CD22 CHIMERIC ANTIGEN RECEPTOR (CAR) THERAPIES

Information

  • Patent Application
  • 20220047633
  • Publication Number
    20220047633
  • Date Filed
    September 27, 2019
    4 years ago
  • Date Published
    February 17, 2022
    2 years ago
Abstract
The invention provides compositions and methods for treating cancer, e.g., hematological cancer, by administering a CD22 CAR-expressing cell described herein according to a dosage regimen described herein. Also disclosed are methods of making and compositions comprising the same.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Sep. 20, 2019, is named N2067-7162WO_SL.txt and is 447,205 bytes in size.


FIELD OF THE INVENTION

The present invention relates generally to the use of T cells engineered to express a Chimeric Antigen Receptor (CAR) expressing CD22, to treat a disease, e.g., a hematological cancer.


BACKGROUND OF THE INVENTION

Many patients with B cell malignancies are incurable with standard therapy. In addition, traditional treatment options often have serious side effects. Attempts have been made in cancer immunotherapy, however, several obstacles render this a very difficult goal to achieve clinical effectiveness. Although hundreds of so-called tumor antigens have been identified, these are generally derived from self and thus are poorly immunogenic. Furthermore, tumors use several mechanisms to render themselves hostile to the initiation and propagation of immune attack.


Recent developments using chimeric antigen receptor (CAR) modified autologous T cell (CART) therapy, which relies on redirecting T cells to a suitable cell-surface molecule on cancer cells such as B cell malignancies, show promising results in harnessing the power of the immune system to treat B cell malignancies and other cancers (see, e.g., Sadelain et al., Cancer Discovery 3:388-398 (2013)). The clinical results of the murine derived CART19 (i.e., “CTL019”) have shown promise in establishing complete remissions in patients suffering with CLL as well as in childhood ALL (see, e.g., Kalos et al., Sci Transl Med 3:95ra73 (2011), Porter et al., NEJM 365:725-733 (2011), Grupp et al., NEJM 368:1509-1518 (2013)). Besides the ability for the chimeric antigen receptor on the genetically modified T cells to recognize and destroy the targeted cells, a successful therapeutic T cell therapy needs to have the ability to proliferate and persist over time, in order to survey for leukemic relapse. The variable quality of T cells, resulting from anergy, suppression, or exhaustion, will have effects on CAR-transformed T cells' performance, over which skilled practitioners have limited control at this time. To be effective, CAR transformed patient T cells need to persist and maintain the ability to proliferate in response to the cognate antigen. It has been shown that ALL patient T cells perform can do this with CART19 comprising a murine scFv (see, e.g., Grupp et al., NEJM 368:1509-1518 (2013)).


SUMMARY OF THE INVENTION

The disclosure provides, inter alia, a method of treating a hematological cancer, comprising administering cells that express a CAR molecule that binds CD22, e.g., a CD22 CAR as described herein. In some embodiments, the CD22 CAR is administered according to a fractionated dosing regimen, e.g., a split-dosing regimen, e.g., as described herein. In some embodiments, the hematological cancer is B-cell ALL, e.g., relapsed and/or refractory ALL. In some embodiments, the subject is a young adult or a pediatric subject. Also described herein are compositions comprising CD22 CAR-expressing cells, methods of manufacturing the same and combination therapies comprising CD22 CAR-expressing cells, e.g., a combination therapy comprising CD22 CAR-expressing cells and CD19 CAR-expressing cells.


In an aspect, provided herein is a method of treating a young adult or a pediatric subject having a hematological cancer comprising administering to the subject an effective number of cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR. In some embodiments, the CD22 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., a split-dosing regimen.


In another aspect, provided herein is a composition comprising cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR, for use in the treatment of a young adult or a pediatric subject having a hematological cancer comprising administering to the subject an effective number of said cells. In some embodiments, the CD22 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., a split-dosing regimen.


In some embodiments, the CD22 CAR-expressing cells are administered at a dose of about 0.02×107 to 5.0×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg.


In some embodiments, the CD22 CAR-expressing cells are administered at a dose of about 0.5×108 to 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.


In another aspect, provided herein is a method of treating a subject having a hematological cancer, comprising administering to the subject an effective number of cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR.


In a related aspect, provided herein is a composition comprising cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR, for use in the treatment of a subject having a hematological cancer comprising administering to the subject an effective number of said cells.


In some embodiments, the CD22 CAR-expressing cells are administered at a dose of about 0.02×107 to 5.0×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg.


In some embodiments, the CD22 CAR-expressing cells are administered at a dose of about 0.5×108 to 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.


In some embodiments, the CD22 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., a split-dosing regimen, e.g., as described herein.


In some embodiments of any of the methods or compositions for use provided herein, the hematological cancer is associated with expression of CD22 or CD19.


In some embodiments of any of the methods or compositions for use provided herein, the hematological cancer is a leukemia or a lymphoma. In some embodiments, the hematological cancer is a relapsed and/or refractory cancer. In some embodiments, the hematological cancer is chosen from B-cell acute Lymphoid Leukemia (B-ALL), T-cell acute Lymphoid Leukemia (T-ALL), acute lymphoid leukemia (ALL), Prolymphocytic leukemia, Chronic myeloid leukemia (CML), blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma, Follicular lymphoma, Hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), Marginal zone lymphoma, multiple myeloma, acute myeloid leukemia (AML), myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, or plasmablastic lymphoma. In some embodiments, the hematological cancer is a relapsed and/or refractory B-ALL.


In some embodiments of any of the methods or compositions for use provided herein, the subject having a hematological cancer, e.g., as described herein, is a young adult or a pediatric subject. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years.


In some embodiments of any of the methods or compositions for use provided herein, the hematological cancer is B-cell ALL. In some embodiments of any of the methods or compositions for use provided herein, the hematological cancer is relapsed and/or refractory B cell ALL. In some embodiments, the subject having a relapsed and/or refractory B-cell ALL is a young adult or a pediatric subject. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years.


In some embodiments of any of the methods or compositions for use provided herein, the dose fractionation dosing regimen comprises a total dose administered in, e.g., one, two, three, or more separate administrations of a partial dose. In some embodiments, the dose fractionation dosing regimen comprising a total dose comprises three administrations of a partial dose, e.g., a first partial dose, a second partial dose and a third partial dose. In some embodiments, the first partial dose comprises a first percentage of the total dose and is administered on a first day of treatment. In some embodiments, the first percentage comprising the first partial dose is about 10% (e.g., 10%) of the total dose. In some embodiments, the second partial dose comprises a second percentage of the total dose and is administered on a second day of treatment. In some embodiments, the second percentage comprising the second partial dose is about 30% (e.g., 30%) of the total dose. In some embodiments, the third partial dose comprises a third percentage (e.g., the remaining percentage) of the total dose and is administered on a third day of treatment. In some embodiments, the third percentage comprising the third partial dose is about 60% (e.g., 60%) of the total dose. In some embodiments, the first partial dose, the second partial dose and the third partial dose are administered on consecutive days.


In some embodiments of any of the methods or compositions for use provided herein, the total cell dose of a CAR-expressing cell, e.g., a CD22 CAR-expressing cell, comprises about 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CD22 CAR-expressing cells/kg or about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CD22 CAR-expressing cells. In some embodiments, the total cell dose comprises about 0.2×107 to 1.0×107 CD22 CAR-expressing cells/kg. In some embodiments, the total cell dose comprises about 1×108 to 5×108 CD22 CAR-expressing cells.


