MODULATING T CELL FUNCTION AND RESPONSE

Abstract
The present disclosure describes a method of producing T cells exhibiting an enhanced memory T cell phenotype, the method comprising: modulating a population of T cells to enhance the expression and/or function of high mobility group protein Y (HMGY). In embodiments, the method may include introducing a polynucleotide encoding HMGY into a population of T cells, wherein expression of HMGY is higher in the population of T cells as compared to a population of T cells that are not introduced with the polynucleotide, and the memory T cell phenotype of the population of T cells is enhanced as compared to T cells that are not introduced with the polynucleotide. In embodiments, the method can also include introducing a polynucleotide encoding one or more genes associated with HMGY, for example, upstream or downstream of the signaling pathway associated with HMGY and/or a transcription factor associated with HMGY.
Description
SEQUENCE LISTING INFORMATION

A computer-readable textfile, entitled “SDS1.0082US_ST25.txt,” created on or about Jul. 10, 2020, with a file size of about 128 KB, contains the sequence listing for this application and is hereby incorporated by reference in its entirety.


TECHNICAL FIELD

The present disclosure relates to compositions and methods for expanding and maintaining modified cells including genetically modified cells and uses thereof in the treatment of diseases, including cancer.


BACKGROUND

T cells genetically targeted to certain malignancies have demonstrated tremendous clinical outcomes. During CAR-T cell therapy, physicians draw patients' blood and harvest their cytotoxic T cells. The cells are re-engineered in a lab to attack her particular cancer. Recent progress in genome editing technologies allows scientists to modulate gene expression in T-cells to enhance effector functions or to bypass tumor immune suppression and metabolically hostile tumor microenvironment. Thus, there is a need to modulate T cells to overcome these problems.


SUMMARY

Embodiments relate to a method of producing T cells exhibiting an enhanced memory T cell phenotype, the method comprising: modulating a population of T cells to enhance the expression and/or function of HMGY. For example, the method may include introducing a polynucleotide encoding HMGY into a population of T cells, wherein expression of HMGY is higher as compared to T cells that are not introduced with the polynucleotide, and the memory T cell phenotype of the population of T cells is enhanced as compared to T cells that are not introduced with the polynucleotide. In embodiments, the method may include introducing a polynucleotide encoding one or more genes associated with HMGY, for example, upstream or downstream of the signaling pathway associated with HMGY and/or a transcription factor associated with HMGY.


Embodiments relate to a method of producing T cells exhibiting an enhanced memory T cell phenotype, the method comprising: introducing a polynucleotide encoding HMGY into a population of T cells, wherein expression of HMGY is higher as compared to T cells that are not introduced with the polynucleotide, and the memory T cell phenotype of the population of T cells is enhanced as compared to T cells that are not introduced with the polynucleotide. In embodiments, the population of T cells exhibiting an increased gene expression level in CD62L and/or CCR7 as compared to T cells that are not introduced with the polynucleotide. In embodiments, the method further comprises culturing the population; and measuring cell expansion of the population of T cells. In embodiments, expansion of the population of T cells is enhanced as compared to T cells that are not introduced with the polynucleotide.


Embodiments relate to a modified cell engineered to express an antigen binding molecule, wherein the expression and/or function of one or more genes in the modified cell has been enhanced. In embodiments, the one or more genes comprise at least one of BATF, HMGA1, STAT5A, ZNF580, GLMP, JAZF1, RUNX1, ZGPAT, ZNF511, GTF2IRD2B, ATF4, MBD4, TBPL1, GTF2B, RBCK1, ZBTB38, PIN1, DRAP1, THYN1, HSF1, PRDM1, ZNF428, NFYC, and ZNF706. In embodiments, one or more genes are HMGA1 and/or ZBTB38.


Embodiments relate to a modified cell engineered to express an antigen binding molecule, wherein the expression and/or function of one or more genes in the modified cell has been reduced or eliminated. In embodiments, the one or more genes comprise at least one of GTF3A, JUN, IRF1, JUNB, TMF1, ELF1, AKNA, BCL11B, KLF2, ZNF292, RORA, HMGN3, KDM2A, ASCL2, SP140L, NFATC2, RUNX3, NFE2L2, KLF6, MTERF4, PHF20, RELB, MAZ, ARID5A, REL, ZEB2, ARID5B, KLF3, CREM, ZNF207, IRF7, DR1, SP140, BBX, MECP2, STAT4, ZBTB1, CREBZF, NFATC3, GPBP1, IKZF1, SON, ZNF800, STAT3, STATE, CGGBP1, FOXN2, DNMT1, SP100, GATA3, EOMES, YY1, SP110, SAFB, REST, NR3C1, FOXN3, ELF2, GTF2I, BAZ2A, ZNF683, STAT1, BHLHE40, ZNF276, ETS1, NFAT5, BPTF, KMT2A, FOS, PA2G4, IKZF3, ZNF148, CDC5L, CREB1, HBP1, ZNF721, KAT7, SP4, ZC3H8, AKAP8L, ZNF326, ZNF451, ZNF131, CEBPZ, TOPORS, ZNF33A, NCOA3, STAT2, DDIT3, ZNF217, KLF9, CSRNP1, NCOA1, SAFB2, ZNF107, ZFX, E2F4, HIF1A, ZNF480, CTCF, ZBTB44, NCOA2, ZHX1, ZNF644, ASH1L, STAT5B, AEBP2, MYSM1, ZNF91, CEBPB, MXD4, YBX3, RLF, JUND, ZNF600, SMAD4, TET2, ZNF267, PRDM2, ZBTB7A, THAP12, ETV3, NFKB2, KLF13, SATB1, ZNF791, RBPJ, SPEN, PURA, ZNF507, FOSL2, IRF8, ELK4, ATF3, KCMF1, ZNF639, SKI, FOXO1, NR4A2, ZNF331, NFKB1, CEBPD, FOSB, SKIL, NR4A3, and NR4A1. In embodiments, the one or more genes are AKNA.


This Summary is not intended to identify key features or essential features of the claimed subject matter, nor is it intended to be used to limit the scope of the claimed subject matter.





BRIEF DESCRIPTION OF THE DRAWINGS

The Detailed Description is described with reference to the accompanying figures. The use of the same reference numbers in different figures indicates similar or identical items.



FIG. 1 shows TCR clonal enrichment in a clinical trial.



FIG. 2 shows TCR monoclonal TRBV9 is highly enriched in a clinical trial.



FIGS. 3 and 4 show the analysis of intracellular pathways based on single-cell sequencing and existing databases.



FIG. 5 shows the expression of HMGY in various cells.



FIGS. 6 and 7 show flow cytometry results of expression of markers CD62L and CCR7 of various cells.



FIGS. 8 and 9 show flow cytometry results of expression of marker KLRG and CD137 of various cells.



FIGS. 10 and 11 shows flow cytometry results of cell expansion of various cells.





DETAILED DESCRIPTION

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which the disclosure belongs. Although any method and material similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, preferred methods and materials are described. For the purposes of the present disclosure, the following terms are defined below.


The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.


By “about” is meant a quantity, level, value, number, frequency, percentage, dimension, size, amount, weight or length that varies by as much as 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1% to a reference quantity, level, value, number, frequency, percentage, dimension, size, amount, weight or length.


The term “activation,” as used herein, refers to the state of a cell that has been sufficiently stimulated to induce detectable cellular proliferation. Activation can also be associated with induced cytokine production and detectable effector functions. The term “activated T cells” refers to, among other things, T cells that are undergoing cell division.


The term “antibody” is used in the broadest sense and refers to monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multi-specific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity or function. The antibodies in the present disclosure may exist in a variety of forms including, for example, polyclonal antibodies; monoclonal antibodies; Fv, Fab, Fab′, and F(ab′)2 fragments; as well as single chain antibodies and humanized antibodies (Harlow et al., 1999, In: Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow et al., 1989, In: Antibodies: A Laboratory Manual, Cold Spring Harbor, N.Y.; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; Bird et al., 1988, Science 242:423-426).


The term “antibody fragments” refers to a portion of a full-length antibody, for example, the antigen binding or variable region of the antibody. Other examples of antibody fragments include Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multi-specific antibodies formed from antibody fragments.


The term “Fv” refers to the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in a tight, non-covalent association. From the folding of these two domains emanates six hypervariable loops (3 loops each from the H and L chain) that contribute amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv including only three complementarity determining regions (CDRs) specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site (the dimer).


An “antibody heavy chain,” as used herein, refers to the larger of the two types of polypeptide chains present in all antibody molecules in their naturally occurring conformations. An “antibody light chain,” as used herein, refers to the smaller of the two types of polypeptide chains present in all antibody molecules in their naturally occurring conformations. K and A light chains refer to the two major antibody light chain isotypes.


The term “synthetic antibody” refers to an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage. The term also includes an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and the expression of the DNA molecule to obtain the antibody or to obtain an amino acid encoding the antibody. The synthetic DNA is obtained using technology that is available and well known in the art.


The term “antigen” refers to a molecule that provokes an immune response, which may involve either antibody production, or the activation of specific immunologically-competent cells, or both. Antigens include any macromolecule, including all proteins or peptides, or molecules derived from recombinant or genomic DNA. For example, DNA including a nucleotide sequence or a partial nucleotide sequence encoding a protein or peptide that elicits an immune response, and therefore, encodes an “antigen” as the term is used herein. An antigen need not be encoded solely by a full-length nucleotide sequence of a gene. An antigen can be generated, synthesized, or derived from a biological sample including a tissue sample, a tumor sample, a cell, or a biological fluid.


The term “anti-tumor effect” as used herein, refers to a biological effect associated with a decrease in tumor volume, a decrease in the number of tumor cells, a decrease in the number of metastases, decrease in tumor cell proliferation, decrease in tumor cell survival, an increase in life expectancy of a subject having tumor cells, or amelioration of various physiological symptoms associated with the cancerous condition. An “anti-tumor effect” can also be manifested by the ability of the peptides, polynucleotides, cells, and antibodies in the prevention of the occurrence of tumors in the first place.


The term “auto-antigen” refers to an endogenous antigen mistakenly recognized by the immune system as being foreign. Auto-antigens include cellular proteins, phosphoproteins, cellular surface proteins, cellular lipids, nucleic acids, glycoproteins, including cell surface receptors.


The term “autologous” is used to describe a material derived from a subject that is subsequently re-introduced into the same subject.


The term “allogeneic” is used to describe a graft derived from a different subject of the same species. As an example, a donor subject may be related or unrelated to the recipient subject, but the donor subject has immune system markers that are similar to the recipient subject.


The term “xenogeneic” is used to describe a graft derived from a subject of a different species. As an example, the donor subject is from a different species than a recipient subject, and the donor subject and the recipient subject can be genetically and immunologically incompatible.


The term “cancer” is used to refer to a disease characterized by the rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body. Examples of various cancers include breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, lung cancer, and the like.


Throughout this specification, unless the context requires otherwise, the words “comprise,” “includes” and “including” will be understood to imply the inclusion of a stated step or element or group of steps or elements but not the exclusion of any other step or element or group of steps or elements.


The phrase “consisting of” is meant to include, and is limited to, whatever follows the phrase “consisting of.” Thus, the phrase “consisting of” indicates that the listed elements are required or mandatory and that no other elements may be present.


The phrase “consisting essentially of” is meant to include any element listed after the phrase and can include other elements that do not interfere with or contribute to the activity or action specified in the disclosure for the listed elements. Thus, the phrase “consisting essentially of” indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present depending upon whether or not they affect the activity or action of the listed elements.


The terms “complementary” and “complementarity” refer to polynucleotides (i.e., a sequence of nucleotides) related by the base-pairing rules. For example, the sequence “A-G-T” is complementary to the sequence “T-C-A.” Complementarity may be “partial,” in which only some of the nucleic acids' bases are matched according to the base-pairing rules, or there may be “complete” or “total” complementarity between the nucleic acids. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands.


The term “corresponds to” or “corresponding to” refers to (a) a polynucleotide having a nucleotide sequence that is substantially identical or complementary to all or a portion of a reference polynucleotide sequence or encoding an amino acid sequence identical to an amino acid sequence in a peptide or protein, or (b) a peptide or polypeptide having an amino acid sequence that is substantially identical to a sequence of amino acids in a reference peptide or protein.


The term “co-stimulatory ligand,” refers to a molecule on an antigen-presenting cell (e.g., an APC, dendritic cell, B cell, and the like) that specifically binds a cognate co-stimulatory molecule on a T cell, thereby providing a signal which, in addition to the primary signal provided by, for instance, binding of a TCR/CD3 complex with an MHC molecule loaded with peptide, mediates a T cell response, including at least one of proliferation, activation, differentiation, and other cellular responses. A co-stimulatory ligand can include B7-1 (CD80), B7-2 (CD86), PD-L1, PD-L2, 4-1BBL, OX40L, inducible co-stimulatory ligand (ICOS-L), intercellular adhesion molecule (ICAM), CD30L, CD40, CD70, CD83, HLA-G, MICA, MICB, HVEM, lymphotoxin beta receptor, 3/TR6, ILT3, ILT4, HVEM, a ligand for CD7, an agonist or antibody that binds the Toll ligand receptor, and a ligand that specifically binds with B7-H3. A co-stimulatory ligand also includes, inter alia, an agonist or an antibody that specifically binds with a co-stimulatory molecule present on a T cell, such as CD27, CD28, 4-1BB, OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, and a ligand that specifically binds CD83.


The term “co-stimulatory molecule” refers to the cognate binding partner on a T cell that specifically binds with a co-stimulatory ligand, thereby mediating a co-stimulatory response by the T cell, such as proliferation. Co-stimulatory molecules include an MHC class I molecule, BTLA, and a Toll-like receptor.


The term “co-stimulatory signal” refers to a signal, which in combination with a primary signal, such as TCR/CD3 ligation, leads to T cell proliferation and/or upregulation or downregulation of key molecules.


The terms “disease” and “condition” may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out), and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians. The term “disease” is a state of health of a subject wherein the subject cannot maintain homeostasis, and wherein if the disease is not ameliorated then the subject's health continues to deteriorate. In contrast, a “disorder” in a subject is a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health.


