTACGATT






GCCAGTCAGCCTCTTTCACT






GCGGCCTGAGGCCAGCAGA






CCAGCTGCCGGCGGTGCCG






TCCATACAAGAGGACTGGA






CTTCGCGTCCGATAAACCTA






CTACCACTCCAGCCCCAAGG






CCCCCAACCCCAGCACCGAC






TATCGCATCACAGCCTTTGT






CACTGCGTCCTGAAGCCAG






CCGGCCAGCTGCAGGGGG






GGCCGTCCACACAAGGGGA






CTCGACTTTGCGAGTGATA






AACCTACTACAACTCCTGCC






CCCCGGCCTCCTACACCAGC






TCCTACTATCGCCTCCCAGC






CACTCAGTCTCAGACCCGA






GGCTTCTAGGCCAGCGGCC






GGAGGCGCGGTCCACACCC






GCGGGCTGGACTTTGCATC






CGATAAGCCCACCACCACCC






CTGCCCCTAGACCTCCAACC






CCAGCCCCTACAATCGCCAG






CCAGCCCCTGAGCCTGAGG






CCCGAAGCCTGTAGACCTG






CCGCTGGCGGAGCCGTGCA






CACCAGAGGCCTGGATTTC






GCCTGCGACATCTACATCTG






GGCCCCTCTGGCCGGCACC






TGTGGCGTGCTGCTGCTGA






GCCTGGTCATCACCCTGTAC






TGCAACCACCGGAATAGGA






GCAAGCGGAGCAGAGGCG






GCCACAGCGACTACATGAA






CATGACCCCCCGGAGGCCT






GGCCCCACCCGGAAGCACT






ACCAGCCCTACGCCCCTCCC






AGGGACTTCGCCGCCTACC






GGAGCCGGGTGAAGTTCAG






CCGGAGCGCCGACGCCCCT






GCCTACCAGCAGGGCCAGA






ACCAGCTGTACAACGAGCT






GAACCTGGGCCGGAGGGA






GGAGTACGACGTGCTGGAC






AAGCGGAGAGGCCGGGAC






CCTGAGATGGGCGGCAAGC






CCCGGAGAAAGAACCCTCA






GGAGGGCCTGTATAACGAA






CTGCAGAAAGACAAGATGG






CCGAGGCCTACAGCGAGAT






CGGCATGAAGGGCGAGCG






GCGGAGGGGCAAGGGCCA






CGACGGCCTGTACCAGGGC






CTGAGCACCGCCACCAAGG






ATACCTACGACGCCCTGCAC






ATGCAGGCCCTGCCCCCCA






GA





Murine
603
DIKMTQSPSSMYASLGERVTITCKA
604
GACATCAAGATGACCCAGA


ROR1

SPDINSYLSWFQQKPGKSPKTLIYR

GCCCCAGCTCTATGTACGCC


(VL-VH).

ANRLVDGVPSRFSGGGSGQDYSLT

AGCCTGGGCGAGCGCGTGA


LNGFR ECD.

INSLEYEDMGIYYCLQYDEFPYTFG

CCATCACATGCAAGGCCAG


CD8TM.

GGTKLEMKGSTSGSGKPGSGEGST

CCCCGACATCAACAGCTACC


CD28z

KGEVKLVESGGGLVKPGGSLKLSCA

TGTCCTGGTTCCAGCAGAA




ASGFTFSSYAMSWVRQIPEKRLEW

GCCCGGCAAGAGCCCCAAG




VASISRGGTTYYPDSVKGRFTISRD

ACCCTGATCTACCGGGCCA




NVRNILYLQMSSLRSEDTAMYYCG

ACCGGCTGGTGGACGGCGT




RYDYDGYYAMDYWGQGTSVTVSS

GCCAAGCAGATTTTCCGGC




KEACPTGLYTHSGECCKACNLGEG

GGAGGCAGCGGCCAGGAC




VAQPCGANQTVCEPCLDSVTFSDV

TACAGCCTGACCATCAACA




VSATEPCKPCTECVGLQSMSAPCV

GCCTGGAATACGAGGACAT




EADDAVCRCAYGYYQDETTGRCEA

GGGCATCTACTACTGCCTGC




CRVCEAGSGLVFSCQDKCINTVCEE

AGTACGACGAGTTCCCCTAC




CPDGTYSDEANHVDPCLPCTVCED

ACCTTCGGAGGCGGCACCA




TERQLRECTRWADAECEEIPGRWI

AGCTGGAAATGAAGGGCA




TRSTPPEGSDSTAPSTQEPEAPPEQ

GCACCTCCGGCAGCGGCAA




DLIASTVAGVVTTVMGSSQPVVTR

GCCTGGCAGCGGCGAGGG




GTTDNIYIWAPLAGTCGVLLLSLVIT

CAGCACCAAGGGCGAAGTG




LYCNHRNRSKRSRGGHSDYMNMT

AAGCTGGTGGAAAGCGGC




PRRPGPTRKHYQPYAPPRDFAAYR

GGAGGCCTGGTGAAACCTG




SRVKFSRSADAPAYQQGONQLYN

GCGGCAGCCTGAAGCTGAG




ELNLGRREEYDVLDKRRGRDPEMG

CTGCGCCGCCAGCGGCTTC




GKPRRKNPQEGLYNELQKDKMAE

ACCTTCAGCAGCTACGCCAT




AYSEIGMKGERRRGKGHDGLYQGL

GAGCTGGGTCCGACAGATC




STATKDTYDALHMQALPPR

CCCGAGAAGCGGCTGGAAT






GGGTGGCCAGCATCAGCAG






GGGCGGCACCACCTACTAC






CCCGACAGCGTGAAGGGCC






GGTTCACCATCAGCCGGGA






CAACGTGCGGAACATCCTG






TACCTGCAGATGAGCAGCC






TGCGGAGCGAGGACACCGC






CATGTACTACTGCGGCAGA






TACGACTACGACGGCTACT






ACGCCATGGATTACTGGGG






CCAGGGCACCAGCGTGACC






GTGTCTAGCAAGGAGGCAT






GCCCCACAGGCCTGTACAC






ACACAGCGGTGAGTGCTGC






AAAGCCTGCAACCTGGGCG






AGGGTGTGGCCCAGCCTTG






TGGAGCCAACCAGACCGTG






TGTGAGCCCTGCCTGGACA






GCGTGACGTTCTCCGACGT






GGTGAGCGCGACCGAGCC






GTGCAAGCCGTGCACCGAG






TGCGTGGGGCTCCAGAGCA






TGTCGGCGCCGTGCGTGGA






GGCCGACGACGCCGTGTGC






CGCTGCGCCTACGGCTACT






ACCAGGATGAGACGACTGG






GCGCTGCGAGGCGTGCCGC






GTGTGCGAGGCGGGCTCG






GGCCTCGTGTTCTCCTGCCA






GGACAAGCAGAACACCGTG






TGCGAGGAGTGCCCCGACG






GCACGTATTCCGACGAGGC






CAACCACGTGGACCCGTGC






CTGCCCTGCACCGTGTGCG






AGGACACCGAGCGCCAGCT






CCGCGAGTGCACACGCTGG






GCCGACGCCGAGTGCGAG






GAGATCCCTGGCCGTTGGA






TTACACGGTCCACACCCCCA






GAGGGCTCGGACAGCACAG






CCCCCAGCACCCAGGAGCC






TGAGGCACCTCCAGAACAA






GACCTCATAGCCAGCACGG






TGGCAGGTGTGGTGACCAC






AGTGATGGGCAGCTCCCAG






CCCGTGGTGACCCGAGGCA






CCACCGACAACATCTACATC






TGGGCCCCTCTGGCCGGCA






CCTGTGGCGTGCTGCTGCT






GAGCCTGGTCATCACCCTGT






ACTGCAACCACCGGAATAG






GAGCAAGCGGAGCAGAGG






CGGCCACAGCGACTACATG






AACATGACCCCCCGGAGGC






CTGGCCCCACCCGGAAGCA






CTACCAGCCCTACGCCCCTC






CCAGGGACTTCGCCGCCTA






CCGGAGCCGGGTGAAGTTC






AGCCGGAGCGCCGACGCCC






CTGCCTACCAGCAGGGCCA






GAACCAGCTGTACAACGAG






CTGAACCTGGGCCGGAGGG






AGGAGTACGACGTGCTGGA






CAAGCGGAGAGGCCGGGA






CCCTGAGATGGGCGGCAAG






CCCCGGAGAAAGAACCCTC






AGGAGGGCCTGTATAACGA






ACTGCAGAAAGACAAGATG






GCCGAGGCCTACAGCGAGA






TCGGCATGAAGGGCGAGCG






GCGGAGGGGCAAGGGCCA






CGACGGCCTGTACCAGGGC






CTGAGCACCGCCACCAAGG






ATACCTACGACGCCCTGCAC






ATGCAGGCCCTGCCCCCCA






GA





Murine
605
EVKLVESGGGLVKPGGSLKLSCAAS
606
GAAGTGAAGCTGGTGGAAA


ROR1

GFTFSSYAMSWVRQIPEKRLEWVA

GCGGCGGAGGCCTGGTGA


(VH-VL).

SISRGGTTYYPDSVKGRFTISRDNVR

AACCTGGCGGCAGCCTGAA


CD8a(3x).

NILYLQMSSLRSEDTAMYYCGRYD

GCTGAGCTGCGCCGCCAGC


41BBz

YDGYYAMDYWGQGTSVTVSSGST

GGCTTCACCTTCAGCAGCTA




SGSGKPGSGEGSTKGDIKMTQSPS

CGCCATGAGCTGGGTCCGA




SMYASLGERVTITCKASPDINSYLS

CAGATCCCCGAGAAGCGGC




WFQQKPGKSPKTLIYRANRLVDGV

TGGAATGGGTGGCCAGCAT




PSRFSGGGSGQDYSLTINSLEYEDM

CAGCAGGGGCGGCACCACC




GIYYCLQYDEFPYTFGGGTKLEMKK

TACTACCCCGACAGCGTGA




PTTTPAPRPPTPAPTIASQPLSLRPE

AGGGCCGGTTCACCATCAG




ASRPAAGGAVHTRGLDFASDKPTT

CCGGGACAACGTGCGGAAC




TPAPRPPTPAPTIASQPLSLRPEASR

ATCCTGTACCTGCAGATGA




PAAGGAVHTRGLDFASDKPTTTPA

GCAGCCTGCGGAGCGAGG




PRPPTPAPTIASQPLSLRPEACRPAA

ACACCGCCATGTACTACTGC




GGAVHTRGLDFACDIYIWAPLAGT

GGCAGATACGACTACGACG




CGVLLLSLVITLYCNHRNKRGRKKLL

GCTACTACGCCATGGATTAC




YIFKQPFMRPVQTTQEEDGCSCRF

TGGGGCCAGGGCACCAGC




PEEEEGGCELRVKFSRSADAPAYQ

GTGACCGTGTCTAGCGGCA




QGQNQLYNELNLGRREEYDVLDKR

GCACCTCCGGCAGCGGCAA




RGRDPEMGGKPRRKNPQEGLYNE

GCCTGGCAGCGGCGAGGG




LQKDKMAEAYSEIGMKGERRRGK

CAGCACCAAGGGCGACATC




GHDGLYQGLSTATKDTYDALHMQ

AAGATGACCCAGAGCCCCA




ALPPR

GCTCTATGTACGCCAGCCTG






GGCGAGCGCGTGACCATCA






CATGCAAGGCCAGCCCCGA






CATCAACAGCTACCTGTCCT






GGTTCCAGCAGAAGCCCGG






CAAGAGCCCCAAGACCCTG






ATCTACCGGGCCAACCGGC






TGGTGGACGGCGTGCCAAG






CAGATTTTCCGGCGGAGGC






AGCGGCCAGGACTACAGCC






TGACCATCAACAGCCTGGA






ATACGAGGACATGGGCATC






TACTACTGCCTGCAGTACGA






CGAGTTCCCCTACACCTTCG






GAGGCGGCACCAAGCTGGA






AATGAAGAAGCCTACCACC






ACCCCCGCACCTCGTCCTCC






AACCCCTGCACCTACGATTG






CCAGTCAGCCTCTTTCACTG






CGGCCTGAGGCCAGCAGAC






CAGCTGCCGGCGGTGCCGT






CCATACAAGAGGACTGGAC






TTCGCGTCCGATAAACCTAC






TACCACTCCAGCCCCAAGGC






CCCCAACCCCAGCACCGACT






ATCGCATCACAGCCTTTGTC






ACTGCGTCCTGAAGCCAGC






CGGCCAGCTGCAGGGGGG






GCCGTCCACACAAGGGGAC






TCGACTTTGCGAGTGATAA






GCCCACCACCACCCCTGCCC






CTAGACCTCCAACCCCAGCC






CCTACAATCGCCAGCCAGCC






CCTGAGCCTGAGGCCCGAA






GCCTGTAGACCTGCCGCTG






GCGGAGCCGTGCACACCAG






AGGCCTGGATTTCGCCTGC






GACATCTACATCTGGGCCCC






TCTGGCCGGCACCTGTGGC






GTGCTGCTGCTGAGCCTGG






TCATCACCCTGTACTGCAAC






CACCGGAATAAGAGAGGCC






GGAAGAAACTGCTGTACAT






CTTCAAGCAGCCCTTCATGC






GGCCCGTGCAGACCACCCA






GGAAGAGGACGGCTGCAG






CTGCCGGTTCCCCGAGGAA






GAGGAAGGCGGCTGCGAA






CTGCGGGTGAAGTTCAGCC






GGAGCGCCGACGCCCCTGC






CTACCAGCAGGGCCAGAAC






CAGCTGTACAACGAGCTGA






ACCTGGGCCGGAGGGAGG






AGTACGACGTGCTGGACAA






GCGGAGAGGCCGGGACCC






TGAGATGGGCGGCAAGCCC






CGGAGAAAGAACCCTCAGG






AGGGCCTGTATAACGAACT






GCAGAAAGACAAGATGGCC






GAGGCCTACAGCGAGATCG






GCATGAAGGGCGAGCGGC






GGAGGGGCAAGGGCCACG






ACGGCCTGTACCAGGGCCT






GAGCACCGCCACCAAGGAT






ACCTACGACGCCCTGCACAT






GCAGGCCCTGCCCCCCAGA





Murine
607
EVKLVESGGGLVKPGGSLKLSCAAS
608
GAAGTGAAGCTGGTGGAAA


ROR1

GFTFSSYAMSWVRQIPEKRLEWVA

GCGGCGGAGGCCTGGTGA


(VH-VL).