In some embodiments, the total cell dose comprises about 0.2×107 to 1.0×107 CD22 CAR-expressing cells/kg. In some embodiments, the first partial dose, e.g., 10% of the total dose, comprises about 0.2×106 to 1×106 CD22 CAR-expressing cells/kg. In some embodiments, the second partial dose, e.g., 30% of the total dose, comprises about 0.6×106 to 3×106 CD22 CAR-expressing cells/kg. In some embodiments, the third partial dose, e.g., 60% of the total dose, comprises about 1.2×106 to 6×106 CD22 CAR-expressing cells/kg. In some embodiments, the subject weighs less than 50 kg.


In some embodiments, the total cell dose comprises about 1×108 to 5×108 CD22 CAR-expressing cells. In some embodiments, the first partial dose, e.g., 10% of the total dose, comprises about 1×107 to 5×107 CD22 CAR-expressing cells. In some embodiments, the second partial dose, e.g., 30% of the total dose, comprises about 0.3×107 to 1.5×108 CD22 CAR-expressing cells. In some embodiments, the third partial dose, e.g., 60% of the total dose, comprises about 0.6×108 to 3×108 CD22 CAR-expressing cells. In some embodiments, the subject weighs 50 kg or more than 50 kg.


In some embodiments of any of the methods or compositions for use provided herein, a CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., a CD22 CAR expressing cell, is administered at a dose of about 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg, when the subject weighs <50 kg. In some embodiments, the CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., CD22 CAR expressing cell, is administered at a dose of about 0.2×107 to 1.0×107 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, the CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., CD22 CAR expressing cell, is administered at a dose of about 0.02×107 to 1×107, about 1×107 to 1.5×107, about 1.5×107 to 2×107, about 2×107 to 2.5×107, about 2.5×107 to 3×107, about 3×107 to 3.5×107, about 3.5×107 to 4×107, about 4×107 to 4.5×107, or about 4.5×107 to 5×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg. In some embodiments, the CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., CD22 CAR expressing cell, is administered at a dose of about 0.02×107, about 0.04×107, about 0.06×107, about 0.08×107, about 1×107, about 1.5×107, about 2×107, about 2.5×107, about 3×107, about 3.5×107, about 4×107, about 4.5×107, or about 5×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg.


In some embodiments of any of the methods or compositions for use provided herein, a CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., a CD22 CAR expressing cell, is administered at a dose of about 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg, when the subject weighs <50 kg, according to a dose fractionation regimen described herein, e.g., a split dosing regimen. In some embodiments, the dose fractionation dosing regimen comprising a total dose comprises three administrations of a partial dose, e.g., a first partial dose, a second partial dose and a third partial dose. In some embodiments, the first partial dose, e.g., 10% of the total dose, comprises about 0.2×106 to 1×106 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, the second partial dose, e.g., 30% of the total dose, comprises about 0.6×106 to 3×106 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, the third partial dose, e.g., 60% of the total dose, comprises about 1.2×106 to 6×106 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg.


In some embodiments of any of the methods or compositions for use provided herein, a CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., a CD22 CAR expressing cell, is administered at a dose of about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CAR-expressing cells, e.g., CD22 CAR-expressing cells, when the subject weighs ≥50 kg. In some embodiments, the CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., CD22 CAR expressing cell, is administered at a dose of about 1×108 to 5×108 CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, the CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., CD22 CAR expressing cell, is administered at a dose of about 0.5×108 to 1×108, about 1×108 to 1.5×108, about 1.5×108 to 2×108, about 2×108 to 2.5×108, about 2.5×108 to 3×108, about 3×108 to 3.5×108, about 3.5×108 to 4×108, about 4×108 to 4.5×108, about 4.5×108 to 5×108, about 5×108 to 6×108, about 6×108 to 7×108, about 7×108 to 8×108, about 8×108 to 9×108, or about 9×108 to 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg. In some embodiments, the CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., CD22 CAR expressing cell, is administered at a dose of about 0.5×108, about 1×108, about 1.5×108, about 2×108, about 2.5×108, about 3×108, about 3.5×108, about 4×108, about 4.5×108, about 5×108, about 6×108, about 7×108, about 8×108, about 9×108, about 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.


In some embodiments of any of the methods or compositions for use provided herein, a CAR expressing cell, e.g., CAR-expressing cell described herein, e.g., a CD22 CAR expressing cell, is administered at a dose of about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CAR-expressing cells, e.g., CD22 CAR-expressing cells, when the subject weighs ≥50 kg, according to a dose fractionation regimen described herein, e.g., a split dosing regimen. In some embodiments, the dose fractionation dosing regimen comprising a total dose comprises three administrations of a partial dose, e.g., a first partial dose, a second partial dose and a third partial dose. In some embodiments, the first partial dose, e.g., 10% of the total dose, comprises about 1×107 to 5×107 CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, the second partial dose, e.g., 30% of the total dose, comprises about 0.3×107 to 1.5×108 CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, the third partial dose, e.g., 60% of the total dose, comprises about 0.6×108 to 3×108 CAR-expressing cells, e.g., CD22 CAR-expressing cells.


In some embodiments of any of the methods or compositions for use provided herein, comprising administering an additional dose, e.g., supplemental dose, of CAR-expressing cells, e.g, CD22 CAR-expressing cells. In some embodiments, the additional dose comprises, about 0.6×106 to 3×106 CAR-expressing cells/kg or about 0.3×108 to 1.5×108 CAR-expressing cells. In some embodiments, the additional dose comprises, less than about 1.2×106 to 6×106 CAR-expressing cells/kg or about 0.6×108 to 3×108 CAR-expressing cells. In some embodiments, the additional dose administered at least 13 days after administration of the first partial dose of CAR-expressing cell, e.g., CD22 CAR-expressing cells, e.g., administered on day 14 or later.


In some embodiments, any of the methods or compositions for use provided herein, further comprise administering to the subject an effective number of cells that express a CAR molecule that binds CD19, e.g., a CD19 CAR. In some embodiments, the CD22 CAR-expressing cells are administered before, after or concurrently with the administration of the CD19 CAR-expressing cells.


In some embodiments of any of the methods or compositions for use provided herein, the CD19 CAR-expressing cells are administered as a single dose infusion, e.g., a total dose is administered in a single infusion, e.g., at a dose of about 0.2×106 to 10×106 CD19 CAR-expressing cells/kg, e.g., about 2.0×106 CD19 CAR-expressing cells/kg.


In some embodiments of any of the methods or compositions for use provided herein, the CD19 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., split-dosing regimen described herein, wherein the dose fractionation dosing regimen comprises a total dose administered in, e.g., one, two, three, or more separate administrations of a partial dose. In some embodiments, the dose fractionation dosing regimen comprising a total dose comprises three administrations of a partial dose, e.g., a first partial dose, a second partial dose and a third partial dose. In some embodiments, the first partial dose comprises a first percentage (e.g., about 10%) of the total dose; and is administered on a first day of treatment. In some embodiments, the second partial dose comprises a second percentage (e.g., about 30%) of the total dose; and is administered on a second day of treatment. In some embodiments, the third partial dose comprises a third percentage (e.g., about 60%) of the total dose; and is administered on a third day of treatment. In some embodiments, the first partial dose, the second partial dose and the third partial dose are administered on consecutive days. In some embodiments, the total cell dose comprises about 0.2×106 to 10×106 CD19 CAR-expressing cells/kg, e.g., about 2.0×106 CD19 CAR-expressing cells/kg. In some embodiments, the first partial dose comprises about 0.2×106 CD19 CAR-expressing cells/kg, the second partial dose comprises about 0.6×106 CD19 CAR-expressing cells/kg, and the third partial dose comprises about 1.2×106 CD19 CAR-expressing cells/kg.


In some embodiments of any of the methods or compositions for use provided herein, the subject has been administered lymphodepleting chemotherapy.