The term “effective” refers to adequate to accomplish a desired, expected, or intended result. For example, an “effective amount” in the context of treatment may be an amount of a compound sufficient to produce a therapeutic or prophylactic benefit.


The term “encoding” refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as a template for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence (except that a “T” is replaced by a “U”) and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA.


The term “exogenous” refers to a molecule that does not naturally occur in a wild-type cell or organism but is typically introduced into the cell by molecular biological techniques. Examples of exogenous polynucleotides include vectors, plasmids, and/or man-made nucleic acid constructs encoding the desired protein. With regard to polynucleotides and proteins, the term “endogenous” or “native” refers to a naturally-occurring polynucleotide or amino acid sequences that may be found in a given wild-type cell or organism. Also, a particular polynucleotide sequence that is isolated from a first organism and transferred to a second organism by molecular biological techniques is typically considered an “exogenous” polynucleotide or amino acid sequence with respect to the second organism. In specific embodiments, polynucleotide sequences can be “introduced” by molecular biological techniques into a microorganism that already contains such a polynucleotide sequence, for instance, to create one or more additional copies of an otherwise naturally-occurring polynucleotide sequence, and thereby facilitate overexpression of the encoded polypeptide.


The term “expression or overexpression” refers to the transcription and/or translation of a particular nucleotide sequence into a precursor or mature protein, for example, driven by its promoter. “Overexpression” refers to the production of a gene product in transgenic organisms or cells that exceeds levels of production in normal or non-transformed organisms or cells. As defined herein, the term “expression” refers to expression or overexpression.


The term “expression vector” refers to a vector including a recombinant polynucleotide including expression control (regulatory) sequences operably linked to a nucleotide sequence to be expressed. An expression vector includes sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system. Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentiviruses, retroviruses, adenoviruses, and adeno-associated viruses) that incorporate the recombinant polynucleotide.


Viruses can be used to deliver nucleic acids into a cell in vitro and in vivo (in a subject). Examples of viruses useful for delivery of nucleic acids into cells include retrovirus, adenovirus, herpes simplex virus, vaccinia virus, and adeno-associated virus.


There also exist non-viral methods for delivering nucleic acids into a cell, for example, electroporation, gene gun, sonoporation, magnetofection, and the use of oligonucleotides, lipoplexes, dendrimers, and inorganic nanoparticles.


The term “homologous” refers to sequence similarity or sequence identity between two polypeptides or between two polynucleotides when a position in both of the two compared sequences is occupied by the same base or amino acid monomer subunit, e.g., if a position in each of two DNA molecules is occupied by adenine, then the molecules are homologous at that position. The percent of homology between two sequences is a function of the number of matching or homologous positions shared by the two sequences divided by the number of positions compared ×100. For example, if 6 of 10 of the positions in two sequences are matched or homologous, then the two sequences are 60% homologous. By way of example, the DNA sequences ATTGCC and TATGGC share 50% homology. A comparison is made when two sequences are aligned to give maximum homology.


The term “immunoglobulin” or “Ig,” refers to a class of proteins, which function as antibodies. The five members included in this class of proteins are IgA, IgG, IgM, IgD, and IgE. IgA is the primary antibody that is present in body secretions, such as saliva, tears, breast milk, gastrointestinal secretions, and mucus secretions of the respiratory and genitourinary tracts. IgG is the most common circulating antibody. IgM is the main immunoglobulin produced in the primary immune response in most subjects. It is the most efficient immunoglobulin in agglutination, complement fixation, and other antibody responses, and is important in defense against bacteria and viruses. IgD is the immunoglobulin that has no known antibody function but may serve as an antigen receptor. IgE is the immunoglobulin that mediates immediate hypersensitivity by causing the release of mediators from mast cells and basophils upon exposure to the allergen.


The term “isolated” refers to a material that is substantially or essentially free from components that normally accompany it in its native state. The material can be a cell or a macromolecule such as a protein or nucleic acid. For example, an “isolated polynucleotide,” as used herein, refers to a polynucleotide, which has been purified from the sequences which flank it in a naturally-occurring state, e.g., a DNA fragment which has been removed from the sequences that are normally adjacent to the fragment. Alternatively, an “isolated peptide” or an “isolated polypeptide” and the like, as used herein, refer to in vitro isolation and/or purification of a peptide or polypeptide molecule from its natural cellular environment, and from association with other components of the cell.


The term “substantially purified” refers to a material that is substantially free from components that are normally associated with it in its native state. For example, a substantially purified cell refers to a cell that has been separated from other cell types with which it is normally associated in its naturally occurring or native state. In some instances, a population of substantially purified cells refers to a homogenous population of cells. In other instances, this term refers simply to a cell that has been separated from the cells with which they are naturally associated in their natural state. In embodiments, the cells are cultured in vitro. In embodiments, the cells are not cultured in vitro.


In the context of the present disclosure, the following abbreviations for the commonly occurring nucleic acid bases are used. “A” refers to adenosine, “C” refers to cytosine, “G” refers to guanosine, “T” refers to thymidine, and “U” refers to uridine.


Unless otherwise specified, a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. The phrase nucleotide sequence that encodes a protein or an RNA may also include introns to the extent that the nucleotide sequence encoding the protein may, in some version, contain an intron(s).


The term “lentivirus” refers to a genus of the Retroviridae family. Lentiviruses are unique among the retroviruses in being able to infect non-dividing cells; they can deliver a significant amount of genetic information into the DNA of the host cell, so they are one of the most efficient methods of a gene delivery vector. Moreover, the use of lentiviruses enables the integration of the genetic information into the host chromosome, resulting in stably transduced genetic information. HIV, SIV, and FIV are all examples of lentiviruses. Vectors derived from lentiviruses offer the means to achieve significant levels of gene transfer in vivo.


The term “modulating,” refers to mediating a detectable increase or decrease in the level of a response in a subject compared with the level of a response in the subject in the absence of a treatment or compound, and/or compared with the level of a response in an otherwise identical but untreated subject. The term encompasses perturbing and/or affecting a native signal or response, thereby mediating a beneficial therapeutic response in a subject, preferably, a human.


Nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence, or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.


The term “under transcriptional control” refers to a promoter being operably linked to and in the correct location and orientation in relation to a polynucleotide to control (regulate) the initiation of transcription by RNA polymerase and expression of the polynucleotide.


The term “overexpressed” tumor antigen or “overexpression” of the tumor antigen is intended to indicate an abnormal level of expression of the tumor antigen in a cell from a disease area such as a solid tumor within a specific tissue or organ of the patient relative to the level of expression in a normal cell from that tissue or organ. Patients having solid tumor or a hematological malignancy characterized by overexpression of the tumor antigen can be determined by standard assays known in the art.


Solid tumors are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors, such as sarcomas and carcinomas, include fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, lymphoid malignancy, pancreatic cancer, breast cancer, lung cancers, ovarian cancer, prostate cancer, hepatocellular carcinoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, pheochromocytomas sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, Wilms' tumor, cervical cancer, testicular tumor, seminoma, bladder carcinoma, melanoma, and CNS tumors (such as a glioma (such as brainstem glioma and mixed gliomas), glioblastoma (also known as glioblastoma multiforme), astrocytoma, CNS lymphoma, germinoma, medulloblastoma, Schwannoma craniopharyogioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, neuroblastoma, retinoblastoma, and brain metastases).


A solid tumor antigen is an antigen expressed on a solid tumor. In embodiments, solid tumor antigens are also expressed at low levels on healthy tissue. Examples of solid tumor antigens and their related disease tumors are provided in Table 1.












TABLE 1







Solid Tumor antigen
Disease tumor









PRLR
Breast cancer



CLCA1
colorectal cancer



MUC12
colorectal cancer



GUCY2C
colorectal cancer



GPR35
colorectal cancer



CR1L
Gastric cancer



MUC 17
Gastric cancer



TMPRSS11B
esophageal cancer



MUC21
esophageal cancer



TMPRSS11E
esophageal cancer



CD207
bladder cancer



SLC30A8
pancreatic cancer



CFC1
pancreatic cancer



SLC12A3
Cervical cancer



SSTR1
Cervical tumor



GPR27
Ovary tumor



FZD10
Ovary tumor



TSHR
Thyroid Tumor



SIGLEC15
Urothelial cancer



SLC6A3
Renal cancer



KISS1R
Renal cancer



QRFPR
Renal cancer:



GPR119
Pancreatic cancer



CLDN6
Endometrial cancer/Urothelial cancer



UPK2
Urothelial cancer (including bladder




cancer)



ADAM12
Breast cancer, pancreatic cancer and




the like



SLC45A3
Prostate cancer



ACPP
Prostate cancer



MUC21
Esophageal cancer



MUC16
Ovarian cancer



MS4A12
Colorectal cancer



ALPP
Endometrial cancer



CEA
Colorectal carcinoma



EphA2
Glioma



FAP
Mesothelioma



GPC3
Lung squamous cell carcinoma



IL13-Rα2
Glioma



Mesothelin
Metastatic cancer



PSMA
Prostate cancer



ROR1
Breast lung carcinoma



VEGFR-II
Metastatic cancer



GD2
Neuroblastoma



FR-α
Ovarian carcinoma



ErbB2
Carcinoma



EpCAM
Carcinoma



EGFRvIII
Glioma-Glioblastoma



EGFR
Glioma-NSCL cancer



tMUC1
Cholangiocarcinoma, Pancreatic




cancer, Breast



PSCA
pancreas, stomach, or prostate cancer



FCER2, GPR18, FCRLA,
breast cancer



CXCR5, FCRL3, FCRL2,




HTR3A, and CLEC17A




TRPMI, SLC45A2, and
lymphoma



SLC24A5




DPEP3
melanoma



KCNK16
ovarian, testis



LIM2 or KCNV2
pancreatic



SLC26A4
thyroid cancer



CD171
Neuroblastoma



Glypican-3
Sarcoma



IL-13
Glioma



CD79a/b
Lymphoma



MAGE A4
Lung and other cancer types










The term “parenteral administration” of a composition includes, e.g., subcutaneous (s.c.), intravenous (i.v.), intramuscular (i.m.), intrasternal injection, or infusion techniques.


The terms “patient,” “subject,” and “individual,” and the like are used interchangeably herein and refer to any human or animal, amenable to the methods described herein. In certain non-limiting embodiments, the patient, subject, or individual is a human or animal. In embodiments, the term “subject” is intended to include living organisms in which an immune response can be elicited (e.g., mammals). Examples of subjects include humans, and animals, such as dogs, cats, mice, rats, and transgenic species thereof.


A subject in need of treatment or in need thereof includes a subject having a disease, condition, or disorder that needs to be treated. A subject in need thereof also includes a subject that needs treatment for the prevention of a disease, condition, or disorder.


The term “polynucleotide” or “nucleic acid” refers to mRNA, RNA, cRNA, rRNA, cDNA or DNA. The term typically refers to a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide. The term includes all forms of nucleic acids including single and double-stranded forms of nucleic acids.


The terms “polynucleotide variant” and “variant” and the like refer to polynucleotides displaying substantial sequence identity with a reference polynucleotide sequence or polynucleotides that hybridize with a reference sequence under stringent conditions that are defined hereinafter. These terms also encompass polynucleotides that are distinguished from a reference polynucleotide by the addition, deletion, or substitution of at least one nucleotide. Accordingly, the terms “polynucleotide variant” and “variant” include polynucleotides in which one or more nucleotides have been added or deleted or replaced with different nucleotides. In this regard, it is well understood in the art that certain alterations inclusive of mutations, additions, deletions, and substitutions can be made to a reference polynucleotide whereby the altered polynucleotide retains the biological function or activity of the reference polynucleotide or has increased activity in relation to the reference polynucleotide (i.e., optimized). Polynucleotide variants include, for example, polynucleotides having at least 50% (and at least 51% to at least 99% and all integer percentages in between, e.g., 90%, 95%, or 98%) sequence identity with a reference polynucleotide sequence described herein. The terms “polynucleotide variant” and “variant” also include naturally-occurring allelic variants and orthologs.


The terms “polypeptide,” “polypeptide fragment,” “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues and to variants and synthetic analogues of the same. Thus, these terms apply to amino acid polymers in which one or more amino acid residues are synthetic non-naturally occurring amino acids, such as a chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally-occurring amino acid polymers. In certain aspects, polypeptides may include enzymatic polypeptides, or “enzymes,” which typically catalyze (i.e., increase the rate of) various chemical reactions.


The term “polypeptide variant” refers to polypeptides that are distinguished from a reference polypeptide sequence by the addition, deletion, or substitution of at least one amino acid residue. In certain embodiments, a polypeptide variant is distinguished from a reference polypeptide by one or more substitutions, which may be conservative or non-conservative. In certain embodiments, the polypeptide variant comprises conservative substitutions, and, in this regard, it is well understood in the art that some amino acids may be changed to others with broadly similar properties without changing the nature of the activity of the polypeptide. Polypeptide variants also encompass polypeptides in which one or more amino acids have been added or deleted or replaced with different amino acid residues.


The term “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence. The term “expression control (regulatory) sequences” refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.


The term “bind,” “binds,” or “interacts with” refers to a molecule recognizing and adhering to a second molecule in a sample or organism but does not substantially recognize or adhere to other structurally unrelated molecules in the sample. The term “specifically binds,” as used herein with respect to an antibody, refers to an antibody which recognizes a specific antigen, but does not substantially recognize or bind other molecules in a sample. For example, an antibody that specifically binds an antigen from one species may also bind that antigen from one or more species. But, such cross-species reactivity does not itself alter the classification of an antibody as specific. In another example, an antibody that specifically binds an antigen may also bind different allelic forms of the antigen. However, such cross reactivity does not itself alter the classification of an antibody as specific. In some instances, the terms “specific binding” or “specifically binding,” can be used in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds a specific protein structure rather than to any protein. If an antibody is specific for epitope “A,” the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled “A” and the antibody, will reduce the amount of labeled A bound to the antibody.