SISRGGTTYYPDSVKGRFTISRDNVR

AACCTGGCGGCAGCCTGAA


IgG4 Fcm.

NILYLQMSSLRSEDTAMYYCGRYD

GCTGAGCTGCGCCGCCAGC


CD8aTM.

YDGYYAMDYWGQGTSVTVSSGST

GGCTTCACCTTCAGCAGCTA


41BBz

SGSGKPGSGEGSTKGDIKMTQSPS

CGCCATGAGCTGGGTCCGA




SMYASLGERVTITCKASPDINSYLS

CAGATCCCCGAGAAGCGGC




WFQQKPGKSPKTLIYRANRLVDGV

TGGAATGGGTGGCCAGCAT




PSRFSGGGSGQDYSLTINSLEYEDM

CAGCAGGGGCGGCACCACC




GIYYCLQYDEFPYTFGGGTKLEMKE

TACTACCCCGACAGCGTGA




SKYGPPCPPCPAPEFEGGPSVFLFP

AGGGCCGGTTCACCATCAG




PKPKDTLMISRTPEVTCVVVDVSQE

CCGGGACAACGTGCGGAAC




DPEVQFNWYVDGVEVHNAKTKPR

ATCCTGTACCTGCAGATGA




EEQFQSTYRVVSVLTVLHQDWLNG

GCAGCCTGCGGAGCGAGG




KEYKCKVSNKGLPSSIEKTISKAKGQ

ACACCGCCATGTACTACTGC




PREPQVYTLPPSQEEMTKNQVSLT

GGCAGATACGACTACGACG




CLVKGFYPSDIAVEWESNGQPENN

GCTACTACGCCATGGATTAC




YKTTPPVLDSDGSFFLYSRLTVDKSR

TGGGGCCAGGGCACCAGC




WQEGNVFSCSVMHEALHNHYTQ

GTGACCGTGTCTAGCGGCA




KSLSLSLGKMIYIWAPLAGTCGVLLL

GCACCTCCGGCAGCGGCAA




SLVITLYCNHRNKRGRKKLLYIFKQP

GCCTGGCAGCGGCGAGGG




FMRPVQTTQEEDGCSCRFPEEEEG

CAGCACCAAGGGCGACATC




GCELRVKFSRSADAPAYQQGQNQ

AAGATGACCCAGAGCCCCA




LYNELNLGRREEYDVLDKRRGRDPE

GCTCTATGTACGCCAGCCTG




MGGKPRRKNPQEGLYNELQKDK

GGCGAGCGCGTGACCATCA




MAEAYSEIGMKGERRRGKGHDGL

CATGCAAGGCCAGCCCCGA




YQGLSTATKDTYDALHMQALPPR

CATCAACAGCTACCTGTCCT






GGTTCCAGCAGAAGCCCGG






CAAGAGCCCCAAGACCCTG






ATCTACCGGGCCAACCGGC






TGGTGGACGGCGTGCCAAG






CAGATTTTCCGGCGGAGGC






AGCGGCCAGGACTACAGCC






TGACCATCAACAGCCTGGA






ATACGAGGACATGGGCATC






TACTACTGCCTGCAGTACGA






CGAGTTCCCCTACACCTTCG






GAGGCGGCACCAAGCTGGA






AATGAAGGAGAGCAAGTAC






GGCCCTCCCTGCCCCCCTTG






CCCTGCCCCCGAGTTCGAG






GGCGGACCCAGCGTGTTCC






TGTTCCCCCCCAAGCCCAAG






GACACCCTGATGATCAGCC






GGACCCCCGAGGTGACCTG






TGTGGTGGTGGACGTGTCC






CAGGAGGACCCCGAGGTCC






AGTTCAACTGGTACGTGGA






CGGCGTGGAGGTGCACAAC






GCCAAGACCAAGCCCCGGG






AGGAGCAGTTCCAGAGCAC






CTACCGGGTGGTGTCCGTG






CTGACCGTGCTGCACCAGG






ACTGGCTGAACGGCAAGGA






ATACAAGTGTAAGGTGTCC






AACAAGGGCCTGCCCAGCA






GCATCGAGAAAACCATCAG






CAAGGCCAAGGGCCAGCCT






CGGGAGCCCCAGGTGTACA






CCCTGCCCCCTAGCCAAGA






GGAGATGACCAAGAATCAG






GTGTCCCTGACCTGCCTGGT






GAAGGGCTTCTACCCCAGC






GACATCGCCGTGGAGTGGG






AGAGCAACGGCCAGCCCGA






GAACAACTACAAGACCACC






CCCCCTGTGCTGGACAGCG






ACGGCAGCTTCTTCCTGTAC






AGCAGGCTGACCGTGGACA






AGAGCCGGTGGCAGGAGG






GCAACGTCTTTAGCTGCTCC






GTGATGCACGAGGCCCTGC






ACAACCACTACACCCAGAA






GAGCCTGTCCCTGAGCCTG






GGCAAGATGATCTACATCT






GGGCCCCTCTGGCCGGCAC






CTGTGGCGTGCTGCTGCTG






AGCCTGGTCATCACCCTGTA






CTGCAACCACCGGAATAAG






AGAGGCCGGAAGAAACTGC






TGTACATCTTCAAGCAGCCC






TTCATGCGGCCCGTGCAGA






CCACCCAGGAAGAGGACGG






CTGCAGCTGCCGGTTCCCC






GAGGAAGAGGAAGGCGGC






TGCGAACTGCGGGTGAAGT






TCAGCCGGAGCGCCGACGC






CCCTGCCTACCAGCAGGGC






CAGAACCAGCTGTACAACG






AGCTGAACCTGGGCCGGAG






GGAGGAGTACGACGTGCTG






GACAAGCGGAGAGGCCGG






GACCCTGAGATGGGCGGCA






AGCCCCGGAGAAAGAACCC






TCAGGAGGGCCTGTATAAC






GAACTGCAGAAAGACAAGA






TGGCCGAGGCCTACAGCGA






GATCGGCATGAAGGGCGA






GCGGCGGAGGGGCAAGGG






CCACGACGGCCTGTACCAG






GGCCTGAGCACCGCCACCA






AGGATACCTACGACGCCCT






GCACATGCAGGCCCTGCCC






CCCAGA





Murine
609
DVQITQSPSSLYASLGERVTITCKAS
610
GACGTGCAGATCACCCAGA


ROR1_v2 VL

PDINSYLSWFQQKPGKSPKTLIYRA

GCCCCAGCAGCCTGTATGC




NRLVDGVPSRFSGGGSGQDYSLTI

CAGCCTGGGCGAGAGAGTG




NSLEYEDMGIYYCLQYDEFPYTFGG

ACCATTACCTGCAAGGCCA




GTKLEMK

GCCCCGACATCAACAGCTA






CCTGAGCTGGTTCCAGCAG






AAGCCCGGCAAGAGCCCCA






AGACCCTGATCTACCGGGC






CAACAGACTGGTGGATGGC






GTGCCCAGCAGATTCAGCG






GCGGAGGCTCTGGCCAGGA






CTACAGCCTGACCATCAACT






CCCTGGAATACGAGGACAT






GGGCATCTACTACTGCCTGC






AGTACGACGAGTTCCCCTAC






ACCTTCGGAGGCGGCACCA






AGCTGGAAATGAAG





Murine
611
EVKLVESGGGLVKPGGSLKLSCAAS
612
GAAGTGAAGCTGGTGGAAT


ROR1_v2 VH

GFTFSSYAMSWVRQIPEKRLEWVA

CTGGCGGCGGACTCGTGAA




SISRGGTTYYPDSVKGRFTISRDNVR

GCCTGGCGGCTCTCTGAAG




NILYLQMSSLRSEDTAMYYCGRYD

CTGTCTTGTGCCGCCAGCG




YDGYYAMDYWGQGTSVTVSS

GCTTCACCTTCAGCAGCTAC






GCCATGAGCTGGGTGCGGC






AGATCCCCGAGAAGCGGCT






GGAATGGGTGGCCAGCATC






AGCAGAGGCGGAACCACCT






ACTACCCCGACTCTGTGAAG






GGCCGGTTCACCATCAGCC






GGGACAACGTGCGGAACAT






CCTGTACCTGCAGATGAGC






AGCCTGCGGAGCGAGGACA






CCGCCATGTACTACTGTGGC






AGATACGACTACGACGGCT






ACTATGCCATGGATTACTG






GGGCCAGGGCACCAGCGT






GACCGTGTCATCT





Murine
613
DVQITQSPSSLYASLGERVTITCKAS
614
GACGTGCAGATCACCCAGA


ROR1_v2

PDINSYLSWFQQKPGKSPKTLIYRA

GCCCCAGCAGCCTGTATGC


(VL-VH).

NRLVDGVPSRFSGGGSGQDYSLTI

CAGCCTGGGCGAGAGAGTG


CD8a(3x).