In some embodiments of any of the methods or compositions for use provided herein, the subject has not been administered lymphodepleting chemotherapy.


In some embodiments of any of the methods or compositions for use provided herein, has not relapsed to treatment with cells that express a CAR molecule, e.g., a CD19 CAR or a CD22 CAR therapy (e.g., a CD19 CAR monotherapy or a CD22 CAR monotherapy).


In some embodiments of any of the methods or compositions for use provided herein, administration of the combination comprising a CD19 CAR and CD22 CAR prevents relapse in the subject, relative to a CD19 CAR monotherapy or a CD22 CAR monotherapy, or relative to a subject that has not received the combination therapy.


In some embodiments of any of the methods or compositions for use provided herein, the CAR expressing cell, e.g., CD22 CAR22-expressing cell, is administered to a subject who has previously received stem cell transplantation (SCT) therapy, e.g., allogeneic or autologous SCT. In some embodiments, the CAR expressing cell, e.g., CD22 CAR22-expressing cell, is administered to a subject who is not eligible for SCT therapy, e.g., e.g., allogeneic or autologous SCT. In some embodiments, the subject has previously received SCT therapy and the subject has, or is identified as having relapsed from the SCT therapy. In some embodiments, the relapse from SCT therapy comprises a blood or bone marrow relapse and/or the relapse occurs at least 1-12 months, e.g., at least 6 months, after SCT therapy.


In some embodiments of any of the methods or compositions for use provided herein, the CAR expressing cell, e.g., CD22 CAR22-expressing cell, is administered to a subject who has, or is identified as having, a relapse, e.g., a second or greater relapse, e.g., bone marrow relapse, e.g., as described herein.


In some embodiments of any of the methods or compositions for use provided herein, the CAR expressing cell, e.g., CD22 CAR22-expressing cell, is administered to a subject who has, or is identified as having relapsed from a CAR-expressing cell therapy, e.g., a CAR-expressing cell therapy other than a CAR22-expressing cell therapy or a CAR19-expressing cell therapy.


In some embodiments of any of the methods or compositions for use provided herein, the CAR expressing cell, e.g., CD22 CAR22-expressing cell, is administered to a subject who has, or is identified as not having a response, e.g., a complete response or partial response, in response to one or more (e.g., 2, 3, 4, 5, 6, 7, or 8) chemotherapeutic agents, e.g., as described herein.


Timing and Dosage of the Combination Administration

The one or more therapies described herein can be administered to the subject substantially at the same time or in any order. For instance, a CD22 CAR, e.g., a CD22 CAR-expressing cell described herein, and a CD19 CAR, e.g., a CD19 CAR-expressing cell described herein can be administered simultaneously, in the same or in separate compositions, or sequentially. In another example, a CD22 CAR, e.g., a CD22 CAR-expressing cell described herein, and a CD19 CAR, e.g., a CD19 CAR-expressing cell described herein and/or optionally at least one additional therapeutic agent can be administered simultaneously, in the same or in separate compositions, or sequentially.


For sequential administration, the CD22 CAR-expressing cell can be administered first, and the CD19 CAR-expressing cell can be administered second, or the order of administration can be reversed. In some embodiments, the first therapy (e.g., a CD22 CAR-expressing cell such as a CD22 CART cell) is continued when the second therapy (e.g., a CD19 CAR-expressing cell such as a CD19 CART cell) is introduced, and in other embodiments the first therapy is withdrawn before, after, or at the same time as the second therapy is introduced. In instances of sequential administration, in some embodiments, the second therapy is initiated after a predetermined amount of time, or after the subject displays one or more indications that relapse has occurred or is likely to occur. The indication can be, e.g., the presence of cancer cells having a disturbance in the target of the first therapy, e.g., CD19, CD20, or CD22. The disturbance may be, e.g., a frameshift mutation and/or a premature stop codon.


In other embodiments, the two or more therapies (e.g., a CD22 CAR-expressing cell and CD19 CAR-expressing cell) are administered simultaneously. Without being bound by theory, in some embodiments, simultaneous administration of the therapies can reduce the likelihood of relapse and/or delay relapse.


When administered in combination, the first therapy (e.g., CD22 CAR-expressing cell) and the second therapy (e.g., CD19 CAR-expressing cells), or both, can be administered in an amount or dose that is higher, lower, or the same as the amount or dosage of each therapy, e.g., agent, used individually, e.g., as a monotherapy. In certain embodiments, the administered amount or dosage of the first therapy, second therapy, optionally a third therapy, or all, is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy. In other embodiments, the amount or dosage of the first therapy, second therapy, optionally a third therapy, or all, that results in a desired effect (e.g., treatment of cancer) is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower) than the amount or dosage of each agent used individually, e.g., as a monotherapy, required to achieve the same therapeutic effect. In certain embodiments, the lower dose results in reduced side effects compared to those seen when the regular (monotherapy) dose is administered.


In an embodiment, the therapy comprises a population of cells. In embodiments, the cells are immune effector cells, e.g., CAR-expressing cells, e.g., CD22 CAR-expressing cells and/or CD19 CAR-expressing cells.


Alternatively, or in combination with the methods described herein, methods are disclosed that comprise a diagnostic step or a patient selection step, for instance as described below.


In one aspect, the invention provides a method of evaluating a subject, e.g., a patient, for relapser status (e.g. a relapser or a non-relapser after a CAR-therapy). In one embodiment, the method identifies a subject, e.g., a patient, who has relapsed (“relapser”) or who is are likely to relapse, or who has not relapsed (“non-relapser”) or who is likely not to relapse, after treatment with a CAR therapy (e.g., a CD19 CART therapy, e.g., described herein, e.g., a CTL019 therapy). In an embodiment, relapser status (e.g. relapser or non-relapser after a CART therapy) is determined by assaying for one or more characteristics of CD19. In some embodiments, the subject is a young adult or a pediatric subject. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years.


In an embodiment, methods are provided for identifying a subject having cancer, e.g., a hematological cancer, e.g., ALL (e.g., relapsed and/or refractory ALL), as being a relapser or non-relapser after a treatment that comprises a CAR therapy, e.g., a CD19 CART therapy. The method comprises: (1) acquiring a sample from the subject (e.g., an apheresis sample obtained from the blood of the subject; and/or e.g., a manufactured product sample, e.g., genetically engineered T cells obtained from the blood of the subject); (2) determining a characteristic of CD19, e.g., a sequence or level as described herein; and (3) (optionally) comparing the determined characteristic of CD19 to a reference characteristic; wherein the difference, e.g., statistically significant difference, between the determined characteristic compared to the reference characteristic is predictive of relapse to the CAR therapy; and (4) identifying the subject as a relapser or non-relapser to the CAR therapy, e.g., based on the determined characteristic of CD19. In one embodiment, the presence or absence of the characteristic of CD19 is the presence or absence of a premature stop codon, e.g., by an insertion or deletion leading to a frameshift.


In an embodiment, the provided methods comprise (1) acquiring a sample from the subject (e.g., an apheresis sample obtained from the blood of the subject; and/or, e.g., a manufactured product sample, e.g., genetically engineered T cells obtained from the blood of the subject, e.g., a manufactured CART19 product); (2) determining a characteristic of CD19, e.g., a sequence or level as described herein; and (3) (optionally) comparing the determined characteristic of CD19 to a reference characteristic; wherein the presence of the characteristic of CD19 (e.g., the difference, e.g., a statistically significant difference, between the determined characteristic compared to the reference characteristic) is predictive of relapse to the CAR therapy. In one embodiment, the presence of the characteristic of CD19 is the presence of a premature stop codon, e.g., by an insertion or deletion leading to a frameshift.