By “statistically significant,” it is meant that the result was unlikely to have occurred by chance. Statistical significance can be determined by any method known in the art. Commonly used measures of significance include the p-value, which is the frequency or probability with which the observed event would occur if the null hypothesis were true. If the obtained p-value is smaller than the significance level, then the null hypothesis is rejected. In simple cases, the significance level is defined at a p-value of 0.05 or less. A “decreased” or “reduced” or “lesser” amount is typically a “statistically significant” or a physiologically significant amount, and may include a decrease that is about 1.1, 1.2, 1.3, 1.4, 1.5, 1.6 1.7, 1.8, 1.9, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, or 50 or more times (e.g., 100, 500, 1000 times) (including all integers and decimal points in between and above 1, e.g., 1.5, 1.6, 1.7. 1.8, etc.) an amount or level described herein.


The term “stimulation” refers to a primary response induced by binding of a stimulatory molecule (e.g., a TCR/CD3 complex) with its cognate ligand, thereby mediating a signal transduction event, such as signal transduction via the TCR/CD3 complex. Stimulation can mediate altered expression of certain molecules, such as downregulation of TGF-β and/or reorganization of cytoskeletal structures.


The term “stimulatory molecule” refers to a molecule on a T cell that specifically binds a cognate stimulatory ligand present on an antigen-presenting cell. For example, a functional signaling domain derived from a stimulatory molecule is the zeta chain associated with the T cell receptor complex. The stimulatory molecule includes a domain responsible for signal transduction.


The term “stimulatory ligand” refers to a ligand that when present on an antigen-presenting cell (e.g., an APC, a dendritic cell, a B-cell, and the like.) can specifically bind with a cognate binding partner (referred to herein as a “stimulatory molecule”) on a cell, for example, a T cell, thereby mediating a primary response by the T cell, including activation, initiation of an immune response, proliferation, and similar processes. Stimulatory ligands are well-known in the art and encompass, inter alia, an MHC Class I molecule loaded with a peptide, an anti-CD3 antibody, a superagonist anti-CD28 antibody, and a superagonist anti-CD2 antibody.


The term “therapeutic” refers to treatment and/or prophylaxis. A therapeutic effect is obtained by suppression, remission, or eradication of a disease state or alleviating the symptoms of a disease state.


The term “therapeutically effective amount” refers to the amount of the subject compound that will elicit the biological or medical response of a tissue, system, or subject that is being sought by the researcher, veterinarian, medical doctor or another clinician. The term “therapeutically effective amount” includes that amount of a compound that, when administered, is sufficient to prevent the development of, or alleviate to some extent, one or more of the signs or symptoms of the disorder or disease being treated. The therapeutically effective amount will vary depending on the compound, the disease and its severity and the age, weight, etc., of the subject to be treated.


The term “treat a disease” refers to the reduction of the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject.


The term “transfected” or “transformed” or “transduced” refers to a process by which an exogenous nucleic acid is transferred or introduced into the host cell. A “transfected” or “transformed” or “transduced” cell is one which has been transfected, transformed, or transduced with an exogenous nucleic acid. The cell includes the primary subject cell and its progeny.


The term “vector” refers to a polynucleotide that comprises an isolated nucleic acid and which can be used to deliver the isolated nucleic acid to the interior of a cell. Numerous vectors are known in the art including linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses. Thus, the term “vector” includes an autonomously replicating plasmid or a virus. The term also includes non-plasmid and non-viral compounds which facilitate the transfer of nucleic acid into cells, such as, for example, polylysine compounds, liposomes, and the like. Examples of viral vectors include adenoviral vectors, adeno-associated virus vectors, retroviral vectors, and others. For example, lentiviruses are complex retroviruses, which, in addition to the common retroviral genes gag, pol, and env, contain other genes with regulatory or structural function. Lentiviral vectors are well known in the art. Some examples of lentivirus include the Human Immunodeficiency Viruses: HIV-1, HIV-2, and the Simian Immunodeficiency Virus: SIV. Lentiviral vectors have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu, and nef are deleted, making the vector biologically safe.


Ranges: throughout this disclosure, various aspects of the disclosure can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6, etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range.


A “chimeric antigen receptor” (CAR) molecule is a recombinant polypeptide including at least an extracellular domain, a transmembrane domain, and a cytoplasmic domain or intracellular domain. In embodiments, the domains of the CAR are on the same polypeptide chain, for example, a chimeric fusion protein. In embodiments, the domains are on different polypeptide chains, for example, the domains are not contiguous.


The extracellular domain of a CAR molecule includes an antigen binding domain. The antigen binding domain is for expanding and/or maintaining the modified cells, such as a CAR T cell or for killing a tumor cell, such as a solid tumor. In embodiments, the antigen binding domain for expanding and/or maintaining modified cells binds an antigen, for example, a cell surface molecule or marker, on the surface of a WBC. In embodiments, the WBC is at least one of GMP (granulocyte macrophage precursor), MDP (monocyte-macrophage/dendritic cell precursors), cMoP (common monocyte precursor), basophil, eosinophil, neutrophil, SatM (Segerate-nucleus-containing atypical monocyte), macrophage, monocyte, CDP (common dendritic cell precursor), cDC (conventional DC), pDC (plasmacytoid DC), CLP (common lymphocyte precursor), B cell, ILC (Innate Lymphocyte), NK cell, megakaryocyte, myeloblast, promyelocyte, myelocyte, meta-myelocyte, band cells, lymphoblast, prolymphocyte, monoblast, megakaryoblast, promegakaryocyte, megakaryocyte, platelets, or MSDC (Myeloid-derived suppressor cell). In embodiments, the WBC is a granulocyte, monocyte, and or lymphocyte. In embodiments, the WBC is a lymphocyte, for example, a B cell. In embodiments, the WBC is a B cell. In embodiments, the cell surface molecule of a B cell includes CD19, CD22, CD20, BCMA, CD5, CD7, CD2, CD16, CD56, CD30, CD14, CD68, CD11b, CD18, CD169, CD1c, CD33, CD38, CD138, or CD13. In embodiments, the cell surface molecule of the B cell is CD19, CD20, CD22, or BCMA. In embodiments, the cell surface molecule of the B cell is CD19.


The cells described herein, including modified cells such as CAR cells and modified T cells, can be derived from stem cells. Stem cells may be adult stem cells, embryonic stem cells, more particularly non-human stem cells, cord blood stem cells, progenitor cells, bone marrow stem cells, induced pluripotent stem cells, totipotent stem cells or hematopoietic stem cells. A modified cell may also be a dendritic cell, an NK-cell, a B-cell, or a T cell selected from the group consisting of inflammatory T-lymphocytes, cytotoxic T-lymphocytes, regulatory T lymphocytes or helper T-lymphocytes. In embodiments, Modified cells may be derived from the group consisting of CD4+ T lymphocytes and CD8+ T lymphocytes. Prior to expansion and genetic modification of the cells of the invention, a source of cells may be obtained from a subject through a variety of non-limiting methods. T cells may be obtained from a number of non-limiting sources, including peripheral blood mononuclear cells, 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 and known to those skilled in the art may be used. In embodiments, modified cells may be derived from a healthy donor, from a patient diagnosed with cancer or from a patient diagnosed with an infection. In embodiments, a modified cell is part of a mixed population of cells that present different phenotypic characteristics.


A population of cells refers to a group of two or more cells. The cells of the population could be the same, such that the population is a homogenous population of cells. The cells of the population could be different, such that the population is a mixed population or a heterogeneous population of cells. For example, a mixed population of cells could include modified cells comprising a first CAR and cells comprising a second CAR, wherein the first CAR and the second CAR bind different antigens.


The term “stem cell” refers to any of certain types of cell which have the capacity for self-renewal and the ability to differentiate into other kind(s) of a cell. For example, a stem cell gives rise either to two daughter stem cells (as occurs in vitro with embryonic stem cells in culture) or to one stem cell and a cell that undergoes differentiation (as occurs, e.g., in hematopoietic stem cells, which give rise to blood cells). Different categories of stem cells may be distinguished on the basis of their origin and/or on the extent of their capacity for differentiation into other types of cells. For example, stem cells may include embryonic stem (ES) cells (i.e., pluripotent stem cells), somatic stem cells, induced pluripotent stem cells, and any other types of stem cells.


The pluripotent embryonic stem cells are found in the inner cell mass of a blastocyst and have an innate capacity for differentiation. For example, pluripotent embryonic stem cells have the potential to form any type of cell in the body. When grown in vitro for long periods of time, ES cells maintain pluripotency as progeny cells retain the potential for multilineage differentiation.


Somatic stem cells can include fetal stem cells (from the fetus) and adult stem cells (found in various tissues, such as bone marrow). These cells have been regarded as having a capacity for differentiation that is lower than that of the pluripotent ES cells—with the capacity of fetal stem cells being greater than that of adult stem cells. Somatic stem cells apparently differentiate into only a limited number of types of cells and have been described as multipotent. The “tissue-specific” stem cells normally give rise to only one type of cell. For example, embryonic stem cells may be differentiated into blood stem cells (e.g., hematopoietic stem cells (HSCs)), which may be further differentiated into various blood cells (e.g., red blood cells, platelets, white blood cells, etc.).


Induced pluripotent stem cells (i.e., iPS cells or iPSCs) may include a type of pluripotent stem cell artificially derived from a non-pluripotent cell (e.g., an adult somatic cell) by inducing expression of specific genes. Induced pluripotent stem cells are similar to natural pluripotent stem cells, such as embryonic stem (ES) cells, in many aspects, such as the expression of certain stem cell genes and proteins, chromatin methylation patterns, doubling time, embryoid body formation, teratoma formation, viable chimera formation, and potency and differentiability. Induced pluripotent cells can be obtained from adult stomach, liver, skin, and blood cells.


In embodiments, the antigen binding domain for killing a tumor binds an antigen on the surface of a tumor, for example, a tumor antigen or tumor marker. Tumor antigens are proteins that are produced by tumor cells that elicit an immune response, particularly T cell-mediated immune responses. Tumor antigens are well known in the art and include, for example, tumor-associated MUC1 (tMUC1), a glioma-associated antigen, carcinoembryonic antigen (CEA), β-human chorionic gonadotropin, alphafetoprotein (AFP), lectin-reactive AFP, thyroglobulin, RAGE-1, MN-CA IX, human telomerase reverse transcriptase, RU1, RU2 (AS), intestinal carboxyl esterase, mut hsp70-2, M-CSF, prostase, prostate-specific antigen (PSA), PAP, NY-ESO-1, LAGE-1a, p53, prostein, PSMA, Her2/neu, surviving, telomerase, prostate-carcinoma tumor antigen-1 (PCTA-1), MAGE, ELF2M, neutrophil elastase, ephrinB2, CD22, insulin growth factor (IGF)-I, IGF-II, IGF-I receptor, CD19, and mesothelin. For example, when the tumor antigen is CD19, the CAR thereof can be referred to as CD19 CAR or 19CAR, which is a CAR molecule that includes an antigen binding domain that binds CD19.


In embodiments, the extracellular antigen binding domain of a CAR includes at least one scFv or at least a single domain antibody. As an example, there can be two scFvs on a CAR. The scFv includes a light chain variable (VL) region and a heavy chain variable (VH) region of a target antigen-specific monoclonal antibody joined by a flexible linker. Single chain variable region fragments can be made by linking light and/or heavy chain variable regions by using a short linking peptide (Bird et al., Science 242:423-426, 1988). An example of a linking peptide is the GS linker having the amino acid sequence (GGGGS)3 (SEQ ID NO: 278), which bridges approximately 3.5 nm between the carboxy terminus of one variable region and the amino terminus of the other variable region. Linkers of other sequences have been designed and used (Bird et al., 1988, supra). In general, linkers can be short, flexible polypeptides and preferably comprised of about 20 or fewer amino acid residues. The single-chain variants can be produced either recombinantly or synthetically. For synthetic production of scFv, an automated synthesizer can be used. For recombinant production of scFv, a suitable plasmid containing a polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as E. coli. Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides. The resultant scFv can be isolated using standard protein purification techniques known in the art.


The cytoplasmic domain of the CAR molecules described herein includes one or more co-stimulatory domains and one or more signaling domains. The co-stimulatory and signaling domains function to transmit the signal and activate molecules, such as T cells, in response to antigen binding. The one or more co-stimulatory domains are derived from stimulatory molecules and/or co-stimulatory molecules, and the signaling domain is derived from a primary signaling domain, such as the CD3 zeta domain. In embodiments, the signaling domain further includes one or more functional signaling domains derived from a co-stimulatory molecule. In embodiments, the co-stimulatory molecules are cell surface molecules (other than antigens receptors or their ligands) that are required for activating a cellular response to an antigen.


In embodiments, the co-stimulatory domain includes the intracellular domain of CD27, CD28, 4-1BB, OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, or any combination thereof. In embodiments, the signaling domain includes a CD3 zeta domain derived from a T cell receptor.


The CAR molecules described herein also include a transmembrane domain. The incorporation of a transmembrane domain in the CAR molecules stabilizes the molecule. In embodiments, the transmembrane domain of the CAR molecules is the transmembrane domain of a CD28 or 4-1BB molecule.


Between the extracellular domain and the transmembrane domain of the CAR, there may be incorporated a spacer domain. As used herein, the term “spacer domain” generally means any oligo- or polypeptide that functions to link the transmembrane domain to the extracellular domain and/or the cytoplasmic domain on the polypeptide chain. A spacer domain may include up to 300 amino acids, preferably 10 to 100 amino acids, and most preferably 25 to 50 amino acids.


In embodiments, the modified cell comprises a binding molecule, which is a CAR. In embodiments, the CAR comprises an extracellular domain, a transmembrane domain, and an intracellular domain, and the extracellular domain binds a tumor antigen. In embodiments, the intracellular domain comprising a costimulatory domain comprises an intracellular domain of a co-stimulatory molecule selected from the group consisting of CD27, CD28, 4-1BB, OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, and any combination thereof. In embodiments, the intracellular domain comprises a CD3 zeta signaling domain. In embodiments, the CAR is a bispecific CAR or Tan CAR.