NSLEYEDMGIYYCLQYDEFPYTFGG

ACCATTACCTGCAAGGCCA


CD28z

GTKLEMKGSTSGSGKPGSGEGSTK

GCCCCGACATCAACAGCTA




GEVKLVESGGGLVKPGGSLKLSCAA

CCTGAGCTGGTTCCAGCAG




SGFTFSSYAMSWVRQIPEKRLEWV

AAGCCCGGCAAGAGCCCCA




ASISRGGTTYYPDSVKGRFTISRDN

AGACCCTGATCTACCGGGC




VRNILYLQMSSLRSEDTAMYYCGR

CAACAGACTGGTGGATGGC




YDYDGYYAMDYWGQGTSVTVSSK

GTGCCCAGCAGATTCAGCG




PTTTPAPRPPTPAPTIASQPLSLRPE

GCGGAGGCTCTGGCCAGGA




ASRPAAGGAVHTRGLDFASDKPTT

CTACAGCCTGACCATCAACT




TPAPRPPTPAPTIASQPLSLRPEASR

CCCTGGAATACGAGGACAT




PAAGGAVHTRGLDFASDKPTTTPA

GGGCATCTACTACTGCCTGC




PRPPTPAPTIASQPLSLRPEACRPAA

AGTACGACGAGTTCCCCTAC




GGAVHTRGLDFACDIYIWAPLAGT

ACCTTCGGAGGCGGCACCA




CGVLLLSLVITLYCNHRNRSKRSRG

AGCTGGAAATGAAGGGCA




GHSDYMNMTPRRPGPTRKHYQPY

GCACAAGCGGCAGCGGCAA




APPRDFAAYRSRVKFSRSADAPAY

GCCTGGATCTGGCGAGGGA




QQGQNQLYNELNLGRREEYDVLD

AGCACCAAGGGCGAAGTGA




KRRGRDPEMGGKPRRKNPQEGLY

AGCTGGTGGAATCTGGCGG




NELQKDKMAEAYSEIGMKGERRR

CGGACTCGTGAAGCCTGGC




GKGHDGLYQGLSTATKDTYDALH

GGCTCTCTGAAGCTGTCTTG




MQALPPR

TGCCGCCAGCGGCTTCACCT






TCAGCAGCTACGCCATGAG






CTGGGTGCGGCAGATCCCC






GAGAAGCGGCTGGAATGG






GTGGCCAGCATCAGCAGAG






GCGGAACCACCTACTACCCC






GACTCTGTGAAGGGCCGGT






TCACCATCAGCCGGGACAA






CGTGCGGAACATCCTGTAC






CTGCAGATGAGCAGCCTGC






GGAGCGAGGACACCGCCAT






GTACTACTGTGGCAGATAC






GACTACGACGGCTACTATG






CCATGGATTACTGGGGCCA






GGGCACCAGCGTGACCGTG






TCATCTAAGCCTACCACCAC






CCCCGCACCTCGTCCTCCAA






CCCCTGCACCTACGATTGCC






AGTCAGCCTCTTTCACTGCG






GCCTGAGGCCAGCAGACCA






GCTGCCGGCGGTGCCGTCC






ATACAAGAGGACTGGACTT






CGCGTCCGATAAACCTACTA






CCACTCCAGCCCCAAGGCCC






CCAACCCCAGCACCGACTAT






CGCATCACAGCCTTTGTCAC






TGCGTCCTGAAGCCAGCCG






GCCAGCTGCAGGGGGGGC






CGTCCACACAAGGGGACTC






GACTTTGCGAGTGATAAGC






CCACCACCACCCCTGCCCCT






AGACCTCCAACCCCAGCCCC






TACAATCGCCAGCCAGCCCC






TGAGCCTGAGGCCCGAAGC






CTGTAGACCTGCCGCTGGC






GGAGCCGTGCACACCAGAG






GCCTGGATTTCGCCTGCGA






CATCTACATCTGGGCCCCTC






TGGCCGGCACCTGTGGCGT






GCTGCTGCTGAGCCTGGTC






ATCACCCTGTACTGCAACCA






CCGGAATAGGAGCAAGCG






GAGCAGAGGCGGCCACAG






CGACTACATGAACATGACC






CCCCGGAGGCCTGGCCCCA






CCCGGAAGCACTACCAGCC






CTACGCCCCTCCCAGGGACT






TCGCCGCCTACCGGAGCCG






GGTGAAGTTCAGCCGGAGC






GCCGACGCCCCTGCCTACCA






GCAGGGCCAGAACCAGCTG






TACAACGAGCTGAACCTGG






GCCGGAGGGAGGAGTACG






ACGTGCTGGACAAGCGGAG






AGGCCGGGACCCTGAGATG






GGCGGCAAGCCCCGGAGA






AAGAACCCTCAGGAGGGCC






TGTATAACGAACTGCAGAA






AGACAAGATGGCCGAGGCC






TACAGCGAGATCGGCATGA






AGGGCGAGCGGCGGAGGG






GCAAGGGCCACGACGGCCT






GTACCAGGGCCTGAGCACC






GCCACCAAGGATACCTACG






ACGCCCTGCACATGCAGGC






CCTGCCCCCCAGA





hROR1
615
EVQLVESGGGLVQPGGSLRLSCAT
616
GAAGTGCAGCTGGTGGAGT


(VH-VL)_

SGFTFSSYAMSWMRQAPGKGLE

CTGGCGGCGGTCTGGTGCA


14.

WVASISRGGTTYYADSVKGRFTISV

GCCCGGCGGCTCTCTGCGC


CD8a(3x).

DKSKNTLYLQMNSLRAEDTAVYYC

CTCTCCTGTGCCACCTCTGG


CD28z

GRYDYDGYYAMDYWGQGTLVTVS

TTTTACATTCTCCTCCTACGC




SGGGGSGGGGSGGGGSDIQMTQ

TATGTCCTGGATGCGGCAA




SPSSLSASVGDRVTITCKASPDINSY

GCCCCCGGCAAGGGCCTAG




LNWYQQKPGKAPKLLIYRANRLVD

AGTGGGTCGCCTCAATCAG




GVPSRFSGSGSGTDYTLTISSLQPED

CAGGGGCGGGACGACTTAT




FATYYCLQYDEFPYTFGAGTKVEIKK

TATGCCGATTCAGTTAAGG




PTTTPAPRPPTPAPTIASQPLSLRPE

GGAGATTCACAATTTCCGT




ASRPAAGGAVHTRGLDFASDKPTT

GGATAAATCCAAGAATACC




TPAPRPPTPAPTIASQPLSLRPEASR

TTATACCTCCAGATGAACTC




PAAGGAVHTRGLDFASDKPTTTPA

TCTGCGGGCCGAAGATACG




PRPPTPAPTIASQPLSLRPEACRPAA

GCCGTATATTATTGTGGGA




GGAVHTRGLDFACDIYIWAPLAGT

GGTATGACTACGACGGATA




CGVLLLSLVITLYCNHRNRSKRSRG

TTACGCCATGGATTATTGG




GHSDYMNMTPRRPGPTRKHYQPY

GGGCAGGGGACACTTGTTA




APPRDFAAYRSRVKFSRSADAPAY

CAGTGAGTTCCGGTGGTGG




QQGQNQLYNELNLGRREEYDVLD

GGGGTCTGGAGGCGGGGG




KRRGRDPEMGGKPRRKNPQEGLY

CAGTGGAGGCGGAGGGTC




NELQKDKMAEAYSEIGMKGERRR

TGATATACAGATGACACAG




GKGHDGLYQGLSTATKDTYDALH

AGCCCTTCAAGTTTATCTGC




MQALPPR

AAGCGTCGGCGATCGTGTT






ACAATAACTTGCAAGGCAT






CTCCCGACATCAATTCCTAC






CTCAACTGGTATCAGCAGA






AGCCTGGGAAGGCTCCTAA






GCTGCTTATTTACAGAGCAA






ATCGCCTGGTGGACGGCGT






GCCCAGTCGGTTTTCCGGG






TCTGGGAGCGGAACGGATT






ACACACTGACCATCTCAAGC






CTGCAACCCGAAGACTTCG






CTACATATTACTGCCTTCAG






TATGATGAGTTCCCATATAC






CTTCGGCGCTGGGACCAAG






GTGGAGATAAAGAAGCCTA






CCACCACCCCCGCACCTCGT






CCTCCAACCCCTGCACCTAC






GATTGCCAGTCAGCCTCTTT






CACTGCGGCCTGAGGCCAG






CAGACCAGCTGCCGGCGGT






GCCGTCCATACAAGAGGAC






TGGACTTCGCGTCCGATAA






ACCTACTACCACTCCAGCCC






CAAGGCCCCCAACCCCAGC






ACCGACTATCGCATCACAGC






CTTTGTCACTGCGTCCTGAA






GCCAGCCGGCCAGCTGCAG






GGGGGGCCGTCCACACAAG






GGGACTCGACTTTGCGAGT






GATAAGCCCACCACCACCCC






TGCCCCTAGACCTCCAACCC






CAGCCCCTACAATCGCCAGC






CAGCCCCTGAGCCTGAGGC






CCGAAGCCTGTAGACCTGC






CGCTGGCGGAGCCGTGCAC






ACCAGAGGCCTGGATTTCG






CCTGCGACATCTACATCTGG






GCCCCTCTGGCCGGCACCT






GTGGCGTGCTGCTGCTGAG






CCTGGTCATCACCCTGTACT






GCAACCACCGGAATAGGAG






CAAGCGGAGCAGAGGCGG






CCACAGCGACTACATGAAC






ATGACCCCCCGGAGGCCTG






GCCCCACCCGGAAGCACTA






CCAGCCCTACGCCCCTCCCA






GGGACTTCGCCGCCTACCG






GAGCCGGGTGAAGTTCAGC






CGGAGCGCCGACGCCCCTG






CCTACCAGCAGGGCCAGAA






CCAGCTGTACAACGAGCTG






AACCTGGGCCGGAGGGAG






GAGTACGACGTGCTGGACA






AGCGGAGAGGCCGGGACC






CTGAGATGGGCGGCAAGCC






CCGGAGAAAGAACCCTCAG






GAGGGCCTGTATAACGAAC






TGCAGAAAGACAAGATGGC






CGAGGCCTACAGCGAGATC






GGCATGAAGGGCGAGCGG






CGGAGGGGCAAGGGCCAC






GACGGCCTGTACCAGGGCC






TGAGCACCGCCACCAAGGA






TACCTACGACGCCCTGCACA






TGCAGGCCCTGCCCCCCAG






A





hROR1
617
DIQMTQSPSSLSASVGDRVTITCKA
618
GATATTCAGATGACCCAGTC


(VL-VH)_

SPDINSYLSWYQQKPGKAPKLLIYR

ACCTTCGAGTCTGAGCGCA


05.

ANRLVDGVPSRFSGSGSGTDFTLTI

TCCGTGGGCGACAGAGTGA


CD8a(3x).

SSLQPEDIATYYCLQYDEFPYTFGQ

CCATTACCTGTAAGGCCAGC


CD28z

GTKLEIKGGGGSGGGGSGGGGSEV

CCGGACATTAACAGCTACCT




QLVESGGGLVQPGGSLRLSCAASG

ATCGTGGTATCAGCAAAAG




FTFSSYAMSWVRQAPGKGLEWVS

CCTGGTAAGGCCCCTAAACT




SISRGGTTYYPDSVKGRFTISRDNSK

CCTTATCTACAGGGCTAATA




NTLYLOMNSLRAEDTAVYYCGRYD

GGTTGGTAGACGGGGTGCC




TAGCCGGTTCTCTGGTTCCG

GCAGCGGTACGGACTTTAC




YDGYYAMDYWGQGTLVTVSSKPT

TCTGACCATAAGCTCTCTGC




TTPAPRPPTPAPTIASQPLSLRPEAS

AACCAGAAGACATCGCAAC




RPAAGGAVHTRGLDFASDKPTTTP

ATACTACTGTTTACAATACG




APRPPTPAPTIASQPLSLRPEASRPA

ACGAATTTCCTTATACCTTT




AGGAVHTRGLDFASDKPTTTPAPR

GGCCAGGGGACCAAGTTAG




PPTPAPTIASQPLSLRPEACRPAAG

AGATCAAGGGGGGCGGCG




GAVHTRGLDFACDIYIWAPLAGTC

GAAGTGGTGGAGGGGGAA




GVLLLSLVITLYCNHRNRSKRSRGG

GTGGTGGAGGAGGAAGCG




HSDYMNMTPRRPGPTRKHYQPYA

AAGTGCAACTGGTCGAGTC




PPRDFAAYRSRVKFSRSADAPAYQ

TGGGGGCGGCCTTGTGCAA




QGQNQLYNELNLGRREEYDVLDKR

CCTGGAGGCAGCCTTCGAC




RGRDPEMGGKPRRKNPQEGLYNE

TCAGTTGCGCCGCGTCTGG




LQKDKMAEAYSEIGMKGERRRGK

TTTTACCTTCTCCTCTTACGC




GHDGLYQGLSTATKDTYDALHMQ

GATGAGCTGGGTTCGCCAG




ALPPR

GCCCCCGGCAAGGGACTTG






AGTGGGTTAGTTCGATCTCC






CGCGGAGGCACCACATATT






ATCCTGACTCGGTTAAGGG






ACGCTTCACTATCTCTAGGG






ACAATTCAAAGAACACACT






GTATCTCCAAATGAACTCCT






TGCGGGCCGAGGACACTGC






TGTGTATTATTGCGGACGAT






ACGACTACGATGGGTATTA






CGCCATGGATTACTGGGGG






CAAGGTACACTGGTCACTG






TGAGTTCGAAGCCTACCACC






ACCCCCGCACCTCGTCCTCC






AACCCCTGCACCTACGATTG






CCAGTCAGCCTCTTTCACTG






CGGCCTGAGGCCAGCAGAC






CAGCTGCCGGCGGTGCCGT






CCATACAAGAGGACTGGAC






TTCGCGTCCGATAAACCTAC






TACCACTCCAGCCCCAAGGC






CCCCAACCCCAGCACCGACT






ATCGCATCACAGCCTTTGTC






ACTGCGTCCTGAAGCCAGC






CGGCCAGCTGCAGGGGGG






GCCGTCCACACAAGGGGAC






TCGACTTTGCGAGTGATAA






GCCCACCACCACCCCTGCCC






CTAGACCTCCAACCCCAGCC






CCTACAATCGCCAGCCAGCC






CCTGAGCCTGAGGCCCGAA






GCCTGTAGACCTGCCGCTG






GCGGAGCCGTGCACACCAG






AGGCCTGGATTTCGCCTGC






GACATCTACATCTGGGCCCC






TCTGGCCGGCACCTGTGGC






GTGCTGCTGCTGAGCCTGG






TCATCACCCTGTACTGCAAC






CACCGGAATAGGAGCAAGC






GGAGCAGAGGCGGCCACA






GCGACTACATGAACATGAC






CCCCCGGAGGCCTGGCCCC






ACCCGGAAGCACTACCAGC






CCTACGCCCCTCCCAGGGAC






TTCGCCGCCTACCGGAGCC






GGGTGAAGTTCAGCCGGAG






CGCCGACGCCCCTGCCTACC






AGCAGGGCCAGAACCAGCT






GTACAACGAGCTGAACCTG






GGCCGGAGGGAGGAGTAC






GACGTGCTGGACAAGCGGA






GAGGCCGGGACCCTGAGAT






GGGCGGCAAGCCCCGGAG






AAAGAACCCTCAGGAGGGC






CTGTATAACGAACTGCAGA






AAGACAAGATGGCCGAGGC






CTACAGCGAGATCGGCATG






AAGGGCGAGCGGCGGAGG






GGCAAGGGCCACGACGGCC






TGTACCAGGGCCTGAGCAC






CGCCACCAAGGATACCTAC






GACGCCCTGCACATGCAGG






CCCTGCCCCCCAGATGA





hROR1
619
EVQLVESGGGLVQPGGSLRLSCAA
620
GAGGTTCAACTTGTGGAAT


(VH-VL)_

SGFTFSSYAMSWVRQAPGKGLEW

CCGGCGGCGGGTTAGTCCA


14-3.