In an embodiment, methods are provided for determining the relapse of a subject having cancer, e.g., a hematological cancer, e.g., ALL (e.g., relapsed and/or refractory ALL), after a treatment comprising a CAR therapy, e.g., a CD19 CAR therapy as described herein. The method comprises determining a characteristic of CD19 in a sample obtained prior to relapse. In an embodiment, the presence of the characteristic of CD19 (e.g., the difference, e.g., a statistically significant difference, between the determined characteristic compared to the reference characteristic) is indicative of relapse after CAR therapy. In one embodiment, the presence of the characteristic of CD19 is the presence of a premature stop codon, e.g., by an insertion or deletion leading to a frameshift. In some embodiments, the subject is a young adult or a pediatric subject. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years.


In an embodiment, methods are provided for evaluating a subject having cancer, e.g., a hematological cancer, e.g., ALL (e.g., relapsed and/or refractory ALL). The method comprises acquiring a value of relapser status for the subject that comprises a measure of one or characteristics of CD19, e.g., one or more of the characteristics of CD19 as described herein, thereby evaluating the subject. In some embodiments, the subject is a young adult or a pediatric subject. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years.


In an embodiment, methods are provided for evaluating or monitoring the effectiveness of a CAR therapy, e.g., a CD19 CART therapy, in a subject having cancer comprising acquiring a value of relapser status for the subject that comprises a measure of one or more characteristic of CD19, e.g., one or more of the characteristics of CD19 as described herein, thereby evaluating or monitoring the effectiveness of the CAR therapy in the subject. In some embodiments, the subject is a young adult or a pediatric subject. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years.


In an embodiment, methods are provided for providing a prediction for success rate of a CAR therapy, e.g., a CD19 CART therapy, e.g., described herein, in a subject having cancer, said method comprising steps of providing a biological sample from the subject; determining one or more characteristic of CD19, e.g., one or more of the characteristics of CD19 as described herein; and based on the characteristic determined, providing a prognosis to the subject.


In some embodiments of any of the aforesaid methods, the sample is a biological sample selected from a blood, plasma, or a serum sample. In an embodiment, a biological sample is a blood sample. In one embodiment, the sample is an apheresis sample, e.g., T cells obtained from the blood of the subject. In an embodiment, the sample is a manufactured product sample, e.g. genetically engineered T cells obtained from the blood of the subject, e.g., a manufactured CAR product, e.g., a manufactured CART19 product.


In an embodiment, the methods herein can be used to determine if a subject is likely to respond to CAR therapy (e.g., CD19 CART), e.g., if a subject who has not received CAR therapy is likely to respond to CAR therapy, or if a subject who has received CAR therapy is likely to respond to continued CAR therapy. In general, the same CD19 characteristics that predict relapse predict that a subject is less likely to respond to a CD19 CAR therapy. In some embodiments, a subject who is identified as less likely to respond to a CD19 CAR therapy can be administered a CD22 CAR-expressing cell therapy in combination with a CD19 CAR expressing cell therapy.


In another aspect, a method for treating a subject having cancer, e.g., a hematological cancer, e.g., ALL, e.g., relapsed and/or refractory ALL, is provided. In an embodiment, the method includes determining if a subject has a difference, e.g., statistically significant difference, in a characteristic of CD19 relative to a reference characteristic, and if there is a difference, e.g., statistically significant difference between the determined characteristic and reference characteristic, administering to the subject a therapeutically effective dose of a CAR therapy, e.g., CART, thereby treating the subject. In an embodiment, the characteristic is CD19 sequence, e.g., protein or nucleic acid sequence. In an embodiment, the method comprises assaying for the presence or absence of frameshifted CD19, e.g., CD19 comprising a premature stop codon. In some embodiments, the subject is a young adult or a pediatric subject. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years.


In embodiments of any of the aforesaid methods, the treatment comprises administering a CD19 CAR-expressing cell, in combination with a CD22 CAR-expressing cell. In an embodiment, the CD19 CAR therapy is administered simultaneously with CD22 CAR-expressing cell. In an embodiment, the CD19 CAR therapy is administered before the CD22 CAR-expressing cell. In an embodiment, the CD19 CAR therapy is administered after the CD22 CAR-expressing cell.


In an embodiment, the methods of treatment comprise acquiring a value of relapser status for the subject that comprises a measure of a CD19 characteristic, and responsive to a determination of relapser status, performing one, two, three four or more of: (1) identifying the subject as a relapse or non-relapser; (2) administering a CAR therapy; (3) selecting or altering a dosing of a CAR therapy; (4) selecting or altering the schedule or time course of a CAR therapy; (5) administering, e.g., to a relapser, an additional agent in combination with the CAR therapy, e.g., administering one or more B-cell inhibitors; or a checkpoint inhibitor, e.g., a checkpoint inhibitor described herein, or a kinase inhibitor, e.g., a kinase inhibitor described herein; (6) administering to a relapser a therapy that increases the number of naïve T cells in the subject prior to treatment with a CAR therapy; modifying a manufacturing process of a CAR therapy, e.g., enrich for naïve T cells prior to introducing a nucleic acid encoding a CAR, e.g., for a subject identified as a relapser; or (7) selecting an alternative therapy, e.g., a standard of care for a particular cancer (e.g., as described herein), e.g., for a relapser; thereby treating cancer in the subject.


In some embodiments, the method comprises administering one, two, three or more B-cell inhibitors (e.g., one or more inhibitors of CD10, CD20, CD34, CD123, FLT-3, or ROR1 as described herein).


In some embodiments, the methods of treatment described herein further comprise one or both of: determining a level of an immune checkpoint molecule (e.g., PD-L1, PD1, LAG3, or TIM3) in a patient sample; and administering an immune checkpoint inhibitor (e.g., an inhibitor of one or more of PD-L1, PD1, LAG3, and TIM3) to the patient. For example, the method can comprise treating a patient with one or more CAR-expressing cells described herein (e.g., CD19 CAR in combination with a CD22 CAR) and determining the level of an immune checkpoint molecule in the patient before or after the treatment. In some embodiments, the method comprises administering the immune checkpoint inhibitor to a patient that has elevated levels of the immune checkpoint molecule compared to a reference level, e.g., administering a PD-L1 inhibitor in response to elevated PD-L1 levels, administering a PD1 inhibitor in response to elevated PD1 levels, administering a LAG3 inhibitor in response to elevated LAG3 levels, or administering a TIM3 inhibitor in response to elevated TIM3 levels. In some embodiments, the method comprises administering an immune checkpoint inhibitor to a patient who has received, is receiving, or is about to receive therapy with one or more CAR-expressing cells described herein (e.g., CD19 CAR in combination with a CD22 CAR), wherein the patient has, or is identified as having, elevated levels of the immune checkpoint molecule compared to a reference level.


Compositions

In some aspects, the present disclosure provides, e.g., a composition comprising one or more cells that express a CAR molecule that binds CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR.


In some aspects, the present disclosure provides, e.g., a composition comprising: (i) one or more cells that express a CAR molecule that binds CD19, e.g., a CAR molecule that binds CD19 described herein, e.g., a CD19 CAR, and (ii) one or more cells that express a CAR molecule that binds CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR. In embodiments, (i) and (ii) are provided separately, and in embodiments, (i) and (ii) are admixed.


In some aspects, the present disclosure provides, e.g., a nucleic acid encoding a CAR molecule that binds CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR.


In some aspects, the present disclosure provides, e.g., a nucleic acid encoding: (i) a CAR molecule that binds CD19, e.g., a CAR molecule that binds CD19 described herein, e.g., a CD19 CAR, and (ii) a CAR molecule that binds CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR. In embodiments, the nucleic acid comprises RNA or DNA.