In embodiments, the binding molecule is a TCR. In embodiments, the T cell comprises a modified T Cell Receptor (TCR). In embodiments, the TCR is derived from spontaneously occurring tumor-specific T cells in patients. In embodiments, the TCR binds a tumor antigen. In embodiments, the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1. In embodiments, the TCR comprises TCRγ and TCRδ chains, or TCRα and TCRβ chains, or a combination thereof.


In embodiments, the modified cell is derived from tumor-infiltrating lymphocytes (TILs). In embodiments, a T cell clone that expresses a TCR with a high affinity for the target antigen may be isolated. TILs or peripheral blood mononuclear cells (PBMCs) can be cultured in the presence of antigen-presenting cells (APCs) pulsed with a peptide representing an epitope known to elicit a dominant T cell response when presented in the context of a defined HLA allele. High-affinity clones may be then selected on the basis of MHC-peptide tetramer staining and/or the ability to recognize and lyse target cells pulsed with low titrated concentrations of cognate peptide antigen. After the clone has been selected, the TCRα and TCRβ chains or TCRγ and TCRδ chains are identified and isolated by molecular cloning. For example, for TCRα and TCRβ chains, the TCRα and TCRβ gene sequences are then used to generate an expression construct that ideally promotes stable, high-level expression of both TCR chains in human T cells. The transduction vehicle, for example, a gammaretrovirus or lentivirus, can then be generated and tested for functionality (antigen specificity and functional avidity) and used to produce a clinical lot of the vector. An aliquot of the final product can then be used to transduce the target T cell population (generally purified from patient PBMCs), which is expanded before infusion into the patient.


Various methods may be implemented to obtain genes encoding tumor-reactive TCR. More information is provided in Kershaw et al., Clin Transl Immunology. 2014 May; 3(5): e16. In embodiments, specific TCR can be derived from spontaneously occurring tumor-specific T cells in patients. Antigens included in this category include the melanocyte differentiation antigens MART-1 and gp100, as well as the MAGE antigens and NY-ESO-1, with expression in a broader range of cancers. TCRs specific for viral-associated malignancies can also be isolated, as long as viral proteins are expressed by transformed cells. Malignancies in this category include liver and cervical cancer, those associated with hepatitis and papilloma viruses, and Epstein-Barr virus-associated malignancies. In embodiments, target antigens of the TCR include CEA (e.g., for colorectal cancer), gp100, MART-1, p53 (e.g., for melanoma), MAGE-A3 (e.g., melanoma, esophageal and synovial sarcoma), and NY-ESO-1 (e.g., for melanoma and sarcoma as well as multiple myelomas).


In embodiments, preparation and transfusion of tumor-infiltrating lymphocytes (TIL) may be implemented in the following manner. For example, tumor tissue coming from surgical or biopsy specimens can be obtained under aseptic conditions and transported to the cell culture chamber in an icebox. Necrotic tissue and adipose tissue can be removed. The tumor tissue can be cut into small pieces of about 1-3 cubic millimeters. Collagenase, hyaluronidase, and DNA enzyme can be added and digested overnight at 4° C. Filtering with 0.2 um filter, cells can be separated and collected by lymphocyte separation fluid, under 1500 rpm for 5 min. Expanding the cells in a culture medium comprising PHA, 2-mercaptoethanol, and a CD3 monoclonal antibody, and a small dose of IL-2 (10-20 IU/ml) may be added to induce activation and proliferation. The cell density may be carefully measured and maintained within the range of 0.5-2×106/ml for 7-14 days at a temperature of 37° C. with 5% CO2. TIL positive cells having the ability to kill homologous cancer cells can be screened out by co-culture. The TIL positive cells can be amplified in a serum-free medium containing a high dose of IL-2 (5000-6000 IU/ml) until greater than 1×1011 TILs can be obtained. To administer TILs, they are first collected in saline using continuous-flow centrifugation and then filtered through a platelet-administration set into a volume of 200-300 ml containing 5% albumin and 450000 IU of IL-2. The TILs can be infused into patients through a central venous catheter over a period of 30-60 minutes. In embodiments, TILs can be infused in two to four separate bags, and the individual infusions can be separated by several hours.


A bispecific CAR (or tandem CAR (tanCAR)) may include two binding domains: scFv1 and scFv2. In embodiments, scFv1 binds an antigen of a white blood cell (e.g., CD19), and scFv2 binds a solid tumor antigen (e.g., tMUC1). In embodiments, scFv1 binds a solid tumor antigen, and scFv2 binds another solid tumor antigen (e.g., tMUC1 and CLDN 18.2). Claudin18.2 (CLDN 18.2) is a stomach-specific isoform of Claudin-18. CLDN 18.2 is highly expressed in gastric and pancreatic adenocarcinoma. In embodiments, scFv1 binds an antigen expressed on tumor cells but not on normal tissues (e.g., tMUC1); scFv2 binds an antigen expressed on nonessential tissues associated with solid tumor, and the killing of normal cells of the tissue does not cause a life-threatening event (e.g., complications) to the subject (e.g., TSHR, GUCY2C). Examples of the nonessential tissues include organs such as prostate, breast, or melanocyte. In embodiments, scFv1 and scFv2 bind to different antigens that expressed on the same nonessential tissue (e.g., ACPP and SLC45A3 for Prostate cancer, and SIGLEC15 and UPK2 for Urothelial cancer). The sequences of the bispecific CARs and their components may be found in Table 2.















TABLE 2





Variable

Variable

Variable

Variable


domain 1
Linker 1
domain 3
Linker 2
domain 5
Linker 3
domain 7







Anti-TSHR-
3*GGGGS
Anti-TSHR-
4*GGGGS
humanized-
3*GGGGS
humanized-


VL
linker
VH
bispecific
anti CD19-
linker
anti CD19-VL





CAR linker
VH







Anti-TSHR-
3*GGGGS
Anti-TSHR-
4*GGGGS
humanized-
3*GGGGS
humanized-


VH
linker
VL
bispecific
anti CD19-
linker
anti CD19-VH





CAR linker
VL







Tumor-
3*GGGGS
Tumor-
4*GGGGS
anti-CD19-
3*GGGGS
anti-CD19-VH


associated
linker
associated
bispecific
VL
linker



MUC1

MUC1
CAR linker





scFv-1 or 2

scFv-1 or 2






VL

VH









Tumor-
3*GGGGS
Tumor-
4*GGGGS
anti-CD19-
3*GGGGS
anti-CD19-VL


associated
linker
associated
bispecific
VH
linker



MUC1

MUC1
CAR linker





scFv-1 or 2

scFv-1 or 2






VH

VL









humanized- 
3*GGGGS
humanized- 
4*GGGGS
Tumor-
3*GGGGS
Tumor-


anti CD19-
linker
anti CD19-
bispecific
associated 
linker
associated


VH

VL
CAR linker
MUC1

MUC1 scFv-1






scFv-1 or 2

or 2 VH






VL







Tumor-
3*GGGGS
Tumor-
4*GGGGS
Anti-TSHR- 
3*GGGGS
Anti-TSHR-VH


associated
linker
associated
bispecific
VL
linker



MUC1

MUC1
CAR linker





scFv-1 or 2

scFv-1 or 2






VL

VH









Anti-TSHR- 
3*GGGGS
Anti-TSHR-
4*GGGGS
Tumor-
3*GGGGS
Tumor-


VL
linker
VH
bispecific
associated 
linker
associated





CAR linker
MUC1

MUC1 scFv-1






scFv-1 or 2

or 2 VH






VL







Tumor-
3*GGGGS
Tumor-
4*GGGGS
Anti-
3*GGGGS
Anti-GUCY2C-


associated
linker
associated
bispecific
GUCY2C-
linker
VL or VH


MUC1

MUC1
CAR linker
VH or VL




scFv-1 or 2

scFv-1 or 2






VL

VH









Anti-
3*GGGGS
Anti-
4*GGGGS
Tumor-
3*GGGGS
Tumor-


GUCY2C-
linker
GUCY2C-
bispecific
associated 
linker
associated


VH or VL

VL or VH
CAR linker
MUC1

MUC1 scFv-1






scFv-1 or 2

or 2 VH






VL







Tumor-
3*GGGGS
Tumor-
4*GGGGS
Anti-ACPP- 
3*GGGGS
Anti-ACPP-


associated
linker
associated
bispecific
VH or VL
linker
VL or VH


MUC1

MUC1
CAR linker





scFv-1 or 2

scFv-1 or 2






VL

VH









Anti-ACPP- 
3*GGGGS
Anti-ACPP-
4*GGGGS
Tumor-
3*GGGGS
Tumor-


VH or VL
linker
VL or VH
bispecific
associated
linker
associated





CAR linker
MUC1

MUC1 scFv-1






scFv-1 or 2

or 2 VH






VL







Tumor-
3*GGGGS
Tumor-
4*GGGGS
Anti-
3*GGGGS
Anti-


associated 
linker
associated
bispecific 
CLDN18.2-
linker
CLDN18.2-VL


MUC1

MUC1
CAR linker 
VH or VL

or VH


scFv-1 or 2

scFv-1 or 2






VL

VH









Anti-
3*GGGGS
Anti-
4*GGGGS
Tumor-
3*GGGGS
Tumor-


CLDN18.2-
linker
CLDN18.2-
bispecific 
associated
linker
associated


VH or VL

VL or VH
CAR linker 
MUC1

MUC1 scFv-1






scFv-1 or 2

or 2 VH






VL







Tumor-
3*GGGGS
Tumor-
4*GGGGS
Anti-UPK2-
3*GGGGS
Anti-UPK2-


associated 
linker
associated
bispecific 
VH or VL
linker
VL or VH


MUC1

MUC1
CAR linker





scFv-1 or 2

scFv-1 or 2






VL

VH









Anti-UPK2- 
3*GGGGS
Anti-UPK2-
4*GGGGS
Tumor-
3*GGGGS
Tumor-


VH or VL
linker
VL or VH
bispecific 
associated
linker
associated





CAR linker 
MUC1

MUC1 scFv-1






scFv-1 or 2

or 2 VH






VL







Tumor-
3*GGGGS
Tumor-
4*GGGGS
Anti-
3*GGGGS
Anti-


associated 
linker
associated
bispecific 
SIGLEC15-
linker
SIGLEC15-VL


MUC1

MUC1
CAR linker 
VH or VL

or VH


scFv-1 or 2

scFv-1 or 2






VL

VH









Anti-
3*GGGGS
Anti-
4*GGGGS
Tumor-
3*GGGGS
Tumor-


SIGLEC15-
linker
SIGLEC15-
bispecific 
associated
linker
associated


VH or VL

VL or VH
CAR linker
MUC1

MUC1 scFv-1






scFv-1 or 2

or 2 VH






VL





3*(GGGGS) is (GGGGS)3 and 4*(GGGGS) is (GGGGS)4.






Moreover, the present disclosure describes modified cells comprising the nucleic acids or vectors described herein. The cells have been introduced with the nucleic acids or vectors described herein and express at least one or more different antigen binding domains. In embodiments, the cells express one antigen binding domain. In embodiments, the cells include a first antigen binding domain and a second antigen binding domain, wherein the first antigen binding domain binds a cell surface molecule of a WBC, and the second antigen binding domain binds an antigen different from the cell surface molecule of a WBC. In embodiments, the second antigen binding domain binds a tumor antigen. In embodiments, the cells are modified T cells. In embodiments, the modified T cells are CAR T cells including one or more nucleic acids encoding a first antigen binding domain and/or a second antigen binding domain. In embodiments, the modified cells include T cells containing a TCR including the second antigen binding domain.


The methods described herein involve lymphocytes expressing an expansion molecule and a functional molecule. In embodiments, the expansion molecule expands and/or maintains the lymphocytes in a subject, and the function molecule inhibits the growth of or kills a tumor cell in the subject. In embodiments, the expansion molecule and the function molecule are on a single CAR molecule, for example, a bispecific CAR molecule. In embodiments, the expansion molecule and the function molecule are on separate molecules, for example, CAR and TCR or two different CARs. The expansion molecule can include a CAR binding to an antigen associated with blood (e.g., blood cells and blood plasma) or non-essential tissues, and the function molecule can include a CAR or TCR targeting an antigen associated with a tumor cell.


Lymphocyte or T cell response in a subject refers to cell-mediated immunity associated with a helper, killer, regulatory, and other types of T cells. For example, T cell response may include activities such as assisting other WBCs in immunologic processes and identifying and destroying virus-infected cells and tumor cells. T cell response in the subject can be measured via various indicators such as a number of virus-infected cells and/or tumor cells that T cells kill, the amount of cytokines (e.g., IL-6 and IFN-γ) that T cells release in vivo and/or in co-culturing with virus-infected cells and/or tumor cells, indicates a level of proliferation of T cells in the subject, a phenotype change of T cells, for example, changes to memory T cells, and level longevity or lifetime of T cells in the subject.


In embodiments, the method of enhancing T cell response described herein can effectively treat a subject in need thereof, for example, a subject diagnosed with a tumor. The term tumor refers to a mass, which can be a collection of fluid, such as blood, or a solid mass. A tumor can be malignant (cancerous) or benign. Examples of blood cancers include chronic lymphocytic leukemia, acute myeloid leukemia, acute lymphoblastic leukemia, and multiple myeloma.


Solid tumors usually do not contain cysts or liquid areas. The major types of solid malignant tumors include sarcomas and carcinomas. Sarcomas are tumors that develop in soft tissue cells called mesenchymal cells, which can be found in blood vessels, bone, fat tissues, ligament lymph vessels, nerves, cartilage, muscle, ligaments, or tendon, while carcinomas are tumors that form in epithelial cells, which are found in the skin and mucous membranes. The most common types of sarcomas include undifferentiated pleomorphic sarcoma, which involves soft tissue and bone cells; leiomyosarcoma, which involves smooth muscle cells that line blood vessels, gastrointestinal tract, and uterus; osteosarcoma which involves bone cells, and liposarcoma which involves fat cells. Some examples of sarcomas include Ewing sarcoma, Rhabdomyosarcoma, chondrosarcoma, mesothelioma, fibrosarcoma, fibrosarcoma, and glioma.