VASISRGGTTYYADSVKGRFTISRD

GCCCGGCGGGAGCTTGCGG


CD8a(3x).

NSKNTLYLQMNSLRAEDTAVYYCG

CTGTCCTGCGCCGCCTCTGG


CD28z

RYDYDGYYAMDYWGQGTLVTVSS

ATTCACTTTTAGCTCCTATG




GGGGSGGGGSGGGGSDIQMTQS

CTATGTCTTGGGTAAGGCA




PSSLSASVGDRVTITCKASPDINSYL

GGCCCCTGGTAAAGGACTA




NWYQQKPGKAPKLLIYRANRLVDG

GAGTGGGTGGCCTCGATCT




VPSRFSGSGSGTDFTLTISSLQPEDI

CCCGTGGTGGCACTACATA




ATYYCLQYDEFPYTFGGGTKVEIKK

CTACGCCGACTCCGTTAAAG




PTTTPAPRPPTPAPTIASQPLSLRPE

GCCGGTTTACCATCTCCCGT




ASRPAAGGAVHTRGLDFASDKPTT

GACAACTCTAAAAATACTTT




TPAPRPPTPAPTIASQPLSLRPEASR

GTACCTGCAAATGAACTCCC




PAAGGAVHTRGLDFASDKPTTTPA

TGCGGGCAGAAGACACAGC




PRPPTPAPTIASQPLSLRPEACRPAA

CGTGTACTATTGCGGGCGT




GGAVHTRGLDFACDIYIWAPLAGT

TACGATTACGACGGATATTA




CGVLLLSLVITLYCNHRNRSKRSRG

CGCAATGGACTACTGGGGC




GHSDYMNMTPRRPGPTRKHYQPY

CAGGGCACACTGGTCACCG




APPRDFAAYRSRVKFSRSADAPAY

TGAGCAGCGGGGGCGGAG




QQGQNQLYNELNLGRREEYDVLD

GAAGTGGAGGAGGCGGTA




KRRGRDPEMGGKPRRKNPQEGLY

GTGGTGGGGGAGGAAGCG




NELQKDKMAEAYSEIGMKGERRR

ATATACAAATGACTCAGTCC




GKGHDGLYQGLSTATKDTYDALH

CCTAGTAGCCTTAGTGCTAG




MQALPPR

TGTGGGAGACAGAGTGACC






ATCACCTGCAAAGCATCTCC






TGATATCAATTCCTACCTTA






ACTGGTATCAACAGAAGCC






TGGCAAAGCTCCAAAGCTC






CTGATTTATCGCGCGAACA






GATTGGTCGATGGGGTCCC






TTCCAGATTCAGCGGCTCA






GGGTCAGGGACCGATTTCA






CCCTCACAATTAGTTCACTT






CAGCCCGAGGACATCGCCA






CGTATTATTGCCTTCAGTAC






GATGAGTTCCCTTACACCTT






TGGCGGGGGAACTAAAGTC






GAAATTAAGAAGCCTACCA






CCACCCCCGCACCTCGTCCT






CCAACCCCTGCACCTACGAT






TGCCAGTCAGCCTCTTTCAC






TGCGGCCTGAGGCCAGCAG






ACCAGCTGCCGGCGGTGCC






GTCCATACAAGAGGACTGG






ACTTCGCGTCCGATAAACCT






ACTACCACTCCAGCCCCAAG






GCCCCCAACCCCAGCACCG






ACTATCGCATCACAGCCTTT






GTCACTGCGTCCTGAAGCC






AGCCGGCCAGCTGCAGGG






GGGGCCGTCCACACAAGGG






GACTCGACTTTGCGAGTGA






TAAGCCCACCACCACCCCTG






CCCCTAGACCTCCAACCCCA






GCCCCTACAATCGCCAGCCA






GCCCCTGAGCCTGAGGCCC






GAAGCCTGTAGACCTGCCG






CTGGCGGAGCCGTGCACAC






CAGAGGCCTGGATTTCGCC






TGCGACATCTACATCTGGG






CCCCTCTGGCCGGCACCTGT






GGCGTGCTGCTGCTGAGCC






TGGTCATCACCCTGTACTGC






AACCACCGGAATAGGAGCA






AGCGGAGCAGAGGCGGCC






ACAGCGACTACATGAACAT






GACCCCCCGGAGGCCTGGC






CCCACCCGGAAGCACTACC






AGCCCTACGCCCCTCCCAGG






GACTTCGCCGCCTACCGGA






GCCGGGTGAAGTTCAGCCG






GAGCGCCGACGCCCCTGCC






TACCAGCAGGGCCAGAACC






AGCTGTACAACGAGCTGAA






CCTGGGCCGGAGGGAGGA






GTACGACGTGCTGGACAAG






CGGAGAGGCCGGGACCCT






GAGATGGGCGGCAAGCCCC






GGAGAAAGAACCCTCAGGA






GGGCCTGTATAACGAACTG






CAGAAAGACAAGATGGCCG






AGGCCTACAGCGAGATCGG






CATGAAGGGCGAGCGGCG






GAGGGGCAAGGGCCACGA






CGGCCTGTACCAGGGCCTG






AGCACCGCCACCAAGGATA






CCTACGACGCCCTGCACATG






CAGGCCCTGCCCCCCAGA





hROR1
621
EVQLVESGGGLVQPGGSLRLSCAA
622
GAAGTGCAGCTTGTGGAGT


(VH-VL)_

SGFTFSSYAMSWVRQAPGKGLEW

CAGGAGGAGGGCTAGTTCA


14-4.

VASISRGGTTYYPDSVKGRFTISRD

GCCAGGCGGCTCTCTGAGA


CD8a(3x).

NVRNILYLQMSSLRSEDTAMYYCG

CTATCTTGTGCTGCCTCCGG


CD28z

RYDYDGYYAMDYWGQGTLVTVSS

CTTCACATTTAGCTCTTATG




GGGGSGGGGSGGGGSDIQMTQS

CAATGTCCTGGGTCCGCCA




PSSLSASVGDRVTITCKASPDINSYL

GGCCCCTGGTAAAGGCCTG




NWYQQKPGKAPKLLIYRANRLVDG

GAATGGGTTGCTTCTATCTC




VPSRFSGSGSGTDYTLTISSLQPEDF

TAGAGGCGGAACCACTTAC




ATYYCLQYDEFPYTFGAGTKVEIKK

TACCCTGATTCAGTGAAGG




PTTTPAPRPPTPAPTIASQPLSLRPE

GGAGATTCACAATTAGTAG




ASRPAAGGAVHTRGLDFASDKPTT

GGACAACGTGCGGAACATC




TPAPRPPTPAPTIASQPLSLRPEASR

CTCTACCTACAGATGTCAAG




PAAGGAVHTRGLDFASDKPTTTPA

TTTACGCAGTGAGGACACT




PRPPTPAPTIASQPLSLRPEACRPAA

GCGATGTATTACTGCGGTC




GGAVHTRGLDFACDIYIWAPLAGT

GATACGATTATGATGGATA




CGVLLLSLVITLYCNHRNRSKRSRG

TTATGCAATGGATTATTGG




GHSDYMNMTPRRPGPTRKHYQPY

GGCCAGGGCACTCTGGTCA




APPRDFAAYRSRVKFSRSADAPAY

CAGTATCTTCCGGCGGCGG




QQGQNQLYNELNLGRREEYDVLD

TGGTTCTGGCGGTGGTGGA




KRRGRDPEMGGKPRRKNPQEGLY

AGCGGAGGGGGGGGGTCC




NELQKDKMAEAYSEIGMKGERRR

GACATCCAGATGACCCAAT




GKGHDGLYQGLSTATKDTYDALH

CACCATCGAGTCTTAGTGCA




MQALPPR

TCCGTTGGGGATAGAGTGA






CAATCACTTGTAAGGCATCC






CCGGACATCAACTCATATCT






TAATTGGTATCAGCAAAAG






CCGGGCAAGGCCCCTAAGC






TCCTGATTTATAGGGCCAAC






CGCCTTGTGGATGGAGTCC






CCTCCCGCTTTAGTGGAAGC






GGCTCTGGCACAGACTACA






CCCTGACTATCAGCTCCTTG






CAGCCTGAGGATTTTGCTAC






CTACTACTGTCTTCAGTACG






ATGAATTTCCATACACTTTC






GGTGCTGGGACAAAAGTGG






AGATCAAAAAGCCTACCAC






CACCCCCGCACCTCGTCCTC






CAACCCCTGCACCTACGATT






GCCAGTCAGCCTCTTTCACT






GCGGCCTGAGGCCAGCAGA






CCAGCTGCCGGCGGTGCCG






TCCATACAAGAGGACTGGA






CTTCGCGTCCGATAAACCTA






CTACCACTCCAGCCCCAAGG






CCCCCAACCCCAGCACCGAC






TATCGCATCACAGCCTTTGT






CACTGCGTCCTGAAGCCAG






CCGGCCAGCTGCAGGGGG






GGCCGTCCACACAAGGGGA






CTCGACTTTGCGAGTGATA






AGCCCACCACCACCCCTGCC






CCTAGACCTCCAACCCCAGC






CCCTACAATCGCCAGCCAGC






CCCTGAGCCTGAGGCCCGA






AGCCTGTAGACCTGCCGCT






GGCGGAGCCGTGCACACCA






GAGGCCTGGATTTCGCCTG






CGACATCTACATCTGGGCCC






CTCTGGCCGGCACCTGTGG






CGTGCTGCTGCTGAGCCTG






GTCATCACCCTGTACTGCAA






CCACCGGAATAGGAGCAAG






CGGAGCAGAGGCGGCCAC






AGCGACTACATGAACATGA






CCCCCCGGAGGCCTGGCCC






CACCCGGAAGCACTACCAG






CCCTACGCCCCTCCCAGGGA






CTTCGCCGCCTACCGGAGC






CGGGTGAAGTTCAGCCGGA






GCGCCGACGCCCCTGCCTA






CCAGCAGGGCCAGAACCAG






CTGTACAACGAGCTGAACC






TGGGCCGGAGGGAGGAGT






ACGACGTGCTGGACAAGCG






GAGAGGCCGGGACCCTGA






GATGGGCGGCAAGCCCCG






GAGAAAGAACCCTCAGGAG






GGCCTGTATAACGAACTGC






AGAAAGACAAGATGGCCGA






GGCCTACAGCGAGATCGGC






ATGAAGGGCGAGCGGCGG






AGGGGCAAGGGCCACGAC






GGCCTGTACCAGGGCCTGA






GCACCGCCACCAAGGATAC






CTACGACGCCCTGCACATGC






AGGCCCTGCCCCCCAGA





hROR1
623
EVQLVESGGGLVQPGGSLRLSCAA
624
GAAGTGCAACTGGTCGAGT


(VH_5-VL_14).

SGFTFSSYAMSWVRQAPGKGLEW

CTGGGGGCGGCCTTGTGCA


CD8a(3x).