In some aspects, the present disclosure provides, e.g., a nucleic acid encoding a CAR molecule that binds CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR.


In some aspects, the present disclosure provides, e.g., a nucleic acid encoding: (i) a CAR molecule that binds CD19, e.g., a CAR molecule that binds CD19 described herein, e.g., a CD19 CAR, and (ii) a CAR molecule that binds a CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR. In embodiments, the nucleic acid comprises RNA or DNA. In embodiments, the nucleic acid sequences encoding (i) and (ii) are situated in the same orientation, e.g., transcription of the nucleic acid sequences encoding (i) and (ii) proceeds in the same direction. In embodiments, the nucleic acid sequences encoding (i) and (ii) are situated in different orientations. In embodiments, a single promoter controls expression of the nucleic acid sequences encoding (i) and (ii). In embodiments, a nucleic acid encoding a protease cleavage site (such as a T2A, P2A, E2A, or F2A cleavage site) is situated between the nucleic acid sequences encoding (i) and (ii). In embodiments, the protease cleavage site is placed such that a cell can express a fusion protein comprising (i) and (ii), which protein is subsequently processed into two peptides by proteolytic cleavage. In some embodiments, the nucleic acid sequences encoding (i) is upstream of the nucleic acid sequences encoding (ii), or the nucleic acid sequences encoding (ii) is upstream of the nucleic acid sequences encoding (i). In embodiments, a first promoter controls expression of the nucleic acid sequence encoding (i) and a second promoter controls expression of the nucleic acid sequence encoding (ii). In embodiments, the nucleic acid is a plasmid. In embodiments, the nucleic acid comprises a viral packaging element. In some aspects, the present disclosure provides a cell, e.g., an immune effector cell, comprising the nucleic acid described herein, e.g., a nucleic acid comprising (i) and (ii) as described above. The cell may comprise a protease (e.g., endogenous or exogenous) that cleaves a T2A, P2A, E2A, or F2A cleavage site.


In some aspects, the present disclosure provides, e.g., a composition comprising: (i) a first nucleic acid encoding a CAR molecule that binds CD19, e.g., a CAR molecule that binds CD19 described herein, e.g., a CD19 CAR, and (ii) a second nucleic acid encoding a CAR molecule that binds CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR. In embodiments, the first nucleic acid and second nucleic acid each comprises RNA or DNA.


In some aspects, the present disclosure provides, e.g., a vector comprising a nucleic acid or nucleic acids as described herein. The present disclosure also provides, in certain aspects, a cell comprising a vector or nucleic acid as described herein.


This disclosure also provides, in certain aspects, a composition comprising one or more immune effector cells and: (i) a first nucleic acid encoding, or a first polypeptide comprising, a CAR molecule that binds CD19, e.g., a CAR molecule that binds CD19 described herein, e.g., a CD19 CAR, and (ii) a second nucleic acid encoding, or a second polypeptide comprising, a CAR molecule that binds CD22, e.g., a CAR molecule that binds CD22 described herein, e.g., a CD22 CAR. In embodiments, the first nucleic acid or first polypeptide and the second nucleic acid or second polypeptide are each contained within, e.g., expressed by, a first immune effector cell. In embodiments, the composition comprises a first immune effector cell containing e.g., expressing the first nucleic acid or first polypeptide and a second immune effector cell containing e.g., expressing the second nucleic acid or second polypeptide. In embodiments, the composition does not comprise a cell containing, e.g., expressing, both of the first nucleic acid or first polypeptide and the second nucleic acid or second polypeptide.


Indications

In one embodiment, the disease is selected from a proliferative disease such as a cancer or malignancy or a precancerous condition such as a myelodysplasia, a myelodysplastic syndrome or a preleukemia, or is a non-cancer related indication. In one embodiment, the disease is associated with expression of an antigen, e.g., a tumor antigen, e.g., CD19 and/or CD22. In one embodiment, the disease is a solid or a liquid tumor. In one embodiment, the disease is a hematologic cancer, e.g., a leukemia or lymphoma. In one embodiment, the hematologic cancer is a lymphoma, e.g., a relapsed and/or refractory lymphoma. In one embodiment, the hematologic cancer is a leukemia, e.g., a relapsed and/or refractory leukemia. In one embodiment, the cancer is selected from the group consisting of one or more acute leukemias including but not limited to B-cell acute lymphoid leukemia (B-ALL), T-cell acute lymphoid leukemia (TALL), small lymphocytic leukemia (SLL), acute lymphoid leukemia (ALL) (e.g., relapsing and refractory ALL); one or more chronic leukemias including but not limited to chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL). Additional hematologic cancers or conditions include, but are not limited to mantle cell lymphoma (MCL), B cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma (DLBCL), follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, Marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia.” Preleukemia encompasses a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells In embodiments, a disease as used herein includes, but is not limited to atypical and/or non-classical cancers, malignancies, precancerous conditions; and any combination thereof.


In one embodiment, the disease associated with expression of CD22 and/or CD19 is a lymphoma, e.g., ALL, MCL or Hodgkin lymphoma.


In one embodiment, the disease is a leukemia, e.g., SLL, CLL and/or ALL, e.g., B cell ALL. In one embodiment, the disease is ALL, e.g., relapsed and/or refractory ALL. In one embodiment, the ALL, e.g., relapsed and/or refractory ALL is associated with expression of an antigen, e.g., a tumor antigen, e.g., CD19 and/or CD22. In one embodiment, a subject with relapsed and/or refractory ALL is a young adult or a pediatric subject, e.g., aged about 1-29 years, e.g., aged 1-29 years.


In one embodiment, the disease is associated with expression of an antigen, e.g., a tumor antigen. In one embodiment, the disease associated with a tumor antigen, e.g., a tumor antigen described herein, is selected from a proliferative disease such as a cancer or malignancy or a precancerous condition such as a myelodysplasia, a myelodysplastic syndrome or a preleukemia, or is a non-cancer related indication associated with expression of a tumor antigen described herein. In an embodiment, the disease associated with a tumor antigen described herein is a solid tumor, e.g., a solid tumor described herein, e.g., prostatic, colorectal, pancreatic, cervical, gastric, ovarian, head, or lung cancer.


In an embodiment, the cancer is chosen from AML, ALL, B-ALL, T-ALL, B-cell prolymphocytic leukemia, chronic lymphocytic leukemia, CML, hairy cell leukemia, Hodgkin lymphoma, mast cell disorder, myelodysplastic syndrome, myeloproliferative neoplasm, plasma cell myeloma, plasmacytoid dendritic cell neoplasm, or a combination thereof.


In an embodiment, the subject (e.g., a subject to be treated with a CD19 CAR and/or a CD2 CAR, optionally in combination with an additional agent such as a PD1 inhibitor or PD-L1 inhibitor) has, or is identified as having, at least 5%, 6%, 7%, 8%, 9%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of cancer cells which are CD3+/PD1+.


In an embodiment, the subject is predicted to have a relapse (e.g., has not relapsed), has relapsed or is identified as having relapsed after treatment with the one or more cells that express a CAR molecule that binds CD19, e.g., a CD19 CAR. In an embodiment, the subject is predicted to have a relapse (e.g., has not relapsed), has relapsed or is identified as having relapsed based on one or more of reappearance of blasts in the blood, bone marrow (>5%), or any extramedullary site, e.g., after a complete response. In an embodiment, the subject is predicted to have a relapse (e.g., has not relapsed), has relapsed or is identified as having relapsed based on detection of CD19 negative (CD19−) blasts above a predetermined threshold, e.g., over 1%, 2%, 3%, 4%, 5%, or 10%.