The five most common carcinomas include adenocarcinoma which involves organs that produce fluids or mucous, such as the breasts and prostate; basal cell carcinoma which involves cells of the outer-most layer of the skin, for example, skin cancer; squamous cell carcinoma which involves the basal cells of the skin; and transitional cell carcinoma which affects transitional cells in the urinary tract which includes the bladder, kidneys, and ureter. Examples of carcinomas include cancers of the thyroid, breast, prostate, lung, intestine, skin, pancreas, liver, kidneys, and bladder, and cholangiocarcinoma.


The methods described herein can be used to treat a subject diagnosed with cancer. Cancer can be a blood cancer or can be a solid tumor, such as a sarcoma or carcinoma. The method of treating includes administering an effective amount of a mixed population of T cells described herein comprising a first antigen binding domain and/or a second antigen binding domain to the subject to provide a T-cell response, wherein the first antigen binding domain binds a cell surface molecule of a WBC, and the second antigen binding domain binds an antigen different from the cell surface molecule of the WBC. In embodiments, enhancing the T cell response in the subject includes selectively enhancing proliferation of T cell expressing the first antigen binding domain and the second antigen binding domain in vivo.


The present disclosure describes pharmaceutical compositions. The pharmaceutical compositions include one or more of the following: CAR molecules, TCR molecules, modified CAR T cells, modified cells comprising CAR or TCR, mix population of modified cells, nucleic acids, and vectors described herein. Pharmaceutical compositions are 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, although appropriate dosages may be determined by clinical trials.


The term “pharmaceutically acceptable” means approved by a regulatory agency of the U.S. Federal or a state government or the EMA (European Medicines Agency) or listed in the U.S. Pharmacopeia Pharmacopeia (United States Pharmacopeia-33/National Formulary-28 Reissue, published by the United States Pharmacopeia Convention, Inc., Rockville Md., publication date: April 2010) or other generally recognized pharmacopeia for use in animals, and more particularly in humans.


The term “carrier” refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered. Pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origins, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and giyceroi solutions can also be employed as liquid carriers, particularly for injectable solutions.


The present disclosure also describes a pharmaceutical composition comprising the first and the second population of cells described herein. The pharmaceutical composition described herein, comprising a first population of cells comprising a first antigen binding molecule and a second population of cells comprising a second antigen binding domain, are suitable for cancer therapy. For example, the binding of the first antigen binding molecule with an antigen enhances the expansion of the cells suitable for cancer therapy.


When “an immunologically effective amount,” “an anti-tumor effective amount,” “a tumor-inhibiting effective amount,” or “a therapeutically effective amount” is indicated, 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, the extent of infection or metastasis, and condition of the patient (subject). It can be stated that a pharmaceutical composition comprising the modified cells described herein may be administered at a dosage of 104 to 109 cells/kg body weight, preferably 105 to106 cells/kg body weight, including all integer values within those ranges. Modified cell compositions may 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 may be desired to administer activated T cells to a subject and then subsequently redraw the blood (or have apheresis performed), collect the activated and expanded T cells, 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 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 protocols, may select out certain populations of T cells.


The administration of the pharmaceutical compositions described herein may be carried out in any convenient manner, including by aerosol inhalation, injection, ingestion, transfusion, implantation, or transplantation. The compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous (i. v.) injection, or intraperitoneally. In embodiments, the modified cell compositions described herein are administered to subjects by intradermal or subcutaneous injection. In embodiments, the T cell compositions of the present disclosure are administered by i.v. injection. The compositions of modified cells may be injected directly into a tumor, lymph node, or site of infection. In embodiments, cells activated and expanded using the methods described herein, or other methods known in the art where T cells are expanded to therapeutic levels, are administered to patients in conjunction with (e.g., before, simultaneously or following) any number of relevant treatment modalities, for example as a combination therapy, including but not limited to treatment with agents for antiviral therapy, cidofovir, and interleukin-2, Cytarabine (also known as ARA-C); or natalizumab treatment for MS patients; or efalizumab treatment for psoriasis patients or other treatments for PML patients. In further embodiments, the T cells described herein can be used in combination with chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAM PATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludaribine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, cytokines, 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; Isoniemi (supra)). In embodiments, the cell compositions described herein are administered to a subject in conjunction with (e.g., before, simultaneously or following) bone marrow 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 embodiments, the cell compositions described herein are administered following B-cell ablative therapy. For example, agents that react with CD20, e.g., Rituxan, may be administered to patients. In embodiments, subjects may undergo standard treatment with high dose chemotherapy, followed by peripheral blood stem cell transplantation. In certain embodiments, following the transplant, subjects receive an infusion of the expanded immune cells of the present disclosure. In embodiments, expanded cells are administered before or following surgery. The dosage of the above treatments to be administered to a subject in need thereof 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 by a physician depending on various factors. Additional information on the methods of cancer treatment using modified cells is provided in U.S. Pat. No. 8,906,682, incorporated by reference in its entirety.


Embodiments described herein relate to an in vitro method for preparing modified cells. The method may include obtaining a sample of cells from a subject. For example, the sample may include T cells or T cell progenitors. The method may further include transfecting the sample of cells with a DNA encoding at least a CAR and culturing the sample of cells ex vivo in a medium that selectively enhances proliferation of CAR-expressing T cells. The sample of cells can be a mixed population of modified cells described herein.


In embodiments, the sample is a cryopreserved sample. In embodiments, the sample of cells is from umbilical cord blood or a peripheral blood sample from the subject. In embodiments, the sample of cells is obtained by apheresis or venipuncture. In embodiments, the sample of cells is a subpopulation of T cells.


Embodiments relate to a modified cell engineered to express an antigen binding molecule, wherein the expression and/or function of one or more genes in the modified cell has been enhanced. In embodiments, the one or more genes comprise at least one of BATF, HMGA1, STAT5A, ZNF580, GLMP, JAZF1, RUNX1, ZGPAT, ZNF511, GTF2IRD2B, ATF4, MBD4, TBPL1, GTF2B, RBCK1, ZBTB38, PIN1, DRAP1, THYN1, HSF1, PRDM1, ZNF428, NFYC, and ZNF706. In embodiments, the one or more genes are HMGA1 and/or ZBTB38.


Embodiments relate to a modified cell engineered to express an antigen binding molecule, wherein the expression and/or function of one or more genes in the modified cell has been reduced or eliminated. In embodiments, the one or more genes comprise at least one of GTF3A, JUN, IRF1, JUNB, TMF1, ELF1, AKNA, BCL11B, KLF2, ZNF292, RORA, HMGN3, KDM2A, ASCL2, SP140L, NFATC2, RUNX3, NFE2L2, KLF6, MTERF4, PHF20, RELB, MAZ, ARID5A, REL, ZEB2, ARID5B, KLF3, CREM, ZNF207, IRF7, DR1, SP140, BBX, MECP2, STAT4, ZBTB1, CREBZF, NFATC3, GPBP1, IKZF1, SON, ZNF800, STAT3, STATE, CGGBP1, FOXN2, DNMT1, SP100, GATA3, EOMES, YY1, SP110, SAFB, REST, NR3C1, FOXN3, ELF2, GTF2I, BAZ2A, ZNF683, STAT1, BHLHE40, ZNF276, ETS1, NFAT5, BPTF, KMT2A, FOS, PA2G4, IKZF3, ZNF148, CDC5L, CREB1, HBP1, ZNF721, KAT7, SP4, ZC3H8, AKAP8L, ZNF326, ZNF451, ZNF131, CEBPZ, TOPORS, ZNF33A, NCOA3, STAT2, DDIT3, ZNF217, KLF9, CSRNP1, NCOA1, SAFB2, ZNF107, ZFX, E2F4, HIF1A, ZNF480, CTCF, ZBTB44, NCOA2, ZHX1, ZNF644, ASH1L, STAT5B, AEBP2, MYSM1, ZNF91, CEBPB, MXD4, YBX3, RLF, JUND, ZNF600, SMAD4, TET2, ZNF267, PRDM2, ZBTB7A, THAP12, ETV3, NFKB2, KLF13, SATB1, ZNF791, RBPJ, SPEN, PURA, ZNF507, FOSL2, IRF8, ELK4, ATF3, KCMF1, ZNF639, SKI, FOXO1, NR4A2, ZNF331, NFKB1, CEBPD, FOSB, SKIL, NR4A3, and NR4A1. In embodiments, the one or more genes are AKNA.


In embodiments, overexpression of HMGA1 may increase the expansion ability of T cells and inhibit the T cell Andrea Conte1, Cell Death, and Differentiation. At the same time, HMGA1 may promote the secretion of IL2 by T cells and the release of IFNγ. HMGA1 may inhibit autophagy and enhances mitochondrial function, thereby promoting phosphorylation of phosphorylation and providing T cells with More energy. Thus, overexpression of this gene may enhance the function of CAR-T. In embodiments, reduced expression of AKNA gene may promote the release of immune cell factors and enhance the inflammatory response and enhance the killing ability of CAR-T.


In embodiments, overexpression of one or more genes in a modified cell may be implemented by introducing a polynucleotide encoding the one or more genes. In embodiments, the overexpression of the one or more genes in the modified cell may be regulated by a transcription modulator, which is or includes Hif1a, NFAT, FOXP3, and/or NFkB. A promoter comprising one or more binding sites for NFAT responsive elements, such as NFAT1, NFAT2, NFAT3, and/or NFAT4. “NFAT promoter” refers to one or more NFAT responsive elements linked to a minimal promoter of any gene expressed by T-cells. In embodiments, the minimal promoter of a gene expressed by T-cells is a minimal human IL-2 promoter. The NFAT responsive elements may comprise, e.g., NFAT1, NFAT2, NFAT3, and/or NFAT4 responsive elements. The NFAT promoter (or a functional portion or functional variant thereof) may comprise any number of binding motifs, e.g., at least two, at least three, at least four, at least five, or at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, or up to twelve binding motifs. In embodiments, the NFAT promoter comprises six NFAT binding motifs. In an especially preferred embodiment, the NFAT promoter nucleotide sequence comprises or consists of SEQ ID NO: 63 or a functional portion or functional variant thereof. The NFAT promoter (or a functional portion or functional variant thereof) is operatively associated with the nucleotide sequence encoding the one or more genes (or a functional portion or functional variant thereof). “Operatively associated with” means that the nucleotide sequence encoding the one or more genes (or a functional portion or functional variant thereof) is transcribed into the one or more genes mRNA when the NFAT protein binds to the NFAT promoter sequence (or a functional portion or functional variant thereof). Without being bound to a particular theory, it is believed that NFAT is regulated by a calcium signaling pathway. In particular, it is believed that TCR stimulation (by, e.g., an antigen) and/or stimulation of the calcium signaling pathway of the cell (by, e.g., PMA/lonomycin) increases intracellular calcium concentration and activates calcium channels. It is believed that the NFAT protein is then dephosporylated by calmoduin and translocates to the nucleus where it binds with the NFAT promoter sequence (or a functional portion or functional variant thereof) and activates downstream gene expression. By providing an NFAT promoter (or a functional portion or functional variant thereof) that is operatively associated with the nucleotide sequence encoding the one or more genes (or a functional portion or functional variant thereof), the nucleic acids of the invention advantageously make it possible to express the one or more genes (or a functional portion or functional variant thereof) only when the host cell including the nucleic acid is stimulated by, e.g., PMA/lonomycin and/or an antigen. More information can be found at U.S. Pat. No. 8,556,882, which is incorporated by the reference.


In embodiments, the antigen binding molecule is chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain. In embodiments, the antigen-binding domain binds to a tumor antigen is selected from a group consisting of: TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGSS, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LACE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin, telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin 61, MYCN, RhoC, TRP-2, CYP161, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1. In embodiments, the intracellular signaling domain comprises a costimulatory signaling domain, or a primary signaling domain and a costimulatory signaling domain, wherein the costimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-166 (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D.


In embodiments, the antigen binding molecule is a modified TCR. In embodiments, the TCR is derived from spontaneously occurring tumor-specific T cells in patients. In embodiments, the TCR binds to a tumor antigen. In embodiments, the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1. In embodiments, the TCR comprises TCRγ and TCRδ Chains or TCRα and TCRβ chains, or a combination thereof.


In embodiments, the cell is an immune effector cell (e.g., a population of immune effector cells). In embodiments, the immune effector cell is a T cell or an NK cell. In embodiments, the immune effector cell is a T cell. In embodiments, the T cell is a CD4+ T cell, a CD8+ T cell, or a combination thereof. In embodiments, the cell is a human cell.


In embodiments, the modified cell comprises an inhibitor of expression or function of the one or more genes. In embodiments, the inhibitor is (1) a gene-editing system targeted to one or more sites within the gene encoding the one or more genes or a corresponding regulatory elements; (2) nucleic acid encoding one or more components of a gene-editing system of the one or more genes; or (3) combinations thereof.


Embodiments relate to a pharmaceutical composition comprising the population of the cells above. Embodiments relate to a method of cause T cell response in a subject in need thereof and/or treating a tumor of the subject, the method comprising administering an effective amount of the composition.


Embodiments relate to a method of producing T cells exhibiting an enhanced memory T cell phenotype, the method comprising: modulating a population of T cells to enhance the expression and/or function of HMGY. For example, the method may include introducing a polynucleotide encoding HMGY into a population of T cells, wherein expression of HMGY is higher as compared to T cells that are not introduced with the polynucleotide, and the memory T cell phenotype of the population of T cells is enhanced as compared to T cells that are not introduced with the polynucleotide. In embodiments, the method may include introducing a polynucleotide encoding one or more genes associated with HMGY, for example, upstream or downstream of the signaling pathway associated with HMGY and/or a transcription factor associated with HMGY.


Embodiments relate to a method of producing T cells exhibiting an enhanced memory T cell phenotype, the method comprising: introducing a polynucleotide encoding HMGY into a population of T cells, wherein expression of HMGY is higher as compared to T cells that are not introduced with the polynucleotide, and the memory T cell phenotype of the population of T cells is enhanced as compared to T cells that do not include the polynucleotide.