VSSISRGGTTYYPDSVKGRFTISRDN

ACCTGGAGGCAGCCTTCGA


CD28z

SKNTLYLQMNSLRAEDTAVYYCGR

CTCAGTTGCGCCGCGTCTG




YDYDGYYAMDYWGQGTLVTVSSG

GTTTTACCTTCTCCTCTTACG




GGGSGGGGSGGGGSDIQMTQSPS

CGATGAGCTGGGTTCGCCA




SLSASVGDRVTITCKASPDINSYLN

GGCCCCCGGCAAGGGACTT




WYQQKPGKAPKLLIYRANRLVDGV

GAGTGGGTTAGTTCGATCT




PSRFSGSGSGTDYTLTISSLQPEDFA

CCCGCGGAGGCACCACATA




TYYCLQYDEFPYTFGAGTKVEIKKPT

TTATCCTGACTCGGTTAAGG




TTPAPRPPTPAPTIASQPLSLRPEAS

GACGCTTCACTATCTCTAGG




RPAAGGAVHTRGLDFASDKPTTTP

GACAATTCAAAGAACACAC




APRPPTPAPTIASQPLSLRPEASRPA

TGTATCTCCAAATGAACTCC




AGGAVHTRGLDFASDKPTTTPAPR

TTGCGGGCCGAGGACACTG




PPTPAPTIASQPLSLRPEACRPAAG

CTGTGTATTATTGCGGACG




GAVHTRGLDFACDIYIWAPLAGTC

ATACGACTACGATGGGTAT




GVLLLSLVITLYCNHRNRSKRSRGG

TACGCCATGGATTACTGGG




HSDYMNMTPRRPGPTRKHYQPYA

GGCAAGGTACACTGGTCAC




PPRDFAAYRSRVKFSRSADAPAYQ

TGTGAGTTCGGGGGGCGGC




QGQNQLYNELNLGRREEYDVLDKR

GGAAGTGGTGGAGGGGGA




RGRDPEMGGKPRRKNPQEGLYNE

AGTGGTGGAGGAGGAAGC




LQKDKMAEAYSEIGMKGERRRGK

GATATACAGATGACACAGA




GHDGLYQGLSTATKDTYDALHMQ

GCCCTTCAAGTTTATCTGCA




ALPPR

AGCGTCGGCGATCGTGTTA






CAATAACTTGCAAGGCATCT






CCCGACATCAATTCCTACCT






CAACTGGTATCAGCAGAAG






CCTGGGAAGGCTCCTAAGC






TGCTTATTTACAGAGCAAAT






CGCCTGGTGGACGGCGTGC






CCAGTCGGTTTTCCGGGTCT






GGGAGCGGAACGGATTACA






CACTGACCATCTCAAGCCTG






CAACCCGAAGACTTCGCTAC






ATATTACTGCCTTCAGTATG






ATGAGTTCCCATATACCTTC






GGCGCTGGGACCAAGGTG






GAGATAAAGAAGCCTACCA






CCACCCCCGCACCTCGTCCT






CCAACCCCTGCACCTACGAT






TGCCAGTCAGCCTCTTTCAC






TGCGGCCTGAGGCCAGCAG






ACCAGCTGCCGGCGGTGCC






GTCCATACAAGAGGACTGG






ACTTCGCGTCCGATAAACCT






ACTACCACTCCAGCCCCAAG






GCCCCCAACCCCAGCACCG






ACTATCGCATCACAGCCTTT






GTCACTGCGTCCTGAAGCC






AGCCGGCCAGCTGCAGGG






GGGGCCGTCCACACAAGGG






GACTCGACTTTGCGAGTGA






TAAGCCCACCACCACCCCTG






CCCCTAGACCTCCAACCCCA






GCCCCTACAATCGCCAGCCA






GCCCCTGAGCCTGAGGCCC






GAAGCCTGTAGACCTGCCG






CTGGCGGAGCCGTGCACAC






CAGAGGCCTGGATTTCGCC






TGCGACATCTACATCTGGG






CCCCTCTGGCCGGCACCTGT






GGCGTGCTGCTGCTGAGCC






TGGTCATCACCCTGTACTGC






AACCACCGGAATAGGAGCA






AGCGGAGCAGAGGCGGCC






ACAGCGACTACATGAACAT






GACCCCCCGGAGGCCTGGC






CCCACCCGGAAGCACTACC






AGCCCTACGCCCCTCCCAGG






GACTTCGCCGCCTACCGGA






GCCGGGTGAAGTTCAGCCG






GAGCGCCGACGCCCCTGCC






TACCAGCAGGGCCAGAACC






AGCTGTACAACGAGCTGAA






CCTGGGCCGGAGGGAGGA






GTACGACGTGCTGGACAAG






CGGAGAGGCCGGGACCCT






GAGATGGGCGGCAAGCCCC






GGAGAAAGAACCCTCAGGA






GGGCCTGTATAACGAACTG






CAGAAAGACAAGATGGCCG






AGGCCTACAGCGAGATCGG






CATGAAGGGCGAGCGGCG






GAGGGGCAAGGGCCACGA






CGGCCTGTACCAGGGCCTG






AGCACCGCCACCAAGGATA






CCTACGACGCCCTGCACATG






CAGGCCCTGCCCCCCAGA





hROR1
625
EVQLVESGGGLVQPGGSLRLSCAA
626
GAAGTGCAACTGGTCGAGT


(VH_5-VL_16).

SGFTFSSYAMSWVRQAPGKGLEW

CTGGGGGCGGCCTTGTGCA


CD8a(3x).

VSSISRGGTTYYPDSVKGRFTISRDN

ACCTGGAGGCAGCCTTCGA


CD28z

SKNTLYLQMNSLRAEDTAVYYCGR

CTCAGTTGCGCCGCGTCTG




YDYDGYYAMDYWGQGTLVTVSSG

GTTTTACCTTCTCCTCTTACG




GGGSGGGGSGGGGSDIQMTQSPS

CGATGAGCTGGGTTCGCCA




SLSASVGDRVTITCKASPDINSYLN

GGCCCCCGGCAAGGGACTT




WYQQKPGKAPKVLIYRANRLVDG

GAGTGGGTTAGTTCGATCT




VPSRFSGSGSGTDYTLTISSLQPEDF

CCCGCGGAGGCACCACATA




ATYYCLQYDEFPYTFGQGTKVEIKK

TTATCCTGACTCGGTTAAGG




PTTTPAPRPPTPAPTIASQPLSLRPE

GACGCTTCACTATCTCTAGG




ASRPAAGGAVHTRGLDFASDKPTT

GACAATTCAAAGAACACAC




TPAPRPPTPAPTIASQPLSLRPEASR

TGTATCTCCAAATGAACTCC




PAAGGAVHTRGLDFASDKPTTTPA

TTGCGGGCCGAGGACACTG




PRPPTPAPTIASQPLSLRPEACRPAA

CTGTGTATTATTGCGGACG




GGAVHTRGLDFACDIYIWAPLAGT

ATACGACTACGATGGGTAT




CGVLLLSLVITLYCNHRNRSKRSRG

TACGCCATGGATTACTGGG




GHSDYMNMTPRRPGPTRKHYQPY

GGCAAGGTACACTGGTCAC




APPRDFAAYRSRVKFSRSADAPAY

TGTGAGTTCGGGGGGCGGC




QQGQNQLYNELNLGRREEYDVLD

GGAAGTGGTGGAGGGGGA




KRRGRDPEMGGKPRRKNPQEGLY

AGTGGTGGAGGAGGAAGC




NELQKDKMAEAYSEIGMKGERRR

GATATTCAGATGACCCAGTC




GKGHDGLYQGLSTATKDTYDALH

GCCCAGCAGTCTCTCGGCCT




MQALPPR

CAGTGGGCGACCGGGTCAC






TATCACTTGCAAAGCAAGTC






CTGATATAAACTCCTATCTT






AATTGGTATCAGCAGAAGC






CCGGCAAGGCACCTAAGGT






TCTGATATATCGCGCAAATC






GGCTCGTGGATGGAGTACC






CAGCCGATTTTCCGGCAGC






GGCTCAGGCACTGACTACA






CACTGACAATCAGCAGCTT






GCAGCCTGAAGATTTCGCC






ACATACTATTGTCTACAGTA






CGACGAGTTCCCTTATACAT






TCGGCCAGGGGACCAAGGT






CGAGATCAAGAAGCCTACC






ACCACCCCCGCACCTCGTCC






TCCAACCCCTGCACCTACGA






TTGCCAGTCAGCCTCTTTCA






CTGCGGCCTGAGGCCAGCA






GACCAGCTGCCGGCGGTGC






CGTCCATACAAGAGGACTG






GACTTCGCGTCCGATAAACC






TACTACCACTCCAGCCCCAA






GGCCCCCAACCCCAGCACC






GACTATCGCATCACAGCCTT






TGTCACTGCGTCCTGAAGCC






AGCCGGCCAGCTGCAGGG






GGGGCCGTCCACACAAGGG






GACTCGACTTTGCGAGTGA






TAAGCCCACCACCACCCCTG






CCCCTAGACCTCCAACCCCA






GCCCCTACAATCGCCAGCCA






GCCCCTGAGCCTGAGGCCC






GAAGCCTGTAGACCTGCCG






CTGGCGGAGCCGTGCACAC






CAGAGGCCTGGATTTCGCC






TGCGACATCTACATCTGGG






CCCCTCTGGCCGGCACCTGT






GGCGTGCTGCTGCTGAGCC






TGGTCATCACCCTGTACTGC






AACCACCGGAATAGGAGCA






AGCGGAGCAGAGGCGGCC






ACAGCGACTACATGAACAT






GACCCCCCGGAGGCCTGGC






CCCACCCGGAAGCACTACC






AGCCCTACGCCCCTCCCAGG






GACTTCGCCGCCTACCGGA






GCCGGGTGAAGTTCAGCCG






GAGCGCCGACGCCCCTGCC






TACCAGCAGGGCCAGAACC






AGCTGTACAACGAGCTGAA






CCTGGGCCGGAGGGAGGA






GTACGACGTGCTGGACAAG






CGGAGAGGCCGGGACCCT






GAGATGGGCGGCAAGCCCC






GGAGAAAGAACCCTCAGGA






GGGCCTGTATAACGAACTG






CAGAAAGACAAGATGGCCG






AGGCCTACAGCGAGATCGG






CATGAAGGGCGAGCGGCG






GAGGGGCAAGGGCCACGA






CGGCCTGTACCAGGGCCTG






AGCACCGCCACCAAGGATA






CCTACGACGCCCTGCACATG






CAGGCCCTGCCCCCCAGA





hROR1
627
EVQLVESGGGLVQPGGSLRLSCSAS
628
GAGGTTCAACTCGTGGAGT


(VH_18-

GFTFSSYAMSWVRQVPGKGLVWI

CTGGAGGCGGGCTAGTGCA


VL_04).

SSISRGGTTYYADSVRGRFIISRDNA

GCCTGGCGGCTCCCTGCGA


CD8a(3x).