Car Therapies and Dosing Regimens

In certain embodiments, the method of treatment comprises a CAR therapy, e.g., administration of one or more cells that express one or more CAR molecules. A cell expressing one or more CAR molecules can be an immune effector cell, e.g., a T cell (e.g., a CD4+ or CD8+ T cell) or an NK cell. In an embodiment, the subject is a human.


In one embodiment, the cell expressing the CAR molecule comprises a vector that includes a nucleic acid sequence encoding the CAR molecule. In one embodiment, the vector is selected from the group consisting of a DNA, an RNA, a plasmid, a lentivirus vector, adenoviral vector, or a retrovirus vector. In one embodiment, the vector is a lentivirus vector. In one embodiment, the vector further comprises a promoter. In one embodiment, the promoter is an EF-1 promoter. In one embodiment, the EF-1 promoter comprises a sequence of SEQ ID NO: 100. In one embodiment, the vector is an in vitro transcribed vector, e.g., a vector that transcribes RNA of a nucleic acid molecule described herein. In one embodiment, the nucleic acid sequence in the in vitro vector further comprises a poly(A) tail, e.g., a poly A tail described herein, e.g., comprising about 150 adenosine bases. In one embodiment, the nucleic acid sequence in the in vitro vector further comprises a 3′UTR, e.g., a 3′ UTR described herein, e.g., comprising at least one repeat of a 3′UTR derived from human beta-globulin. In one embodiment, the nucleic acid sequence in the in vitro vector further comprises promoter. In one embodiment, the nucleic acid sequence comprises a T2A sequence.


In one embodiment, the cell expressing the CAR molecule (e.g., a CD19 CAR molecule or a CD22 CAR molecule is a cell described herein, e.g., a human T cell or a human NK cell, e.g., a human T cell described herein or a human NK cell described herein. In one embodiment, the human T cell is a CD8+ T cell. In one embodiment, the human T cell is a CD4+ T cell. In one embodiment, the human T cell is a CD4+/CD8+ T cell. In one embodiment the human T cell is a mixture of CD8+ and CD4+ T cells. In one embodiment, the human T cell is a CD3+ T cell. In one embodiment, the cell is an autologous T cell. In one embodiment, the cell is an allogeneic T cell. In one embodiment, the cell is a T cell and the T cell is diacylglycerol kinase (DGK) deficient. In one embodiment, the cell is a T cell and the T cell is Ikaros deficient. In one embodiment, the cell is a T cell and the T cell is both DGK and Ikaros deficient.


In another embodiment, the cell expressing the CAR molecule, e.g., as described herein, can further express another agent, e.g., an agent which enhances the activity of a CAR-expressing cell.


In one embodiment, the method includes administering a cell expressing the CAR molecule, as described herein, in combination with an agent which enhances the activity of a CAR-expressing cell, wherein the agent is a cytokine, e.g., IL-7, IL-15 (e.g., hetIL-15), IL-21, or a combination thereof. The cytokine can be delivered in combination with, e.g., simultaneously or shortly after, administration of the CAR-expressing cell. Alternatively, the cytokine can be delivered after a prolonged period of time after administration of the CAR-expressing cell, e.g., after assessment of the subject's response to the CAR-expressing cell.


For example, in one embodiment, the agent that enhances the activity of a CAR-expressing cell can be an agent which inhibits an immune inhibitory molecule. Examples of immune inhibitory molecules include PD1, PD-L1, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and TGFR beta. In one embodiment, the agent that inhibits an immune inhibitory molecule comprises a first polypeptide, e.g., an inhibitory molecule, associated with a second polypeptide that provides a positive signal to the cell, e.g., an intracellular signaling domain described herein. In one embodiment, the agent comprises a first polypeptide, e.g., of an immune inhibitory molecule such as PD1, PD-L1, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 or TGFR beta, or a fragment of any of these (e.g., at least a portion of the extracellular domain of any of these), and a second polypeptide which is an intracellular signaling domain described herein (e.g., comprising a costimulatory domain (e.g., 41BB, CD27 or CD28, e.g., as described herein) and/or a primary signaling domain (e.g., a CD3 zeta signaling domain described herein). In one embodiment, the agent comprises a first polypeptide of PD1 or a fragment thereof (e.g., at least a portion of the extracellular domain of PD1), and a second polypeptide of an intracellular signaling domain described herein (e.g., a CD28 signaling domain described herein and/or a CD3 zeta signaling domain described herein).


In one embodiment, lymphocyte infusion, for example allogeneic lymphocyte infusion, is used in the treatment of the cancer, wherein the lymphocyte infusion comprises at least one CD19 CAR-expressing cell described herein and/or at least one CD22 CAR-expressing cell. In one embodiment, autologous lymphocyte infusion is used in the treatment of the cancer, wherein the autologous lymphocyte infusion comprises at least one CD19-expressing cell and/or at least one CD22 CAR expressing cell.


In one embodiment, the CAR expressing cell, e.g., T cell, is administered to a subject that has received a previous stem cell transplantation, e.g., autologous or allogeneic stem cell transplantation.


In one embodiment, the CAR expressing cell, e.g., T cell, is administered to a subject that has received chemotherapy, e.g., lymphodepleting chemotherapy, e.g., as described herein.


In one embodiment, the CAR expressing cell, e.g., T cell, is administered to a subject that has received chemotherapy, e.g., bridging chemotherapy, e.g., as described herein.


In one embodiment, the cell expressing the CAR molecule, e.g., a CAR molecule described herein, is administered in combination with an agent that ameliorates one or more side effect associated with administration of a cell expressing a CD19 CAR molecule or with administration of a CD22 CAR molecule.


In one embodiment, the cell expressing the CAR molecules, e.g., a CD19 CAR molecule described herein and/or a CD22 CAR molecule described herein, are administered in combination with an additional agent that treats the disease associated with the tumor antigen, e.g., CD19 and/or CD22, e.g., an additional agent described herein.


In one embodiment, the cells expressing a CAR molecule, e.g., a CAR molecule described herein, are administered at a dose and/or dosing schedule described herein.


In one embodiment, the CAR molecule is introduced into T cells, e.g., using in vitro transcription, and the subject (e.g., human) receives an initial administration of cells comprising a CAR molecule, and one or more subsequent administrations of cells comprising a CAR molecule, wherein the one or more subsequent administrations are administered less than 15 days, e.g., 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 days after the previous administration. In one embodiment, more than one administration of cells comprising a CAR molecule are administered to the subject (e.g., human) per week, e.g., 2, 3, or 4 administrations of cells comprising a CAR molecule are administered per week. In one embodiment, the subject (e.g., human subject) receives more than one administration of cells comprising a CAR molecule per week (e.g., 2, 3 or 4 administrations per week) (also referred to herein as a cycle), followed by a week of no administration of cells comprising a CAR molecule, and then one or more additional administration of cells comprising a CAR molecule (e.g., more than one administration of the cells comprising a CAR molecule per week) is administered to the subject. In another embodiment, the subject (e.g., human subject) receives more than one cycle of cells comprising a CAR molecule, and the time between each cycle is less than 10, 9, 8, 7, 6, 5, 4, or 3 days. In one embodiment, the cells comprising a CAR molecule are administered every other day for 3 administrations per week. In one embodiment, the cells comprising a CAR molecule are administered for at least two, three, four, five, six, seven, eight or more weeks.