In embodiments, the population of T cells exhibiting an increased gene expression level in CD62L and/or CCR7 as compared to T cells that are not introduced with the polynucleotide.


In embodiments, the method further comprises culturing the population; and measuring cell expansion of the population of T cells. In embodiments, expansion of the population of T cells is enhanced as compared to T cells that are not introduced with the polynucleotide.


In embodiments, the polynucleotide comprises the amino acid of SEQ ID NO: 61, and HMGY is overexpressed.


The method further comprises contacting the population of T cells with an antigen that the population of T cells bind. In embodiments, the population of T cells exhibiting a reduced gene expression level in CD137 and/or KLRG as compared to T cells that are not introduced with the polynucleotide.


In embodiments, the enhanced memory T cell phenotype comprises an increased gene expression level in CD62L and/or CCR7. In embodiments, the enhanced memory T cell phenotype comprises a reduced gene expression level in CD137 and/or KLRG.


As used herein, the term “memory T-cells” or TCM, refers to a subgroup or subpopulation of T-cells that express a higher level of genes associated with trafficking to secondary lymphoid organs, including CD62L and/or CCR7. In embodiments, memory T cells express a lower level of genes including CD137 and/or KLRG.


HMGY, HMGA1, or HMG-I/Y may be used interchangeably and refers to bind preferentially to the minor groove of A+T rich regions in double-stranded DNA. It is suggested that these proteins could function in nucleosome phasing and in the 3′-end processing of mRNA transcripts. They are also involved in the transcription regulation of genes containing, or in close proximity to A+T-rich regions. The three known members of the HMGI(Y) family of high-mobility group (HMG) mammalian nonhistone nuclear proteins (HMG-I, HMG-Y, and HMGI-C) are thought to participate in numerous biological processes (transcription, replication, retroviral integration, genetic recombination, etc.) by virtue of their ability to recognize and alter the structure of both DNA and chromatin substrates. More information on HMGY can be found at US Patent Publication NO: US2015315589, which is incorporated herein by its reference.


In embodiments, the population of T cells may comprise an antigen binding molecule. In embodiments, the cell is a human cell.


In embodiments, the antigen binding molecule is chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain. In embodiments, the antigen-binding domain binds to a tumor antigen is selected from a group consisting of: TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGSS, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRCSD, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LAGE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin, telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin B1, MYCN, RhoC, TRP-2, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1. In embodiments, the intracellular signaling domain comprises a co-stimulatory signaling domain, or a primary signaling domain and a co-stimulatory signaling domain, wherein the co-stimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D.


In embodiments, the antigen binding molecule is a modified TCR. In embodiments, the TCR is derived from spontaneously occurring tumor-specific T cells in patients. In embodiments, the TCR binds to a tumor antigen. In embodiments, the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1. In embodiments, the TCR comprises TCRγ and TCRδ Chains or TCRα and TCRβ chains, or a combination thereof.


The present disclosure is further described by reference to the following exemplary embodiments and examples. These exemplary embodiments and examples are provided for purposes of illustration only and are not intended to be limiting unless otherwise specified. Thus, the present disclosure should in no way be construed as being limited to the following exemplary embodiments and examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.


Exemplary Embodiments

The following are exemplary embodiments:

  • 1. A modified cell engineered to express an antigen binding molecule, wherein expression and/or function of one or more genes in the modified cell has been enhanced.
  • 2. The modified cell of embodiment 1, wherein the one or more genes comprise at least one of BATF, HMGA1, STAT5A, ZNF580, GLMP, JAZF1, RUNX1, ZGPAT, ZNF511, GTF2IRD2B, ATF4, MBD4, TBPL1, GTF2B, RBCK1, ZBTB38, PIN1, DRAP1, THYN1, HSF1, PRDM1, ZNF428, NFYC, and ZNF706.
  • 3. The modified cell of embodiment 1, wherein the one or more genes are HMGA1 and/or ZBTB38.
  • 4. A modified cell engineered to express an antigen binding molecule, wherein expression and/or function of one or more genes in the modified cell has been reduced or eliminated.
  • 5. The modified cell of embodiment 4, wherein the one or more genes comprise at least one of GTF3A, JUN, IRF1, JUNB, TMF1, ELF1, AKNA, BCL11B, KLF2, ZNF292, RORA, HMGN3, KDM2A, ASCL2, SP140L, NFATC2, RUNX3, NFE2L2, KLF6, MTERF4, PHF20, RELB, MAZ, ARID5A, REL, ZEB2, ARID5B, KLF3, CREM, ZNF207, IRF7, DR1, SP140, BBX, MECP2, STAT4, ZBTB1, CREBZF, NFATC3, GPBP1, IKZF1, SON, ZNF800, STAT3, STATE, CGGBP1, FOXN2, DNMT1, SP100, GATA3, EOMES, YY1, SP110, SAFB, REST, NR3C1, FOXN3, ELF2, GTF2I, BAZ2A, ZNF683, STAT1, BHLHE40, ZNF276, ETS1, NFATS, BPTF, KMT2A, FOS, PA2G4, IKZF3, ZNF148, CDC5L, CREB1, HBP1, ZNF721, KAT7, SP4, ZC3H8, AKAP8L, ZNF326, ZNF451, ZNF131, CEBPZ, TOPORS, ZNF33A, NCOA3, STAT2, DDIT3, ZNF217, KLF9, CSRNP1, NCOA1, SAFB2, ZNF107, ZFX, E2F4, HIF1A, ZNF480, CTCF, ZBTB44, NCOA2, ZHX1, ZNF644, ASH1L, STAT5B, AEBP2, MYSM1, ZNF91, CEBPB, MXD4, YBX3, RLF, JUND, ZNF600, SMAD4, TET2, ZNF267, PRDM2, ZBTB7A, THAP12, ETV3, NFKB2, KLF13, SATB1, ZNF791, RBPJ, SPEN, PURA, ZNF507, FOSL2, IRF8, ELK4, ATF3, KCMF1, ZNF639, SKI, FOXO1, NR4A2, ZNF331, NFKB1, CEBPD, FOSB, SKIL, NR4A3, and NR4A1.
  • 6. The modified cell of embodiment 4, wherein the one or more genes are AKNA.
  • 7. The modified cell of one of embodiments 1-6, wherein the antigen binding molecule is chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain.
  • 8. The modified cell of embodiment 7, wherein the antigen-binding domain binds to a tumor antigen is selected from a group consisting of: TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGS5, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LACE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin, telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin B1, MYCN, RhoC, TRP-2, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.
  • 9. The modified cell of one of embodiments 7 and 8, wherein the intracellular signaling domain comprises a costimulatory signaling domain, or a primary signaling domain and a costimulatory signaling domain, wherein the costimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D.
  • 10. The modified cell of one of embodiments 1-6, wherein the antigen binding molecule is a modified TCR.
  • 11. The modified cell of embodiment 10, wherein the TCR is derived from spontaneously occurring tumor-specific T cells in patients.
  • 12. The modified cell of embodiment 10, wherein the TCR binds to a tumor antigen.
  • 13. The modified cell of embodiment 12, wherein the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1.
  • 14. The modified cell of embodiment 10, wherein the TCR comprises TCRγ and TCRδ Chains or TCRα and TCRβ chains, or a combination thereof.
  • 15. The modified cell of any of the preceding embodiments, wherein the cell is an immune effector cell (e.g., a population of immune effector cells).
  • 16. The modified cell of embodiment 15, wherein the immune effector cell is a T cell or an NK cell.
  • 17. The modified cell of embodiment 15, wherein the immune effector cell is a T cell.
  • 18. modified cell of embodiment 15, wherein the T cell is a CD4+ T cell, a CD8+ T cell, or a combination thereof.
  • 19. The modified cell of any of the preceding embodiments, wherein the cell is a human cell.
  • 20. The modified cell of any of the preceding embodiments, wherein the modified cell comprises an inhibitor of expression or function of the one or more genes.
  • 21. The modified cell of embodiment 20, wherein the inhibitor is (1) a gene-editing system targeted to one or more sites within the gene encoding the one or more genes or a corresponding regulatory elements; (2) nucleic acid encoding one or more components of a gene-editing system of the one or more genes; or (3) combinations thereof.
  • 22. A pharmaceutical composition comprising the population of the cells of any of embodiments 1-21.
  • 23. A method of cause T cell response in a subject in need thereof and/or treating a tumor of the subject, the method comprising administering an effective amount of the composition of embodiment 22 to the subject.
  • 24. A method of modulating activities of T cells, the method comprising: introducing a polynucleotide encoding HMGY into a population of T cells.
  • 25. A method of producing T cells exhibiting an enhanced memory T cell phenotype, the method comprising: introducing a polynucleotide encoding HMGY into a population of T cells as compared to T cells that are not introduced with the polynucleotide.
  • 26. A method of producing T cells exhibiting a reduced activation level and/or a reduced differentiation level in the presence of an antigen the T cells binds, the method comprising: introducing a polynucleotide encoding HMGY into a population of T cells as compared to T cells that are not introduced with the polynucleotide.
  • 27. A method of enhancing the expansion of T cells in response to the presence of an antigen that the T cell bind, the method comprising: introducing a polynucleotide encoding HMGY into a population of T cells as compared to T cells that are not introduced with the polynucleotide.
  • 28. A method of producing T cells, the method comprising enhancing HMGY gene expression and/or function of the T cells as compared to T cells that do not include enhanced HMGY gene expression and/or function.
  • 29. The method of any preceding embodiments, wherein the population of T cells exhibiting an increased gene expression in CD62L and/or CCR7 as compared to T cells that are not introduced with the polynucleotide or enhanced HMGY gene expression and/or function.
  • 30. The method of any preceding embodiments, wherein the population of T cells exhibiting a reduced gene expression in CD137 and/or KLRG as compared to T cells that are not introduced with the polynucleotide or do not include enhanced HMGY gene expression and/or function
  • 31. The method of any preceding embodiments, further comprising: culturing the population of T cells; and measuring cell expansion of the population of T cells.
  • 32. The method of any preceding embodiments, further comprising: contacting the population of T cells with an antigen that the population of T cells bind.
  • 33. The method of any suitable preceding embodiments, wherein the enhanced memory T cell phenotype comprises a reduced gene expression in CD137 and/or KLRG, or the enhanced memory T cell phenotype comprises an increased gene expression in CD62L and/or CCR7.
  • 34. The population T cells produced using the method of any preceding embodiments.
  • 35. A modified cell engineered to express an antigen binding molecule, wherein expression and/or function of one or more genes in the modified cell has been enhanced.
  • 36. The modified cell of embodiment 35, wherein the one or more genes are HMGA1 and/or ZBTB38 (SEQ ID NO: 62).
  • 37. The modified cell of embodiment 35, wherein the modified cell exhibits an increased gene expression in CD62L and/or CCR7 as compared to a cell that does not include enhanced HMGY gene expression and/or function.
  • 38. The modified cell of embodiment 35, wherein the modified cell exhibits a reduced gene expression in CD137and/or KLRG as compared to a cell that does not include enhanced HMGY gene expression and/or function.
  • 39. The method or modified cell of any preceding embodiments, wherein the modified cell or the population of T cells are engineered to express an antigen binding molecule.
  • 40. The modified cell or the population of T cells of any preceding embodiments, wherein the antigen binding molecule is chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain.
  • 41. The modified cell or the population of T cells of embodiment 40, wherein the antigen-binding domain binds to a tumor antigen is selected from a group consisting of: TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGS5, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LACE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin, telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin 61, MYCN, RhoC, TRP-2, CYP161, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.
  • 42. The modified cell or the population of T cells of embodiment 40 of 19, wherein the intracellular signaling domain comprises a co-stimulatory signaling domain, or a primary signaling domain and a co-stimulatory signaling domain, wherein the co-stimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-166 (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D.
  • 43. The modified cell or the population of T cells of any suitable preceding embodiments, wherein the antigen binding molecule is a modified TCR.
  • 44. The modified cell or the population of T cells of embodiment 43, wherein the TCR is derived from spontaneously occurring tumor-specific T cells in patients.
  • 45. The modified cell or the population of T cells of embodiment 43, wherein the TCR binds to a tumor antigen.
  • 46. The modified cell or the population of T cells of embodiment 45, wherein the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1.
  • 47. The modified cell or the population of T cells of embodiment 43, wherein the TCR comprises TCRγ and TCRδ Chains or TCRα and TCRβ chains, or a combination thereof.
  • 26. The modified cell of any of the preceding embodiments, wherein the modified cell is an immune effector cell (e.g., a population of immune effector cells).
  • 48. The modified cell of embodiment 26, wherein the immune effector cell is a T cell or an NK cell.
  • 49. The modified cell of embodiment 48, wherein the immune effector cell is a T cell.
  • 50. the modified cell of embodiment 48, wherein the T cell is a CD4+ T cell, a CD8+ T cell, or a combination thereof.
  • 51. The modified cell or the population of T cells of any of the preceding embodiments, wherein the cell is a human cell.
  • 52. The modified cell or the population of T cells of the preceding embodiments, wherein the modified cell comprises an inhibitor of expression or function of the one or more genes.
  • 53. The modified cell or the population of T cells of embodiment 52, wherein the inhibitor is (1) a gene-editing system targeted to one or more sites within the gene encoding the one or more genes or a corresponding regulatory elements; (2) nucleic acid encoding one or more components of a gene-editing system of the one or more genes; or (3) combinations thereof.
  • 54. A pharmaceutical composition comprising the population of the cells of any of preceding embodiments.
  • 55. A method of delivering the therapeutic agent, the method comprising administering an effective amount of the composition of embodiment 54 to the subject, or a method of causing or eliciting T cell response in a subject in need thereof and/or treating a tumor of the subject, the method comprising administering an effective amount of the composition of embodiment 54 to the subject.
  • 56. The modified cell, the method, the pharmaceutical composition, the cell of one of embodiments 24-55, wherein the one or more polynucleotides are present in the modified cell in a recombinant DNA construct, in an mRNA, or in a viral vector.
  • 57. The modified cell, the method, the pharmaceutical composition, the cell of embodiment 56, wherein the nucleic acid sequence is an mRNA, which is not integrated into the genome of the modified cell.
  • 58. The modified cell, the method, the pharmaceutical composition, the cell of one of embodiments 55-57, wherein the one or more polynucleotides are associated with an oxygen-sensitive polypeptide domain.
  • 59. The modified cell, the method, the pharmaceutical composition, the cell of embodiment 58, wherein the oxygen-sensitive polypeptide domain comprises HIF VHL binding domain.
  • 60. The modified cell, the method, the pharmaceutical composition, the cell of one of embodiments 55-59, wherein expression of the one or more polynucleotide are regulated by a promoter comprising a binding site for a transcription modulator that modulates the expression and/or secretion of the therapeutic agent in the cell.
  • 61. The modified cell, the method, the pharmaceutical composition, the cell of embodiment 60, wherein the transcription modulator is or includes Hif1a, NFAT, FOXP3, and/or NFkB.
  • 62. The modified cell of any preceding embodiments (24-61), wherein expression the one or more polynucleotide, is regulated by NFAT such that the EV is assembled in response to activation of the modified cell.
  • 43. A polynucleotide comprising a binding site of a transcription modulator (e.g., NFAT) and encoding one or more proteins assembling the extracellular vesicle (EV) and the therapeutic agent.
  • 63. The modified cell, the method, the pharmaceutical composition, the cell of any of embodiments 24-43, wherein the one or more proteins are self-assembling proteins.
  • 64. The modified cell, the method, the pharmaceutical composition, the cell of any of embodiments 24-44 wherein the one or more proteins that direct their release through vesicles as a luminal membrane-bound protein is chosen from the group consisting of: the retroviral group-specific antigen, retroviral group-specific antigen variations, the influenza MI protein, the ARRDCI protein, the ARC protein, the Ebola virus VP40 protein and the M proteins of vesicular stomatitis virus.
  • 65. The modified cell, the method, the pharmaceutical composition, the cell of any of embodiments 24-64, wherein the one or more proteins comprise an Arc protein and the one or more polynucleotides comprise a nucleic acid encoding a therapeutic agent.
  • 66. An EV comprising an Arc protein and a nucleic acid encoding or comprising a therapeutic agent, the nucleic acid is DNA or RNA encoding the therapeutic agent.
  • 67. The modified cell, the method, the pharmaceutical composition, the cell, or the EV of any of embodiments 65 and 66, wherein the therapeutic agent is selected from the group consisting of a siRNA, an shRNA, and RNAi.
  • 68. The modified cell, the method, the pharmaceutical composition, the cell, or the EV of any of embodiments 65 and 66, wherein the nucleic acid encoding a therapeutic agent is linked to a 3′ UTR sequence.
  • 69. The modified cell, the method, the pharmaceutical composition, the cell, or the EV of embodiment 68, wherein the 3′ UTR sequence is bound to said Arc protein.
  • 70. The modified cell, the method, the pharmaceutical composition, the cell, or the EV of embodiment 69, wherein the 3′ UTR sequence is an arc mRNA 3′ UTR sequence.
  • 71. The modified cell, the method, the pharmaceutical composition, the cell, or the EV of any of embodiments 65 and 70 wherein the nucleic acid further comprises a transcription modulator sequence.
  • 72. The modified cell, the method, the pharmaceutical composition, the cell, or the EV of any of embodiments 24-71, wherein the therapeutic agent is scFv binding a tumor antigen on a membrane or inside of a tumor cell.
  • 73. The modified cell, the method, the pharmaceutical composition, the cell, or the EV of embodiment 72, wherein the tumor antigen is at least one of the tumor antigens of embodiments listed in the disclosure.