KNTLYLEMNNLRGEDTAVYYCARY

CTGTCTTGCAGCGCATCAG


CD28z

DYDGYYAMDYWGQGTLVTVSSG

GCTTTACATTCAGTTCTTAT




GGGSGGGGSGGGGSDIQMTQSPS

GCCATGAGCTGGGTGAGGC




SLSASVGDRVTITCQASPDINSYLN

AGGTGCCCGGCAAGGGTCT




WYQQKPGKAPKLLIYRANNLETGV

GGTGTGGATCAGCTCAATC




PSRFSGSGSGTDFTLTISSLQPEDIA

TCCAGGGGCGGGACTACAT




TYYCLQYDEFPYTFGQGTKLEIKKPT

ATTACGCCGATTCGGTCAG




TTPAPRPPTPAPTIASQPLSLRPEAS

GGGTCGTTTTATCATTAGCA




RPAAGGAVHTRGLDFASDKPTTTP

GGGATAATGCCAAGAACAC




APRPPTPAPTIASQPLSLRPEASRPA

CTTGTATTTGGAGATGAAC




AGGAVHTRGLDFASDKPTTTPAPR

AACCTAAGAGGCGAAGACA




PPTPAPTIASQPLSLRPEACRPAAG

CCGCTGTGTACTATTGTGCC




GAVHTRGLDFACDIYIWAPLAGTC

CGTTACGACTACGATGGGT




GVLLLSLVITLYCNHRNRSKRSRGG

ACTACGCCATGGACTATTG




HSDYMNMTPRRPGPTRKHYQPYA

GGGCCAGGGAACCTTGGTG




PPRDFAAYRSRVKFSRSADAPAYQ

ACTGTGTCAAGTGGCGGGG




QGQNQLYNELNLGRREEYDVLDKR

GCGGCAGCGGAGGCGGTG




RGRDPEMGGKPRRKNPQEGLYNE

GCAGCGGAGGCGGCGGTT




LQKDKMAEAYSEIGMKGERRRGK

CTGATATTCAAATGACGCAA




GHDGLYQGLSTATKDTYDALHMQ

AGTCCCAGCAGCCTCTCCGC




ALPPR

CTCCGTTGGAGACAGGGTG






ACTATTACATGCCAAGCCAG






CCCCGATATTAATAGCTACT






TAAATTGGTATCAGCAGAA






ACCTGGGAAGGCACCTAAA






CTTCTCATCTACCGCGCTAA






CAATCTGGAGACCGGCGTG






CCGTCTAGATTTTCCGGCTC






TGGATCAGGGACCGATTTT






ACTCTGACAATTAGTTCCCT






GCAACCCGAAGACATCGCC






ACTTATTATTGCCTGCAATA






TGATGAGTTTCCTTACACAT






TTGGTCAGGGAACTAAACT






AGAGATTAAGAAGCCTACC






ACCACCCCCGCACCTCGTCC






TCCAACCCCTGCACCTACGA






TTGCCAGTCAGCCTCTTTCA






CTGCGGCCTGAGGCCAGCA






GACCAGCTGCCGGCGGTGC






CGTCCATACAAGAGGACTG






GACTTCGCGTCCGATAAACC






TACTACCACTCCAGCCCCAA






GGCCCCCAACCCCAGCACC






GACTATCGCATCACAGCCTT






TGTCACTGCGTCCTGAAGCC






AGCCGGCCAGCTGCAGGG






GGGGCCGTCCACACAAGGG






GACTCGACTTTGCGAGTGA






TAAGCCCACCACCACCCCTG






CCCCTAGACCTCCAACCCCA






GCCCCTACAATCGCCAGCCA






GCCCCTGAGCCTGAGGCCC






GAAGCCTGTAGACCTGCCG






CTGGCGGAGCCGTGCACAC






CAGAGGCCTGGATTTCGCC






TGCGACATCTACATCTGGG






CCCCTCTGGCCGGCACCTGT






GGCGTGCTGCTGCTGAGCC






TGGTCATCACCCTGTACTGC






AACCACCGGAATAGGAGCA






AGCGGAGCAGAGGCGGCC






ACAGCGACTACATGAACAT






GACCCCCCGGAGGCCTGGC






CCCACCCGGAAGCACTACC






AGCCCTACGCCCCTCCCAGG






GACTTCGCCGCCTACCGGA






GCCGGGTGAAGTTCAGCCG






GAGCGCCGACGCCCCTGCC






TACCAGCAGGGCCAGAACC






AGCTGTACAACGAGCTGAA






CCTGGGCCGGAGGGAGGA






GTACGACGTGCTGGACAAG






CGGAGAGGCCGGGACCCT






GAGATGGGCGGCAAGCCCC






GGAGAAAGAACCCTCAGGA






GGGCCTGTATAACGAACTG






CAGAAAGACAAGATGGCCG






AGGCCTACAGCGAGATCGG






CATGAAGGGCGAGCGGCG






GAGGGGCAAGGGCCACGA






CGGCCTGTACCAGGGCCTG






AGCACCGCCACCAAGGATA






CCTACGACGCCCTGCACATG






CAGGCCCTGCCCCCCAGA






GAGGTTCAACTCGTGGAGT






CTGGAGGCGGGCTAGTGCA






GCCTGGCGGCTCCCTGCGA






CTGTCTTGCAGCGCATCAG






GCTTTACATTCAGTTCTTAT






GCCATGAGCTGGGTGAGGC






AGGTGCCCGGCAAGGGTCT






GGTGTGGATCAGCTCAATC






TCCAGGGGCGGGACTACAT





hROR1
629
EVQLVESGGGLVQPGGSLRLSCSAS
630
ATTACGCCGATTCGGTCAG


(VH_18-

GFTFSSYAMSWVRQVPGKGLVWI

GGGTCGTTTTATCATTAGCA


VL_14).

SSISRGGTTYYADSVRGRFIISRDNA

GGGATAATGCCAAGAACAC


CD8a(3x).

KNTLYLEMNNLRGEDTAVYYCARY

CTTGTATTTGGAGATGAAC


CD28z

DYDGYYAMDYWGQGTLVTVSSG

AACCTAAGAGGCGAAGACA




GGGSGGGGSGGGGSDIQMTQSPS

CCGCTGTGTACTATTGTGCC




SLSASVGDRVTITCKASPDINSYLN

CGTTACGACTACGATGGGT




WYQQKPGKAPKLLIYRANRLVDGV

ACTACGCCATGGACTATTG




PSRFSGSGSGTDYTLTISSLQPEDFA

GGGCCAGGGAACCTTGGTG




TYYCLQYDEFPYTFGAGTKVEIKKPT

ACTGTGTCAAGTGGCGGGG




TTPAPRPPTPAPTIASQPLSLRPEAS

GCGGCAGCGGAGGCGGTG




RPAAGGAVHTRGLDFASDKPTTTP

GCAGCGGAGGCGGCGGTT




APRPPTPAPTIASQPLSLRPEASRPA

CTGATATACAGATGACACA




AGGAVHTRGLDFASDKPTTTPAPR

GAGCCCTTCAAGTTTATCTG




PPTPAPTIASQPLSLRPEACRPAAG

CAAGCGTCGGCGATCGTGT




GAVHTRGLDFACDIYIWAPLAGTC

TACAATAACTTGCAAGGCAT




GVLLLSLVITLYCNHRNRSKRSRGG

CTCCCGACATCAATTCCTAC




HSDYMNMTPRRPGPTRKHYQPYA

CTCAACTGGTATCAGCAGA




PPRDFAAYRSRVKFSRSADAPAYQ

AGCCTGGGAAGGCTCCTAA




QGQNQLYNELNLGRREEYDVLDKR

GCTGCTTATTTACAGAGCAA




RGRDPEMGGKPRRKNPQEGLYNE

ATCGCCTGGTGGACGGCGT




LQKDKMAEAYSEIGMKGERRRGK

GCCCAGTCGGTTTTCCGGG




GHDGLYQGLSTATKDTYDALHMQ

TCTGGGAGCGGAACGGATT




ALPPR

ACACACTGACCATCTCAAGC






CTGCAACCCGAAGACTTCG






CTACATATTACTGCCTTCAG






TATGATGAGTTCCCATATAC






CTTCGGCGCTGGGACCAAG






GTGGAGATAAAGAAGCCTA






CCACCACCCCCGCACCTCGT






CCTCCAACCCCTGCACCTAC






GATTGCCAGTCAGCCTCTTT






CACTGCGGCCTGAGGCCAG






CAGACCAGCTGCCGGCGGT






GCCGTCCATACAAGAGGAC






TGGACTTCGCGTCCGATAA






ACCTACTACCACTCCAGCCC






CAAGGCCCCCAACCCCAGC






ACCGACTATCGCATCACAGC






CTTTGTCACTGCGTCCTGAA






GCCAGCCGGCCAGCTGCAG






GGGGGGCCGTCCACACAAG






GGGACTCGACTTTGCGAGT






GATAAGCCCACCACCACCCC






TGCCCCTAGACCTCCAACCC






CAGCCCCTACAATCGCCAGC






CAGCCCCTGAGCCTGAGGC






CCGAAGCCTGTAGACCTGC






CGCTGGCGGAGCCGTGCAC






ACCAGAGGCCTGGATTTCG






CCTGCGACATCTACATCTGG






GCCCCTCTGGCCGGCACCT






GTGGCGTGCTGCTGCTGAG






CCTGGTCATCACCCTGTACT






GCAACCACCGGAATAGGAG






CAAGCGGAGCAGAGGCGG






CCACAGCGACTACATGAAC






ATGACCCCCCGGAGGCCTG






GCCCCACCCGGAAGCACTA






CCAGCCCTACGCCCCTCCCA






GGGACTTCGCCGCCTACCG






GAGCCGGGTGAAGTTCAGC






CGGAGCGCCGACGCCCCTG






CCTACCAGCAGGGCCAGAA






CCAGCTGTACAACGAGCTG






AACCTGGGCCGGAGGGAG






GAGTACGACGTGCTGGACA






AGCGGAGAGGCCGGGACC






CTGAGATGGGCGGCAAGCC






CCGGAGAAAGAACCCTCAG






GAGGGCCTGTATAACGAAC






TGCAGAAAGACAAGATGGC






CGAGGCCTACAGCGAGATC






GGCATGAAGGGCGAGCGG






CGGAGGGGCAAGGGCCAC






GACGGCCTGTACCAGGGCC






TGAGCACCGCCACCAAGGA






TACCTACGACGCCCTGCACA






TGCAGGCCCTGCCCCCCAG






A





IgG4-Fc 12






amino acid
631
ESKYGPPCPPCP




hinge region














RTS-COMPONENTS











VP16
632
GPKKKRKVAPPTDVSLGDELHLDG
633
GGCCCCAAGAAGAAAAGG


activation

EDVAMAHADALDDFDLDMLGDG

AAGGTGGCCCCCCCCACCG


domain

DSPGPGFTPHDSAPYGALDMADF

ACGTGAGCCTGGGCGACGA




EFEQMFTDALGIDEYGG

GCTGCACCTGGACGGCGAG






GACGTGGCCATGGCCCACG






CCGACGCCCTGGACGACTT






CGACCTGGACATGCTGGGC






GACGGCGACAGCCCCGGCC






CCGGCTTCACCCCCCACGAC






AGCGCCCCCTACGGCGCCC






TGGACATGGCCGACTTCGA






GTTCGAGCAGATGTTCACC






GACGCCCTGGGCATCGACG






AGTACGGCGGC





Retinoid x
634
EMPVDRILEAELAVEQKSDQGVEG
635
GAGATGCCCGTGGACAGGA


receptor

PGGTGGSGSSPNDPVTNICQAADK

TTCTGGAGGCCGAACTCGC


(RxR)