In embodiments, a CAR-expressing cell, e.g., a CD22 CAR-expressing cell described herein, is administered to the subject according to a dosing regimen comprising a total dose of cells administered to the subject by dose fractionation (e.g., split dosing), e.g., one, two, three or more separate administration of a partial dose. In embodiments, a total dose comprises one or more partial doses, e.g., 2, 3, 4, 5, or 6 partial doses, e.g., 3 partial doses. In embodiments, a first percentage of the total dose, e.g., the first partial dose, is administered on a first day of treatment, a second percentage of the total dose, e.g., the second partial dose, is administered on a subsequent (e.g., second, third, fourth, fifth, sixth, or seventh or later) day of treatment, and a third percentage (e.g., the remaining percentage) of the total dose, e.g., a third partial dose, is administered on a yet subsequent (e.g., third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, or later) day of treatment.


In an embodiment, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about about 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.02×107 to 1×107, about 1×107 to 1.5×107, about 1.5×107 to 2×107, about 2×107 to 2.5×107, about 2.5×107 to 3×107, about 3×107 to 3.5×107, about 3.5×107 to 4×107, about 4×107 to 4.5×107, or about 4.5×107 to 5×107 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.02×107, about 0.04×107, about 0.06×107, about 0.08×107, about 1×107, about 1.5×107, about 2×107, about 2.5×107, about 3×107, about 3.5×107, about 4×107, about 4.5×107, or about 5×107 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, the subject weighs less than 50 kg.


In an embodiment, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5×108 to 1×108, about 1×108 to 1.5×108, about 1.5×108 to 2×108, about 2×108 to 2.5×108, about 2.5×108 to 3×108, about 3×108 to 3.5×108, about 3.5×108 to 4×108, about 4×108 to 4.5×108, about 4.5×108 to 5×108, about 5×108 to 6×108, about 6×108 to 7×108, about 7×108 to 8×108, about 8×108 to 9×108, or about 9×108 to 10×108 CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5×108, about 1×108, about 1.5×108, about 2×108, about 2.5×108, about 3×108, about 3.5×108, about 4×108, about 4.5×108, about 5×108, about 6×108, about 7×108, about 8×108, about 9×108, about 10×108 CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, the subject weighs ≥50 kg.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15% (e.g., about 5-10%, 10-15%, 6-14%, 7-13%, 8-12%, or 9-11%), of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15%, e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15%, of the total dose. In an embodiment, when a subject weighs less than 50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 5-15% of the total dose. In some embodiments, when a subject weighs less than 50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 10% of the total dose (e.g., about 0.2×106 to 1×106) of the CAR-expressing cells when the total dose is about 0.2×107 to 1.0×107 cells/kg. In some embodiments, a first percentage of a total dose, e.g., a first partial dose, is administered, e.g., delivered or infused, on the first day. In some embodiments, when a subject weighs less than 50 kg, a first partial dose, e.g., about 10% of the total dose (e.g., about 0.2×106 to 1×106 cells/kg), comprises CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% (e.g., about 25-30%, 30-35%, 26-34%, 27-33%, 28-32%, or 29-31%) of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35%, e.g., about 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35%, of the total dose. In an embodiment, when a subject weighs less than 50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% of the total dose. In some embodiments, when a subject weighs less than 50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 30% of the total dose (e.g., about 0.6×106 to 3×106 cells/kg) of the CAR-expressing cells when the total dose is about 0.2×107 to 1.0×107 cells/kg. In some embodiments, a second percentage of a total dose, e.g., a second partial dose, is administered, e.g., delivered or infused, on the second day, e.g., second consecutive day. In some embodiments, when a subject weighs less than 50 kg, a second partial dose, e.g., about 30% of the total dose (e.g., about 0.6×106 to 3×106 cells/kg), comprises CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% (e.g., about 50-55%, 55-60%, 56-64, 57-63, 58-62, 59-61%) of the total dose. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65%, e.g., about 55, 56, 57, 58, 59, 60, 61, 62, 63, 64 or 65%, of the total dose. In an embodiment, when a subject weighs less than 50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% of the total dose. In some embodiments, when a subject weighs less than 50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 60% of the total dose (e.g., about 1.2×106 to 6×106 cells/kg) of the CAR-expressing cells when the total dose is about 0.2×107 to 1.0×107 cells/kg. In some embodiments, when a subject weighs less than 50 kg a third percentage of a total dose, e.g., a third partial dose, is administered, e.g., delivered or infused on the third day, e.g., third consecutive day. In some embodiments, when a subject weighs less than 50 kg a third partial dose, e.g., about 60% of the total dose (e.g., about 1.2×106 to 6×106 cells/kg), comprises CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15% (e.g., about 5-10%, 10-15%, 6-14%, 7-13%, 8-12%, or 9-11%), of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15%, e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15%, of the total dose. In an embodiment, when a subject weighs ≥50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 5-15% of the total dose. In some embodiments, when a subject weighs ≥50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 10% of the total dose (e.g., about 1×107 to 5×107 cells) of the CAR-expressing cells when the total dose is about 1×108 to 5×108 cells. In some embodiments, a first percentage of a total dose, e.g., a first partial dose, is administered, e.g., delivered or infused, on the first day. In some embodiments, when a subject weighs ≥50 kg, a first partial dose, e.g., about 10% of the total dose (e.g., about 1×107 to 5×107 cells), comprises CAR-expressing cells, e.g., CD22 CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% (e.g., about 25-30%, 30-35%, 26-34%, 27-33%, 28-32%, or 29-31%) of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35%, e.g., about 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35%, of the total dose. In an embodiment, when a subject weighs ≥50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% of the total dose. In some embodiments, when a subject weighs ≥50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 30% of the total dose (e.g., about 0.3×107 to 1.5×108 cells) of the CAR-expressing cells when the total dose is about 1×108 to 5×108 cells. In some embodiments, a second percentage of a total dose, e.g., a second partial dose, is administered, e.g., delivered or infused, on the second day, e.g., second consecutive day. In some embodiments, when a subject weighs ≥50 kg, a second partial dose, e.g., about 30% of the total dose (e.g., about 0.3×107 to 1.5×108 cells), comprises CAR-expressing cells, e.g., CD22 CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% (e.g., about 50-55%, 55-60%, 56-64, 57-63, 58-62, or 59-61%) of the total dose. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65%, e.g., about 55, 56, 57, 58, 59, 60, 61, 62, 63, 64 or 65%, of the total dose. In an embodiment, when a subject weighs ≥50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% of the total dose. In some embodiments, when a subject weighs ≥50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 60% of the total dose (e.g., about 0.6×108 to 3×108 cells) of the CAR-expressing cells when the total dose is about 1×108 to 5×108 cells. In some embodiments, when a subject ≥50 kg a third percentage of a total dose, e.g., a third partial dose, is administered, e.g., delivered or infused on the third day, e.g., third consecutive day. In some embodiments, when a subject weighs ≥50 kg a third partial dose, e.g., about 60% of the total dose (e.g., about 0.6×108 to 3×108 cells), comprises CAR-expressing cells, e.g., CD22 CAR-expressing cells.


In some embodiments, the first partial dose of CD22 CAR-expressing cells and the first partial dose of the CD19 CAR-expressing cells are administered, e.g., on the same day, e.g., the first day. In some embodiments, the first partial dose of CD22 CAR-expressing cells and the first partial dose of the CD19 CAR-expressing cells are administered consecutively, e.g., without any lapse of time between administrations, e.g., infusion.


In some embodiments, the second partial dose of CD22 CAR-expressing cells and the second partial dose of the CD19 CAR-expressing cells are administered, e.g., on the same day, e.g., the second day. In some embodiments, the second partial dose of CD22 CAR-expressing cells and the second partial dose of the CD19 CAR-expressing cells are administered consecutively, e.g., without any lapse of time between administrations, e.g., infusion.