EXAMPLES
Expression of CAR and Modified PD-1 on Primary T Cells

Primary T cells were obtained from patients. The obtained primary T cells were transduced with lentiviral vectors. Flow cytometry was performed and analyzed to determine the expression of CAR and various modified PD-1 variants in primary T cells. Techniques related to cell cultures, construction of lentiviral vectors, flow cytometry, and other related techniques are provided in U.S. Pat. No. 9,572,837, assigned to Innovative Cellular Therapeutics Co., Ltd., and incorporated herein by reference in its entirety. Sequences described in the disclosure may be found at Table 7. Additional information of the sequences may be found in PCT Patent Publications WO2020106843 and WO2019140100 and in PCT Patent Application NO: PCT/US20/13099, which are incorporated herein by reference in their entirety.


Cells Expressing Chimeric Receptors Establish Antitumor Effects in Patients with Relapsed/Refractory NHL

This clinical trial was designed to assess the safety and efficacy of infusing autologous T cells modified to express humanized CD19 specific CAR/4-1BB/CD3-ζ and modified PD-1 (SEQ ID NO: 37 of which the intracellular domain comprises SEQ ID NO: 36) into patients with Relapsed/Refractory (R/R) Non-Hodgkin's Lymphoma (NHL). The inclusion criteria were as follows: 1) age not more than 60 years; 2) relapsed or refractory CD19+ NHL, and 3) measurable disease and adequate performance status and organ function. Patients with central nervous system leukemia (CNSL) were ineligible. The protocol was approved by Hospitals and their Institutional Review Boards. All patients provided written informed consent.


Prior to CD19 CART cell infusion, FACS analysis of transduction efficiency and in vitro cytotoxicity assays of CD19 CAR T cells were performed for each patient as described herein. Additionally, CD19 CART cell cultures were checked twice for possible contaminations by a fungus, bacteria, mycoplasma, chlamydia, and endotoxin. The levels of IFN-γ, TNF-α, IL-4, IL-6, IL-10, IL-17, and other cytokines in serum and cerebral spinal fluid (CSF) were measured in a multiplex format according to the manufacturer's instructions.


TCR Clonal Enrichment in a Clinical Trial

In clinical trials, a highly enriched T cell clone was found by TCR sequencing in a patient that expanded from 9% to 74.92% in the peripheral blood 4 days after infusion. The patient received an CD19 CART cell infusion for treating NHL and achieved complete remission (CR). Single-cell RNA sequencing analysis was performed and the result was compared with the T cells and other groups. It was found that the expression of GZMB, PRF, and other tumor killing genes was significantly up-regulated, and the genes related to exhausted T cells, such as TIGIT, were significantly down-regulated. Subsequently, the differentially expressed transcription factors of the T cells was analyzed and correlation analysis was used to find that many of the up-regulated genes promoted the expansion and function of T cells, while down-regulating genes suppressed the T cells' functions. Therefore, it is proposed that knocking out or overexpressing genes in the CAR T cell can help improve the function of CAR T cells.


As shown in FIGS. 1 and 2, a TCR clone was amplified from 9% to 74.92% of total TCR clones in the peripheral blood of the patient in four days (FIG. 2, clinical sample number AYTPB0306, AYTPB0310). To further explore the reasons, single-cell RNA sequencing was performed. Clinical protocols may be found in PCT Patent Publication WO2020106843, which is incorporated herein by reference in its entirety.


Enriched TCR Clones Show Enhanced Efficacy

The killing of T cells is mainly through granzyme B and porins (corresponding genes GZMB, PRF1). In the sequencing results of single-cell RNA, the expression levels of the enriched clones are significantly increased. Table 3 shows the differential expression of killing-related genes in highly enriched clones (unit, UMI).














TABLE 3







Gene
Mean in TRBV9
Mean in others
p-value









PRF1
2.66
2.34
2.19E−33



GZMB
3.11
2.75
9.62E−46










Significant Down-Regulation of Genes Associated with Enriched Clonal Depletion

After T cell activation, due to negative regulation and the immune microenvironment produced by cancer cells, and T cells are exhausted, a representative gene (TIGIT) related to exhausted T cells appears to be significantly down-regulated in the enriched clones (TRBV9: 0.2 UMI, Others: 0.4) UMI, p-value=8.77E-23), which further demonstrates that the enriched cells have strong anti-tumor capabilities.


Overexpression and Knockout of Transcription Factors Screened by Single-Cell Sequencing have Significant Effects on T Cells

Based on the analysis of single-cell sequencing data, the differentially expressed genes in the highly enriched clone TRBV9 were obtained. The transcription factors were sorted out and a gene-related analysis was performed to obtain the phenomenon of enrichment of this clone. The related genes are listed in Table 4. Table 5 shows candidate genes of which overexpression may enhance T cell functions. Table 6 shows candidate genes of which decreased gene expression may enhance T cell functions.










TABLE 4





No.
Gene Name
















1
BATF


2
HMGA1


3
STAT5A


4
ZNF580


5
GLMP


6
JAZF1


7
RUNX1


8
ZGPAT


9
ZNF511


10
GTF2IRD2B


11
ATF4


12
MBD4


13
TBPL1


14
GTF2B


15
RBCK1


16
ZBTB38


17
PIN1


18
DRAP1


19
THYN1


20
HSF1


21
PRDM1


22
ZNF428


23
NFYC


24
ZNF706


25
GTF3A


26
JUN


27
IRF1


28
JUNB


29
TMF1


30
ELF1


31
AKNA


32
BCL11B


33
KLF2


34
ZNF292


35
RORA


36
HMGN3


37
KDM2A


38
ASCL2


39
SP140L


40
NFATC2


41
RUNX3


42
NFE2L2


43
KLF6


44
MTERF4


45
PHF20


46
RELB


47
MAZ


48
ARID5A


49
REL


50
ZEB2


51
ARID5B


52
KLF3


53
CREM


54
ZNF207


55
IRF7


56
DR1


57
SP140


58
BBX


59
MECP2


60
STAT4


61
ZBTB1


62
CREBZF


63
NFATC3


64
GPBP1


65
IKZF1


66
SON


67
ZNF800


68
STAT3


69
STAT6


70
CGGBP1


71
FOXN2


72
DNMT1


73
SP100


74
GATA3


75
EOMES


76
YY1


77
SP110


78
SAFB


79
REST


80
NR3C1


81
FOXN3


82
ELF2


83
GTF2I


84
BAZ2A


85
ZNF683


86
STAT1


87
BHLHE40


88
ZNF276


89
ETS1


90
NFAT5


91
BPTF


92
KMT2A


93
FOS


94
PA2G4


95
IKZF3


96
ZNF148


97
CDC5L


98
CREB1


99
HBP1


100
ZNF721


101
KAT7


102
SP4


103
ZC3H8


104
AKAP8L


105
ZNF326


106
ZNF451


107
ZNF131


108
CEBPZ


109
TOPORS


110
ZNF33A


111
NCOA3


112
STAT2


113
DDIT3


114
ZNF217


115
KLF9


116
CSRNP1


117
NCOA1


118
SAFB2


119
ZNF107


120
ZFX


121
E2F4


122
HIF1A


123
ZNF480


124
CTCF


125
ZBTB44


126
NCOA2


127
ZHX1


128
ZNF644


129
ASH1L


130
STAT5B


131
AEBP2


132
MYSM1


133
ZNF91


134
CEBPB


135
MXD4


136
YBX3


137
RLF


138
JUND


139
ZNF600


140
SMAD4


141
TET2


142
ZNF267


143
PRDM2


144
ZBTB7A


145
THAP12


146
ETV3


147
NFKB2


148
KLF13


149
SATB1


150
ZNF791


151
RBPJ


152
SPEN


153
PURA


154
ZNF507


155
FOSL2


156
IRF8


157
ELK4


158
ATF3


159
KCMF1


160
ZNF639


161
SKI


162
FOXO1


163
NR4A2


164
ZNF331


165
NFKB1


166
CEBPD


167
FOSB


168
SKIL


169
NR4A3


170
NR4A1

















TABLE 5





No.
Gene Name
















1
BATF


2
HMGA1


3
STAT5A


4
ZNF580


5
GLMP


6
JAZF1


7
RUNX1


8
ZGPAT


9
ZNF511


10
GTF2IRD2B


11
ATF4


12
MBD4


13
TBPL1


13
TBPL1


14
GTF2B


15
RBCK1


16
ZBTB38


17
PIN1


18
DRAP1


19
THYN1


20
HSF1


21
PRDM1


22
ZNF428


23
NFYC


24
ZNF706

















TABLE 6





No.
Gene Name
















1
GTF3A


2
JUN


3
IRF1


4
JUNB


5
TMF1


6
ELF1


7
AKNA


8
BCL11B


9
KLF2


10
ZNF292


11
RORA


12
HMGN3


13
KDM2A


14
ASCL2


15
SP140L


16
NFATC2


17
RUNX3


18
NFE2L2


19
KLF6


20
MTERF4


21
PHF20


22
RELB


23
MAZ


24
ARID5A


25
REL


26
ZEB2


27
ARID5B


28
KLF3


29
CREM


30
ZNF207


31
IRF7


32
DR1


33
SP140


34
BBX


35
MECP2


36
STAT4


37
ZBTB1


38
CREBZF


39
NFATC3


40
GPBP1


41
IKZF1


42
SON


43
ZNF800


44
STAT3


45
STAT6


46
CGGBP1


47
FOXN2


48
DNMT1


49
SP100


50
GATA3


51
EOMES


52
YY1


53
SP110


54
SAFB


55
REST


56
NR3C1


57
FOXN3


58
ELF2


59
GTF2I


60
BAZ2A


61
ZNF683


62
STAT1


63
BHLHE40


64
ZNF276


65
ETS1


66
NFAT5


67
BPTF


68
KMT2A


69
FOS


70
PA2G4


71
IKZF3


72
ZNF148


73
CDC5L


74
CREB1


75
HBP1


76
ZNF721


77
KAT7


78
SP4


79
ZC3H8


80
AKAP8L


81
ZNF326


82
ZNF451


83
ZNF131


84
CEBPZ


85
TOPORS


86
ZNF33A


87
NCOA3


88
STAT2


89
DDIT3


90
ZNF217


91
KLF9


92
CSRNP1


93
NCOA1


94
SAFB2


95
ZNF107


96
ZFX


97
E2F4


98
HIF1A


99
ZNF480


100
CTCF


101
ZBTB44


102
NCOA2


103
ZHX1


104
ZNF644


105
ASH1L


106
STAT5B


107
AEBP2


108
MYSM1


109
ZNF91


110
CEBPB


111
MXD4


112
YBX3


113
RLF


114
JUND


115
ZNF600


116
SMAD4


117
TET2


118
ZNF267


119
PRDM2


120
ZBTB7A


121
THAP12


122
ETV3


123
NFKB2


124
KLF13


125
SATB1


126
ZNF791


127
RBPJ


128
SPEN


129
PURA


130
ZNF507


131
FOSL2


132
IRF8


133
ELK4


134
ATF3


135
KCMF1


136
ZNF639


137
SKI


138
FOXO1


139
NR4A2


140
ZNF331


141
NFKB1


142
CEBPD


143
FOSB


144
SKIL


145
NR4A3


146
NR4A1
















TABLE 7







Sequence Listing










SEQ ID

SEQ ID



NO:
Identity
NO:
Identity













1
SP
30
Tumor-associated MUC1 scFv 1


2
Hinge & transmembrane
31
Tumor-associated MUC1 scFv-1 VH



domain




3
Co-stimulatory region
32
Tumor-associated MUC1 scFv-1 VL


4
CD3-zeta
33
Tumor-associated MUC1 scFv 2


5
scFV Humanized CD19
34
Tumor-associated MUC1 scFv2 VH


6
scFV CD19
35
Tumor-associated MUC1 scFv2 VL


7
scFv FZD10
36
Modified PD-1 intracellular domain -1





(two tyrosine kinase mutations)