QLFTLVEWAKRIPHFSSLPLDDQVIL

CGTGGAGCAGAAAAGCGAC




LRAGWNELLIASFSHRSIDVRDGILL

CAGGGCGTGGAGGGCCCC




ATGLHVHRNSAHSAGVGAIFDRVL

GGCGGAACCGGCGGCAGC




TELVSKMRDMRMDKTELGCLRAII

GGCAGCAGCCCCAACGACC




LFNPEVRGLKSAQEVELLREKVYAA

CCGTGACCAACATCTGCCA




LEEYTRTTHPDEPGRFAKLLLRLPSL

GGCCGCCGACAAGCAGCTG




RSIGLKCLEHLFFFRLIGDVPIDTFLM

TTCACCCTGGTGGAGTGGG




EMLESPSDS

CCAAGAGGATTCCCCACTTC






AGCAGCCTGCCCCTGGACG






ACCAGGTGATCCTGCTGAG






GGCCGGATGGAACGAGCTG






CTGATCGCCAGCTTCAGCCA






CAGGAGCATCGACGTGAGG






GACGGCATCCTGCTGGCCA






CCGGCCTGCACGTCCATAG






GAACAGCGCCCACAGCGCC






GGAGTGGGCGCCATCTTCG






ACAGGGTGCTGACCGAGCT






GGTGAGCAAGATGAGGGA






CATGAGGATGGACAAGACC






GAGCTGGGCTGCCTGAGGG






CCATCATCCTGTTCAACCCC






GAGGTGAGGGGCCTGAAA






AGCGCCCAGGAGGTGGAG






CTGCTGAGGGAGAAGGTGT






ACGCCGCCCTGGAGGAGTA






CACCAGGACCACCCACCCC






GACGAGCCCGGCAGATTCG






CCAAGCTGCTGCTGAGGCT






GCCCAGCCTGAGGAGCATC






GGCCTGAAGTGCCTGGAGC






ACCTGTTCTTCTTCAGGCTG






ATCGGCGACGTGCCCATCG






ACACCTTCCTGATGGAGAT






GCTGGAGAGCCCCAGCGAC






AGC





VP16-linker-
636
GPKKKRKVAPPTDVSLGDELHLDG
637
GGCCCCAAGAAGAAAAGG


RxR

EDVAMAHADALDDFDLDMLGDG

AAGGTGGCCCCCCCCACCG




DSPGPGFTPHDSAPYGALDMADF

ACGTGAGCCTGGGCGACGA




EFEQMFTDALGIDEYGGEFEMPVD

GCTGCACCTGGACGGCGAG




RILEAELAVEQKSDQGVEGPGGTG

GACGTGGCCATGGCCCACG




GSGSSPNDPVTNICQAADKQLFTL

CCGACGCCCTGGACGACTT




VEWAKRIPHFSSLPLDDQVILLRAG

CGACCTGGACATGCTGGGC




WNELLIASFSHRSIDVRDGILLATGL

GACGGCGACAGCCCCGGCC




HVHRNSAHSAGVGAIFDRVLTELV

CCGGCTTCACCCCCCACGAC




SKMRDMRMDKTELGCLRAIILFNP

AGCGCCCCCTACGGCGCCC




EVRGLKSAQEVELLREKVYAALEEY

TGGACATGGCCGACTTCGA




TRTTHPDEPGRFAKLLLRLPSLRSIG

GTTCGAGCAGATGTTCACC




LKCLEHLFFFRLIGDVPIDTFLMEML

GACGCCCTGGGCATCGACG




ESPSDS

AGTACGGCGGCGAATTCGA






GATGCCCGTGGACAGGATT






CTGGAGGCCGAACTCGCCG






TGGAGCAGAAAAGCGACCA






GGGCGTGGAGGGCCCCGG






CGGAACCGGCGGCAGCGG






CAGCAGCCCCAACGACCCC






GTGACCAACATCTGCCAGG






CCGCCGACAAGCAGCTGTT






CACCCTGGTGGAGTGGGCC






AAGAGGATTCCCCACTTCA






GCAGCCTGCCCCTGGACGA






CCAGGTGATCCTGCTGAGG






GCCGGATGGAACGAGCTGC






TGATCGCCAGCTTCAGCCAC






AGGAGCATCGACGTGAGG






GACGGCATCCTGCTGGCCA






CCGGCCTGCACGTCCATAG






GAACAGCGCCCACAGCGCC






GGAGTGGGCGCCATCTTCG






ACAGGGTGCTGACCGAGCT






GGTGAGCAAGATGAGGGA






CATGAGGATGGACAAGACC






GAGCTGGGCTGCCTGAGGG






CCATCATCCTGTTCAACCCC






GAGGTGAGGGGCCTGAAA






AGCGCCCAGGAGGTGGAG






CTGCTGAGGGAGAAGGTGT






ACGCCGCCCTGGAGGAGTA






CACCAGGACCACCCACCCC






GACGAGCCCGGCAGATTCG






CCAAGCTGCTGCTGAGGCT






GCCCAGCCTGAGGAGCATC






GGCCTGAAGTGCCTGGAGC






ACCTGTTCTTCTTCAGGCTG






ATCGGCGACGTGCCCATCG






ACACCTTCCTGATGGAGAT






GCTGGAGAGCCCCAGCGAC






AGC





GAL4 DNA
638
MKLLSSIEQACDICRLKKLKCSKEKP
639
ATGAAGCTGCTGAGCAGCA


Binding

KCAKCLKNNWECRYSPKTKRSPLTR

TCGAGCAGGCTTGCGACAT


Domain

AHLTEVESRLERLEQLFLLIFPREDLD

CTGCAGGCTGAAGAAGCTG




MILKMDSLQDIKALLTGLFVQDNV

AAGTGCAGCAAGGAGAAG




NKDAVTDRLASVETDMPLTLRQHR

CCCAAGTGCGCCAAGTGCC




ISATSSSEESSNKGQRQLTVSPEF

TGAAGAACAACTGGGAGTG






CAGATACAGCCCCAAGACC






AAGAGGAGCCCCCTGACCA






GGGCCCACCTGACCGAGGT






GGAGAGCAGGCTGGAGAG






GCTGGAGCAGCTGTTCCTG






CTGATCTTCCCCAGGGAGG






ACCTGGACATGATCCTGAA






GATGGACAGCCTGCAAGAC






ATCAAGGCCCTGCTGACCG






GCCTGTTCGTGCAGGACAA






CGTGAACAAGGACGCCGTG






ACCGACAGGCTGGCCAGCG






TGGAGACCGACATGCCCCT






GACCCTGAGGCAGCACAGG






ATCAGCGCCACCAGCAGCA






GCGAGGAGAGCAGCAACA






AGGGCCAGAGGCAGCTGAC






CGTGAGCCCCGAGTTT





Ecdysone
640
IRPECVVPETQCAMKRKEKKAQKE
641
ATCAGGCCCGAGTGCGTGG


Receptor

KDKLPVSTTTVDDHMPPIMQCEPP

TGCCCGAGACCCAGTGCGC


Ligand

PPEAARIHEVVPRFLSDKLLVTNRQ

CATGAAAAGGAAGGAGAA


Binding

KNIPQLTANQQFLIARLIWYQDGYE

GAAGGCCCAGAAGGAGAA


Domain-VY

QPSDEDLKRITQTWQQADDENEE

GGACAAGCTGCCCGTGAGC


variant (EcR)

SDTPFRQITEMTILTVQLIVEFAKGL

ACCACCACCGTCGATGACC




PGFAKISQPDQITLLKACSSEVMML

ACATGCCCCCCATCATGCAG




RVARRYDAASDSILFANNQAYTRD

TGCGAGCCCCCCCCCCCCGA




NYRKAGMAEVIEDLLHFCRCMYS

GGCCGCCAGGATTCACGAG




MALDNIHYALLTAVVIFSDRPGLEQ

GTCGTGCCCAGGTTCCTGA




PQLVEEIQRYYLNTLRIYILNQLSGS

GCGACAAGCTGCTGGTGAC




ARSSVIYGKILSILSELRTLGMQNSN

CAACAGGCAGAAGAACATC




MCISLKLKNRKLPPFLEEIWDVAD

CCCCAGCTGACCGCCAACC




MSHTQPPPILESPTNL

AGCAGTTCCTGATCGCCAG






GCTGATCTGGTATCAGGAC






GGCTACGAGCAGCCCAGCG






ACGAGGACCTGAAAAGGAT






CACCCAGACCTGGCAGCAG






GCCGACGACGAGAACGAG






GAGAGCGACACCCCCTTCA






GGCAGATCACCGAGATGAC






CATCCTGACCGTGCAGCTG






ATCGTGGAGTTCGCCAAGG






GCCTGCCCGGATTCGCCAA






GATCAGCCAGCCCGACCAG






ATCACCCTGCTGAAGGCTT






GCAGCAGCGAGGTGATGAT






GCTGAGGGTGGCCAGGAG






GTACGACGCCGCCAGCGAC






AGCATCCTGTTCGCCAACAA






CCAGGCTTACACCAGGGAC






AACTACAGGAAGGCTGGCA






TGGCCGAGGTGATCGAGGA






CCTCCTGCACTTCTGCAGAT






GTATGTACAGCATGGCCCT






GGACAACATCCACTACGCC






CTGCTGACCGCCGTGGTGA






TCTTCAGCGACAGGCCCGG






CCTGGAGCAGCCCCAGCTG






GTGGAGGAGATCCAGAGGT






ACTACCTGAACACCCTGAG






GATCTACATCCTGAACCAGC






TGAGCGGCAGCGCCAGGA






GCAGCGTGATCTACGGCAA






GATCCTGAGCATCCTGAGC






GAGCTGAGGACCCTGGGAA






TGCAGAACAGCAATATGTG






TATCAGCCTGAAGCTGAAG






AACAGGAAGCTGCCCCCCT






TCCTGGAGGAGATTTGGGA






CGTGGCCGACATGAGCCAC






ACCCAGCCCCCCCCCATCCT






GGAGAGCCCCACCAACCTG





Ecdysone
642
RPECVVPETQCAMKRKEKKAQKEK
643
CGGCCTGAGTGCGTAGTAC


Receptor

DKLPVSTTTVDDHMPPIMQCEPPP

CCGAGACTCAGTGCGCCAT


Ligand

PEAARIHEVVPRFLSDKLLVTNRQK

GAAGCGGAAAGAGAAGAA


Binding

NIPQLTANQQFLIARLIWYQDGYE

AGCACAGAAGGAGAAGGA


Domain_VY

QPSDEDLKRITQTWQQADDENEE

CAAACTGCCTGTCAGCACG


variant (EcR)

SDTPFRQITEMTILTVQLIVEFAKGL

ACGACGGTGGACGACCACA




PGFAKISQPDQITLLKACSSEVMML

TGCCGCCCATTATGCAGTGT




RVARRYDAASDSILFANNQAYTRD

GAACCTCCACCTCCTGAAGC




NYRKAGMAEVIEDLLHFCRCMYS

AGCAAGGATTCACGAAGTG




MALDNIHYALLTAVVIFSDRPGLEQ

GTCCCAAGGTTTCTCTCCGA




PQLVEEIQRYYLNTLRIYILNQLSGS

CAAGCTGTTGGTGACAAAC




ARSSVIYGKILSILSELRTLGMQNSN

CGGCAGAAAAACATCCCCC




MCISLKLKNRKLPPFLEEIWDVAD

AGTTGACAGCCAACCAGCA




MSHTQPPPILESPTNL

GTTCCTTATCGCCAGGCTCA






TCTGGTACCAGGACGGGTA






CGAGCAGCCTTCTGATGAA






GATTTGAAGAGGATTACGC






AGACGTGGCAGCAAGCGG






ACGATGAAAACGAAGAGTC






GGACACTCCCTTCCGCCAGA






TCACAGAGATGACTATCCTC






ACGGTCCAACTTATCGTGG






AGTTCGCGAAGGGATTGCC






AGGGTTCGCCAAGATCTCG






CAGCCTGATCAAATTACGCT






GCTTAAGGCTTGCTCAAGT






GAGGTAATGATGCTCCGAG






TCGCGCGACGATACGATGC






GGCCTCAGACAGTATTCTGT






TCGCGAACAACCAAGCGTA






CACTCGCGACAACTACCGC






AAGGCTGGCATGGCCGAGG






TCATCGAGGATCTACTGCAC






TTCTGCCGGTGCATGTACTC






TATGGCGTTGGACAACATC






CATTACGCGCTGCTCACGG






CTGTCGTCATCTTTTCTGAC






CGGCCAGGGTTGGAGCAGC






CGCAACTGGTGGAAGAGAT






CCAGCGGTACTACCTGAAT






ACGCTCCGCATCTATATCCT






GAACCAGCTGAGCGGGTCG






GCGCGTTCGTCCGTCATATA






CGGCAAGATCCTCTCAATCC






TCTCTGAGCTACGCACGCTC






GGCATGCAAAACTCCAACA






TGTGCATCTCCCTCAAGCTC






AAGAACAGAAAGCTGCCGC






CTTTCCTCGAGGAGATCTG






GGATGTGGCGGACATGTCG






CACACCCAACCGCCGCCTAT






CCTCGAGTCCCCCACGAATC






TCTAG





GAL4-Linker-
644
MKLLSSIEQACDICRLKKLKCSKEKP
645
ATGAAGCTACTGTCTTCTAT


EcR

KCAKCLKNNWECRYSPKTKRSPLTR

CGAACAAGCATGCGATATT




AHLTEVESRLERLEQLFLLIFPREDLD

TGCCGACTTAAAAAGCTCA




MILKMDSLQDIKALLTGLFVQDNV

AGTGCTCCAAAGAAAAACC




NKDAVTDRLASVETDMPLTLRQHR

GAAGTGCGCCAAGTGTCTG




ISATSSSEESSNKGQRQLTVSPEFPG

AAGAACAACTGGGAGTGTC




IRPECVVPETQCAMKRKEKKAQKE

GCTACTCTCCCAAAACCAAA




KDKLPVSTTTVDDHMPPIMQCEPP

AGGTCTCCGCTGACTAGGG




PPEAARIHEVVPRFLSDKLLVTNRQ

CACATCTGACAGAAGTGGA




KNIPQLTANQQFLIARLIWYQDGYE

ATCAAGGCTAGAAAGACTG




QPSDEDLKRITQTWQQADDENEE

GAACAGCTATTTCTACTGAT




SDTPFRQITEMTILTVQLIVEFAKGL

TTTTCCTCGAGAAGACCTTG




PGFAKISQPDQITLLKACSSEVMML

ACATGATTTTGAAAATGGAT




RVARRYDAASDSILFANNQAYTRD

TCTTTACAGGATATAAAAGC




NYRKAGMAEVIEDLLHFCRCMYS

ATTGTTAACAGGATTATTTG




MALDNIHYALLTAVVIFSDRPGLEQ

TACAAGATAATGTGAATAA




PQLVEEIQRYYLNTLRIYILNQLSGS

AGATGCCGTCACAGATAGA




ARSSVIYGKILSILSELRTLGMQNSN

TTGGCTTCAGTGGAGACTG




MCISLKLKNRKLPPFLEEIWDVAD

ATATGCCTCTAACATTGAGA




MSHTQPPPILESPTNL

CAGCATAGAATAAGTGCGA






CATCATCATCGGAAGAGAG






TAGTAACAAAGGTCAAAGA






CAGTTGACTGTATCGCCGG






AATTCCCGGGGATCCGGCC






TGAGTGCGTAGTACCCGAG






ACTCAGTGCGCCATGAAGC






GGAAAGAGAAGAAAGCAC






AGAAGGAGAAGGACAAAC






TGCCTGTCAGCACGACGAC






GGTGGACGACCACATGCCG






CCCATTATGCAGTGTGAACC






TCCACCTCCTGAAGCAGCAA






GGATTCACGAAGTGGTCCC






AAGGTTTCTCTCCGACAAGC






TGTTGGTGACAAACCGGCA






GAAAAACATCCCCCAGTTG






ACAGCCAACCAGCAGTTCCT






TATCGCCAGGCTCATCTGGT






ACCAGGACGGGTACGAGCA






GCCTTCTGATGAAGATTTGA






AGAGGATTACGCAGACGTG






GCAGCAAGCGGACGATGAA






AACGAAGAGTCGGACACTC






CCTTCCGCCAGATCACAGA






GATGACTATCCTCACGGTCC






AACTTATCGTGGAGTTCGC






GAAGGGATTGCCAGGGTTC






GCCAAGATCTCGCAGCCTG






ATCAAATTACGCTGCTTAAG






GCTTGCTCAAGTGAGGTAA






TGATGCTCCGAGTCGCGCG






ACGATACGATGCGGCCTCA






GACAGTATTCTGTTCGCGA






ACAACCAAGCGTACACTCG






CGACAACTACCGCAAGGCT






GGCATGGCCGAGGTCATCG






AGGATCTACTGCACTTCTGC






CGGTGCATGTACTCTATGG






CGTTGGACAACATCCATTAC






GCGCTGCTCACGGCTGTCG






TCATCTTTTCTGACCGGCCA






GGGTTGGAGCAGCCGCAAC






TGGTGGAAGAGATCCAGCG






GTACTACCTGAATACGCTCC






GCATCTATATCCTGAACCAG






CTGAGCGGGTCGGCGCGTT






CGTCCGTCATATACGGCAA






GATCCTCTCAATCCTCTCTG






AGCTACGCACGCTCGGCAT






GCAAAACTCCAACATGTGC






ATCTCCCTCAAGCTCAAGAA






CAGAAAGCTGCCGCCTTTCC






TCGAGGAGATCTGGGATGT






GGCGGACATGTCGCACACC






CAACCGCCGCCTATCCTCGA






GTCCCCCACGAATCTCTAG





GAL4-Linker-
646
MKLLSSIEQACDICRLKKLKCSKEKP
647
ATGAAGCTGCTGAGCAGCA


EcR

KCAKCLKNNWECRYSPKTKRSPLTR

TCGAGCAGGCTTGCGACAT




AHLTEVESRLERLEQLFLLIFPREDLD

CTGCAGGCTGAAGAAGCTG




MILKMDSLQDIKALLTGLFVQDNV

AAGTGCAGCAAGGAGAAG




NKDAVTDRLASVETDMPLTLRQHR

CCCAAGTGCGCCAAGTGCC




ISATSSSEESSNKGQRQLTVSPEFPG

TGAAGAACAACTGGGAGTG




RPECVVPETQCAMKRKEKKAQKEK

CAGATACAGCCCCAAGACC




DKLPVSTTTVDDHMPPIMQCEPPP

AAGAGGAGCCCCCTGACCA




PEAARIHEVVPRFLSDKLLVTNRQK

GGGCCCACCTGACCGAGGT




NIPQLTANQQFLIARLIWYQDGYE

GGAGAGCAGGCTGGAGAG




QPSDEDLKRITQTWQQADDENEE

GCTGGAGCAGCTGTTCCTG




SDTPFRQITEMTILTVQLIVEFAKGL

CTGATCTTCCCCAGGGAGG




PGFAKISQPDQITLLKACSSEVMML

ACCTGGACATGATCCTGAA




RVARRYDAASDSILFANNQAYTRD

GATGGACAGCCTGCAAGAC




NYRKAGMAEVIEDLLHFCRCMYS

ATCAAGGCCCTGCTGACCG




MALDNIHYALLTAVVIFSDRPGLEQ

GCCTGTTCGTGCAGGACAA




PQLVEEIQRYYLNTLRIYILNQLSGS

CGTGAACAAGGACGCCGTG




ARSSVIYGKILSILSELRTLGMQNSN

ACCGACAGGCTGGCCAGCG




MCISLKLKNRKLPPFLEEIWDVAD

TGGAGACCGACATGCCCCT




MSHTQPPPILESPTNL

GACCCTGAGGCAGCACAGG






ATCAGCGCCACCAGCAGCA






GCGAGGAGAGCAGCAACA






AGGGCCAGAGGCAGCTGAC






CGTGAGCCCCGAGTTTCCC






GGGCGGCCTGAGTGCGTAG






TACCCGAGACTCAGTGCGC






CATGAAGCGGAAAGAGAA






GAAAGCACAGAAGGAGAA






GGACAAACTGCCTGTCAGC






ACGACGACGGTGGACGACC






ACATGCCGCCCATTATGCAG






TGTGAACCTCCACCTCCTGA






AGCAGCAAGGATTCACGAA






GTGGTCCCAAGGTTTCTCTC






CGACAAGCTGTTGGTGACA






AACCGGCAGAAAAACATCC






CCCAGTTGACAGCCAACCA






GCAGTTCCTTATCGCCAGGC






TCATCTGGTACCAGGACGG






GTACGAGCAGCCTTCTGAT






GAAGATTTGAAGAGGATTA






CGCAGACGTGGCAGCAAGC






GGACGATGAAAACGAAGA






GTCGGACACTCCCTTCCGCC






AGATCACAGAGATGACTAT






CCTCACGGTCCAACTTATCG






TGGAGTTCGCGAAGGGATT






GCCAGGGTTCGCCAAGATC






TCGCAGCCTGATCAAATTAC






GCTGCTTAAGGCTTGCTCAA






GTGAGGTAATGATGCTCCG






AGTCGCGCGACGATACGAT






GCGGCCTCAGACAGTATTC






TGTTCGCGAACAACCAAGC






GTACACTCGCGACAACTACC






GCAAGGCTGGCATGGCCGA






GGTCATCGAGGATCTACTG






CACTTCTGCCGGTGCATGTA






CTCTATGGCGTTGGACAAC






ATCCATTACGCGCTGCTCAC






GGCTGTCGTCATCTTTTCTG






ACCGGCCAGGGTTGGAGCA






GCCGCAACTGGTGGAAGAG






ATCCAGCGGTACTACCTGA






ATACGCTCCGCATCTATATC






CTGAACCAGCTGAGCGGGT






CGGCGCGTTCGTCCGTCAT






ATACGGCAAGATCCTCTCAA






TCCTCTCTGAGCTACGCACG






CTCGGCATGCAAAACTCCA






ACATGTGCATCTCCCTCAAG






CTCAAGAACAGAAAGCTGC






CGCCTTTCCTCGAGGAGATC






TGGGATGTGGCGGACATGT






CGCACACCCAACCGCCGCCT






ATCCTCGAGTCCCCCACGAA






TCTCTAG





EMCV IRES


702
CCCCCTCTCCCTCCCCCCCCC






CTAACGTTACTGGCCGAAG






CCGCTTGGAATAAGGCCGG






TGTGCGTTTGTCTATATGTT






ATTTTCCACCATATTGCCGT






CTTTTGGCAATGTGAGGGC






CCGGAAACCTGGCCCTGTC






TTCTTGACGAGCATTCCTAG






GGGTCTTTCCCCTCTCGCCA






AAGGAATGCAAGGTCTGTT






GAATGTCGTGAAGGAAGCA






GTTCCTCTGGAAGCTTCTTG






AAGACAAACAACGTCTGTA






GCGACCCTTTGCAGGCAGC






GGAACCCCCCACCTGGCGA






CAGGTGCCTCTGCGGCCAA






AAGCCACGTGTATAAGATA






CACCTGCAAAGGCGGCACA






ACCCCAGTGCCACGTTGTG






AGTTGGATAGTTGTGGAAA






GAGTCAAATGGCTCTCCTCA






AGCGTATTCAACAAGGGGC






TGAAGGATGCCCAGAAGGT






ACCCCATTGTATGGGATCTG






ATCTGGGGCCTCGGTGCAC






ATGCTTTACATGTGTTTAGT






CGAGGTTAAAAAACGTCTA






GGCCCCCCGAACCACGGGG






ACGTGGTTTTCCTTTGAAAA






ACACGATC





2xRbm3 IRES


703
ACTAGTTTTATAATTTCTTCT






TCCAGAATTTCTGACATTTT






ATAATTTCTTCTTCCAGAAG






ACTCACAACCTC








Claims
  • 1. A non-naturally occurring polynucleotide encoding: (a) a miRNA that inhibits the expression of an immune checkpoint protein; and (b) a chimeric receptor.
  • 2. The polynucleotide of claim 1, wherein the miRNA inhibits the expression of CTLA, PD-1, PD-L1, TIM3, TIGIT, LAG3, GITR, or PIK31P1.
  • 3-6. (canceled)
  • 7. The polynucleotide of claim 2, comprising a nucleic acid sequence having at least 80% sequence identity with SEQ ID NO: 267 or that is capable of hybridizing under stringent hybridization conditions to the complement of SEQ ID NO: 267.
  • 8-9. (canceled)
  • 10. The polynucleotide of claim 1, wherein the chimeric receptor is a T-cell receptor or a chimeric antigen receptor.
  • 11-12. (canceled)
  • 13. The polynucleotide of claim 10, wherein the chimeric antigen receptor comprises an antigen-binding domain that binds to an epitope on ROR1.
  • 14. The polynucleotide of claim 13, wherein the chimeric antigen receptor comprises: (a) a variable light chain domain comprising the amino acid sequence of any one of SEQ ID NOs: 347, 351, 355, 359, 363, 367, 371, 375, 379, 383, 387, 391, 395, 399, 403, 407, 411, 415, 419, 423, 427, 431, 435, 439, 443, 447, 451, 455, 459, and 463, or a functional fragment or variant thereof; and/or(b) a variable heavy chain domain comprising the amino acid sequence of any one of SEQ ID NOs: 349, 353, 357, 361, 365, 369, 373, 377, 381, 385, 389, 393, 397, 401, 405, 409, 413, 417, 421, 425, 429, 433, 437, 441, 445, 449, 453, 457, and 461, or a functional fragment or variant thereof.
  • 15-17. (canceled)
  • 18. The polynucleotide of claim 10, wherein the chimeric antigen receptor comprises a spacer comprising: (a) a stalk region comprising the amino acid sequence of SEQ ID NO: 467, or a functional fragment or variant thereof; and (b) a stalk extension region comprising the amino acid sequence of SEQ ID NO: 473, or a functional fragment or variant thereof.
  • 19-31. (canceled)
  • 32. The polynucleotide of claim 1, further encoding a cytokine.
  • 33-35. (canceled)
  • 36. The polynucleotide of claim 32, encoding a fusion protein comprising: (a) IL-15, or a functional fragment or variant thereof; and (b) IL-15Rα, or a functional fragment or variant thereof.
  • 37. The polynucleotide of claim 36, wherein the fusion protein comprises the amino acid sequence of SEQ ID NO: 523, or a functional fragment or variant thereof.
  • 38. (canceled)
  • 39. The polynucleotide of claim 1, further encoding a cell tag.
  • 40-42. (canceled)
  • 43. The polynucleotide of claim 39, wherein the cell tag comprises: (a) a truncated HER1, or a functional fragment or variant thereof; and a CD28 transmembrane domain, or a functional fragment or variant thereof.
  • 44. (canceled)
  • 45. A vector comprising the polynucleotide of claim 1.
  • 46. (canceled)
  • 47. The vector of claim 45, comprising a Sleeping Beauty transposon.
  • 48. A modified immune effector cell comprising the polynucleotide of claim 1.
  • 49. A composition comprising the polynucleotide of claim 1.
  • 50-57. (canceled)
  • 58. A method for detecting a disease or disorder associated with the overexpression of an antigen in a subject, the method comprising: a) contacting a sample from the subject with one or more of the antibodies, or antigen-binding fragments thereof; and b) detecting an increased level of binding of the antibody or fragment thereof to the sample as compared to such binding to a control sample lacking the disease, thereby detecting the disease in the subject.
  • 59. A method for treating a disease or disorder comprising the serial administration of polynucleotides encoding a chimeric antigen receptor or a cell comprising the same, wherein the encoded chimeric antigen receptors are selected from a collection of chimeric antigen receptors having different structural compositions and binding specificities for an array of antigen targets.
  • 60. (canceled)
  • 61. A kit comprising the polynucleotide of claim 1.
  • 62. A method of treating a subject suffering from a disease or disorder, comprising administering the cell of claim 48 to a subject in need thereof.
Provisional Applications (3)
Number Date Country
63136611 Jan 2021 US
63263001 Oct 2021 US
63263508 Nov 2021 US
Continuations (1)
Number Date Country
Parent 63136074 Jan 2021 US
Child 17572618 US