8
scFv TSHR
37
Modified PD-1 of SEQ ID NO: 36





(extracellular, transmembrane, and





intracellular domains)


9
scFv PRLR
38
Modified PD-1 intracellular domain -2


10
scFv Muc 17
39
Modified PD-1 intracellular domain -3


11
scFv GUCY2C
40
Modified PD-1 intracellular domain -4


12
scFv CD207
41
Modified PD-1 intracellular domain -5


13
Prolactin (ligand)
42
Removed PD-1 intracellular domain -2


14
scFv CD3
43
A hinge


15
scFv CD4
44
Seq1: WT


16
scFv CD4-2
45
Seq2: Y201F


17
scFv CD5
46
Seq3: Y218F


18
WTCD3zeta
47
Seq4:Y201F Y218F


19
WTCD3zeta-
48
Seq5: Truncated (delete internal 190-223)



BCMACAR full length




20
BCMACAR
49
Seq6: Replace with





CD8 transmembrane (delete





161-223, add CD8 transmembrane)


21
MUC1CAR
50
Seq7: L141A Y201F Y218F


22
m19CAR-IRES-MUC1CAR
51
Seq8: Truncated (delete internal 190-223) +





L141A


23
hCD19CAR-IRES-MUC1CAR
52
Seq9: Replace with CD8





transmembrane + L141A


24
hCD22CAR-IRES-MUC1CAR
53
WT CD3 zeta aa


25
BCMACAR-IRES-MUC1CAR
45
Modified PD-1 (WT)


26
mCD19CAR-2A-MUC1CAR
55
Modified PD-1 (Point mutation 1)


27
hCD19CAR-2A-MUC1CAR
56
Modified PD-1 (point mutations 2 sites-2)


28
hCD22CAR-2A-MUC1CAR
57
Modified PD-1 (point mutations 2 sites-3)


29
BCMA-2A-MUC1CAR
58
P2A aa


59
CD28 Co-stimulation
60
HMGY aa



Domain




61
HMGY nucleotide
62
ZBTB38 aa


63
NFAT promoter









Expression of HMGY in CAR T Cells


FIG. 5 shows the expression of HMGY in various cells. On day 0, the peripheral blood of healthy volunteers was drawn, and CD3+ T cells were sorted with pan T Kit. 100 ul of T cell TransAct™ (for activating and expanding human T cells via CD3 and CD28) were added per 1×106 T cells. On day 1, 4×106 6922 cells were transfected with lentivirus vectors, wherein multiplicity of infection (MOI) is 20.79, and 4×106 7413 cells were transfected with lentivirus vectors, wherein MOI is 60.03. 6×106 T cells are non-transfected cells (NT). On day 2, the medium was changed to remove lentivirus and TransAct™, and T cells were resuspended with a fresh medium. On day 7, flow cytometry was used to detect CAR ratio and cell phenotype. Since both vectors were humanized antibodies, human CAR antibodies were used for detection. As shown in FIGS. 5 and 6, human CD19-CD28-CD3zeta (h19-28z) CART cells (T cells including anti-CD19 CAR including CD28 co-stimulation domain and CD3zeta domain) has a total hCAR expression of 27.49%, and h19-28z-2a-HMGY (T cells including anti-CD19 CAR and HMGY) has an expression of 19.89%. After testing, the leveling is 19.89% CAR, and the experiment is carried out according to the following table. The samples were stained with CAR+ multi-color by flow cytometry, and the trace labeled cells were taken for flow cytometry to detect the amplification status at 96 hours (hrs). Sequences described in the Examples and Embodiments are listed in Table 7 above.











TABLE 8





Cell ID
Construction
Notes







6922
CAR-h19-
Humanized anti-CD19 CAR (including



28z
humanized CD19 scFv, CD28 Co-




stimulation Domain, and CD3 zeta




domain)


7413
H19-28z-2a-
Humanized anti-CD19 CAR (including



HMGY
humanized CD19 scFv, CD28 Co-




stimulation Domain and CD3 zeta




domain) + 2A + HMGY









3T3 cell overexpressing 6922 and 7413 were transduced with vectors and cultured for 5 days and then harvested for detection. After extracting RNA from the 3T3 cells, qPCR was performed. Relative quantitation by the SYBR Green method was also performed. FIG. 5 shows the difference in HMGY RNA expression relative to the internal reference β-actin expression, which shows overexpression of 7413 in 3T3 cells. HMGY expression in T cells is high, and HMGY expression in the T cells without vectors transduced is low.









TABLE 9







Experimental Design and Grouping











Substrate cell
E:T
system





T cell
nalm6
3:1
24-well plate 400 u1 x-


NT


vivo without IL2 added


NT
+




6922





6922
+




7413





7413
+










FIGS. 6 and 7 show flow cytometry results of expression of markers CD62L and CCR7 of various cells. NT, 6922, 7413 cells were co-cultured with nalm6 cells for 24 hrs. Various surface marker expression was detected on day 7. FIG. 6 shows flow cytometry results, and FIG. 7 shows statistic data based on the flow cytometry results. 6922 and 7413 cells were co-cultured with or without nalm6 activation for 24 hrs, and the flow-related memory markers CD62L and CCR7 were detected by flow cytometry. Higher expression of CD62L and CCR7 were observed in 7413 cells in the presence or absence of nalm6 activation, keeping T cells in the memory state. After overexpressing the HMGY gene, the background CD62L and CCR7 expression levels were up-regulated, and the percentage of down-regulation decreased significantly after activation.



FIGS. 8 and 9 show flow cytometry results of expression of marker KLRG and CD137 of various cells. 6922 and 7413 cells were co-cultured with nalm6 cells for 24 hrs, and several markers were detected on day 7. FIG. 8 shows flow cytometry results, and FIG. 9 shows statistic data based on the flow cytometry results. 6922 and 7413 cells were co-cultured with or without nalm6 activation for 24 hrs, and flow cytometry was used to detect differentiation-related marker KLRG and activated marker CD137. The results are shown in two CD4 and CD8 T cell subsets. The 7413 cells overexpressing HMGY have lower CD137 expression and lower KLRG expression when stimulated by nalm6, leaving the cells in a weakly activated and lower differentiated state.



FIGS. 10 and 11 shows flow cytometry results of cell expansion of various cells. 6922 and 7413 cells were co-cultured with nalm6 cells for 96 hrs, and cell expansion assay was performed. FIG. 10 shows flow cytometry results, and FIG. 11 shows statistic data based on the flow cytometry results. 6922 and 7413 cells were co-cultured with or without nalm6 activation for 96 hrs, and CellTrace™ was used to label the T cells to show proliferation/expansion by flow cytometry. The results show that in CD4 and CD8 T cell subtypes of the co-cultured cells, the 7413 cells overexpressing HMGY have higher expansion rates and absolute numbers of expanded cells when stimulated by nalm6. Because it is the P2A linked CAR and HMGY genes, 7413 cells also showed greater expansion than 6922 cells in the absence of nalm6 activation.


These results demonstrate that the H19-28z-2a-HMGY vector can effectively express CAR and HMGY at the same time. After overexpressing the HMGY gene, the CAR T cells have higher CD62L and CCR7 expression, which indicates that they have the memory-like phenotype and are activated. The results show that overexpression of the HMGY gene in CAR T cells can also effectively reduce the expression of CD62L and CCR7 and thus promote CAR T cells to be in a better state for killing tumor in a subject. Moreover, after overexpressing the HMGY gene, the stimulated CAR T cells express lower levels of CD137 and KLRG, leaving the CAR T cells in a weakly activated and poorly differentiated state, indicating that they exhibit less of the memory-like phenotype in response to antigens. Further, after overexpressing the HMGY gene, the cells can expand more after being activated.


All publications, patents, and patent applications cited in this specification are incorporated herein by reference in their entireties as if each individual publication, patent, or patent application were specifically and individually indicated to be incorporated by reference. While the foregoing has been described in terms of various embodiments, the skilled artisan will appreciate that various modifications, substitutions, omissions, and changes may be made without departing from the spirit thereof.

Claims
  • 1. A method of producing T cells exhibiting an enhanced memory T cell phenotype, the method comprising: introducing a polynucleotide encoding high-mobility group protein Y (HMGY) into a population of T cells, wherein expression of HMGY is higher in the population of T cells as compared to a population of T cells that are not introduced with the polynucleotide, and the memory T cell phenotype of the population of T cells is enhanced as compared to the population of T cells that are not introduced with the polynucleotide.
  • 2. The method of claim 1, wherein the population of T cells exhibits an increased gene expression level in CD62L and/or CCR7 as compared to a population of T cells that are not introduced with the polynucleotide.
  • 3. The method of claim 1, the method further comprising: obtaining peripheral blood mononuclear cells (PBMCs) from a subject or a healthy donor;isolating the population of T cells from the PBMCs;culturing the population of T cells; andmeasuring expansion of the population of T cells.
  • 4. The method of claim 3, wherein expansion of the population of T cells is enhanced as compared to a population of T cells that are not introduced with the polynucleotide.
  • 5. The method of claim 1, the method further comprising: obtaining blood from a subject or a healthy donor, the blood comprising a population of T cells; andintroducing the polynucleotide encoding HMGY into the blood.
  • 6. The method of claim 1, wherein the polynucleotide comprises SEQ ID NO: 61 or SEQ ID NOS: 61and 63.
  • 7. The method of claim 1, the method further comprising contacting the population of T cells with an antigen that the population of T cells bind.
  • 8. The method of claim 7, wherein the population of T cells exhibits a reduced gene expression level of CD137 and/or KLRG as compared to a population of T cells that are not introduced with the polynucleotide.
  • 9. The method of claim 1, wherein the population of T cells comprising enhanced memory T cell phenotype comprises an increased gene expression level of CD62L and/or CCR7 as compared to a population of T cells that are not introduced with the polynucleotide.
  • 10. The method of claim 1, wherein the population of T cells comprising enhanced memory T cell phenotype comprises a reduced gene expression level of CD137 and/or KLRG as compared to a population of T cells that are not introduced with the polynucleotide.
  • 11. The method of claim 1, wherein the population of T cells comprise an antigen binding molecule.
  • 12. The method of claim 11, wherein the antigen binding molecule is a chimeric antigen receptor (CAR), which comprises an antigen-binding domain, a transmembrane domain, and an intracellular signaling domain.
  • 13. The method of claim 12, wherein the antigen binding domain binds a tumor antigen selected from a group consisting of TSHR, CD19, CD123, CD22, CD30, CD171, CS-1, CLL-1, CD33, EGFRvIII, GD2, GD3, BCMA, Tn Ag, PSMA, ROR1, FLT3, FAP, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3, KIT, IL-13Ra2, Mesothelin, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-beta, SSEA-4, CD20, Folate receptor alpha, ERBB2 (Her2/neu), MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, IGF-I receptor, CAIX, LMP2, gp100, bcr-abl, tyrosinase, EphA2, Fucosyl GM1, sLe, GM3, TGS5, HMWMAA, o-acetyl-GD2, Folate receptor beta, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, Polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, NY-ESO-1, LACE-1a, MAGE-A1, legumain, HPV E6, E7, MAGE A1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53, p53 mutant, prostein, survivin, telomerase, PCTA-1/Galectin 8, MelanA/MART1, Ras mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, Androgen receptor, Cyclin 61, MYCN, RhoC, TRP-2, CYP161, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, RAGE-1, human telomerase reverse transcriptase, RU1, RU2, intestinal carboxyl esterase, mut hsp70-2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.
  • 14. The method of claim 12, wherein the intracellular signaling domain comprises a co-stimulatory signaling domain, or a primary signaling domain and a co-stimulatory signaling domain, wherein the co-stimulatory signaling domain comprises a functional signaling domain of a protein selected from the group consisting of CD27, CD28, 4-166 (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D.
  • 15. The method of claim 11, wherein the antigen binding molecule is a modified TCR.
  • 16. The method of claim 15, wherein the TCR is derived from spontaneously occurring tumor-specific T cells in patients.
  • 17. The method of claim 16, wherein the TCR binds a tumor antigen.
  • 18. The method of claim 17, wherein the tumor antigen comprises CEA, gp100, MART-1, p53, MAGE-A3, or NY-ESO-1.
  • 19. The method of claim 18, wherein the TCR comprises TCRγ and TCRδ chains, TCRα and TCRβ chains, or a combination thereof.
  • 20. The method of claim 1, wherein the cell is a human cell.
CROSS-REFERENCE TO RELATED PATENT APPLICATIONS

This application claims the benefit of U.S. Provisional Application 62/879,186, filed Jul. 26, 2019, which is hereby incorporated by reference in its entirety.

Provisional Applications (1)
Number Date Country
62879186 Jul 2019 US