In some embodiments, the third partial dose of CD22 CAR-expressing cells and the third partial dose of the CD19 CAR-expressing cells are administered, e.g., on the same day, e.g., the third day. In some embodiments, the third partial dose of CD22 CAR-expressing cells and the third partial dose of the CD19 CAR-expressing cells are administered consecutively, e.g., without any lapse of time between administrations, e.g., infusion.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of a total dose, e.g., a first partial dose comprising about 10%, e.g., 10%, of the total dose of cells is delivered on the first day. In an embodiment, a second percentage of a total dose, e.g., a second partial dose comprising about 30%, e.g., 30%, of the total dose of cells is delivered on the second day (e.g., second consecutive day). In an embodiment, a third percentage of a total dose, e.g., a third partial dose comprising, e.g., the remaining dose, of about 60%, e.g., 60%, of the total dose of cells is delivered on the third day of treatment, e.g., third consecutive day of treatment.


In one embodiment, a CD19 CAR-expressing cell described herein is administered to the subject according to a dosing regimen comprising a total dose of CD19 CAR-expressing cells administered to the subject by dose fractionation (e.g., split dosing), e.g., one, two, three of more separate administrations of a partial dose, e.g., three partial doses, of the CD19 CAR-expressing cells. In one embodiment, the total cell dose of the CD19 CAR-expressing cells is about 1-5×106 cells/kg, e.g., about 2.0×106 cells/kg. In an embodiment, a first percentage of the total dose, e.g., a first partial dose, comprising about 5-15%, e.g., about 0.1-0.3×106 cells/kg of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the first day. In an embodiment, a first percentage of the total dose, e.g., a first partial dose, comprising about 10% of the total dose (e.g., about 0.2×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the first day. In an embodiment, a second percentage of the total dose, e.g., a second partial dose, comprising about 25-35% of the total dose, e.g., about 0.5-0.7×106 cells/kg of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day). In an embodiment, a second percentage of the total dose, e.g., a second partial dose, comprising about 30% of the total dose (e.g., about 0.6×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day). In an embodiment, a third percentage of the total dose, e.g., a third partial dose comprising, e.g., comprising the remaining dose, about 55-65% of the total dose, e.g., about 1.1-1.3×106 cells/kg, of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive day of treatment. In an embodiment, a third percentage of the total dose, e.g., a third partial dose comprising, e.g., comprising the remaining dose, about 60% of the total dose (e.g., about 1.2×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive.


In one embodiment, a CD22 CAR-expressing cell described herein is administered to the subject according to a dosing regimen comprising a total dose of CD22 CAR-expressing cells administered to the subject by dose fractionation (e.g., split dosing), e.g., three separate administrations of a partial dose, e.g., three partial doses, of the CD22 CAR-expressing cells. In one embodiment, the total cell dose of the CD22 CAR-expressing cells is 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CD22 CAR-expressing cells/kg or about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CD22 CAR-expressing cells. In an embodiment, a first percentage of the total dose, e.g., a first partial dose, comprising about 5-15%, is administered, e.g., delivered or infused, on the first day. In an embodiment, a first percentage of the total dose, e.g., a first partial dose, comprising about 10% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the first day. In an embodiment, a second percentage of the total dose, e.g., a second partial dose, comprising about 25-35% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day). In an embodiment, a second percentage of the total dose, e.g., a second partial dose, comprising about 30% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day). In an embodiment, a third percentage of the total dose, e.g., a third partial dose comprising, e.g., comprising the remaining dose, about 55-65% of the total dose of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive day. In an embodiment, a third percentage of the total dose, e.g., a third partial dose, e.g., the remaining dose, comprising about 60% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive day.


In one embodiment, a CD22 CAR-expressing cell described herein, and a CD19 CAR-expressing cell described herein is administered to the subject according to a dosing regimen comprising a total dose of CD22 CAR-expressing cells and a total dose of CD19 CAR-expressing cells. In some embodiments, the CD22 CAR-expressing cell and the CD19 CAR-expressing cell is administered to the subject by dose fractionation (e.g., split dosing), e.g., three separate administrations of a partial dose, e.g., three partial doses, of each of the CD22 CAR-expressing cells and CD19 CAR-expressing cells. In one embodiment, the total cell dose of the CD22 CAR-expressing cells is about 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CD22 CAR-expressing cells/kg or about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CD22 CAR-expressing cells. In one embodiment, the total cell dose of the CD19 CAR-expressing cells is about 1-5×106 cells/kg, e.g., about 2.0×106 cells/kg. In one embodiment, the CD22-CAR expressing cells, administered according to a dosing regimen described herein, e.g., a dose fractionation regimen (e.g., split-dosing regimen), are administered before the administration of CD19 CAR-expressing cells. In an embodiment, a first percentage of the total dose of CD22 CAR-expressing cells, e.g., the first partial dose, comprises about 5-15% of the total dose of CD22 CAR-expressing cells. In an embodiment, a first percentage of the total dose of CD22 CAR-expressing cells, e.g., the first partial dose, comprises about 10% of the total dose of the CD22 CAR-expressing cells. In some embodiments, the first partial dose is administered, e.g., delivered or infused, on the first day. In some embodiments, a second percentage of the total dose of CD22 CAR-expressing cells, e.g., the second partial dose, comprises about 25-35% of the total dose of CD22 CAR-expressing cells. In some embodiments, a second percentage of the total dose of CD22 CAR-expressing cells, e.g., the second partial dose, comprises about 30% of the total dose of the CD22 CAR-expressing cells. In some embodiments, the second percentage of the total dose, e.g., the second partial dose, is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day). In some embodiments, a third percentage of the total dose, e.g., a third partial dose, e.g., the remaining dose, comprises about 55-65% of the total dose of CD22 CAR-expressing cells. In some embodiments, a third percentage of the total dose, e.g., a third partial dose, e.g., the remaining dose, comprises about 60% of the total dose of the CD22 CAR-expressing cells. In some embodiments, the third percentage of the total dose, e.g., the third partial dose, is administered, e.g., delivered or infused, on the third day, e.g., third consecutive day.


In one embodiment, the CD22 CAR-expressing cell described herein, and a CD19 CAR-expressing cell described herein are administered to the subject according to a dose regimen as described herein. In one embodiment, the total cell dose of the CD19 CAR-expressing cells is about 1.5×106 cells/kg, e.g., about 2.0×106 cells/kg. In an embodiment a first percentage of the total dose of CD19 CAR-expressing cells, e.g., the first partial dose, comprises about 5-15% of the total dose, e.g., about 0.1-0.3×106 cells/kg of CD19 CAR-expressing cells. In an embodiment, a first percentage of the total dose of CD19 CAR-expressing cells, e.g., the first partial dose, comprises about 10% of the total dose (e.g., about 0.2×106 cells/kg) of the CD19 CAR-expressing cells. In some embodiments, the first percentage of the total dose, e.g., the first partial dose, is administered, e.g., delivered or infused, on the first day. In some embodiments, a second percentage of the total dose of CD19 CAR-expressing cells, e.g., the second partial dose, comprises about 25-35% of the total dose, e.g., about 0.5-0.7×106 cells/kg of CD19 CAR-expressing cells. In some embodiments, a second percentage of the total dose of CD19 CAR-expressing cells, e.g., the second partial dose, comprises about 30% of the total dose (e.g., about 0.6×106 cells/kg) of the CD19 CAR-expressing cells. In some embodiments, the second percentage of the total dose, e.g., the second partial dose, is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day). In some embodiments, a third percentage of the total dose, e.g., a third partial dose, e.g., the remaining dose, comprises about 55-65% of the total dose of CD19 CAR-expressing cells, e.g., about 1.1-1.3×106 cells/kg. In some embodiments, a third percentage of the total dose, e.g., a third partial dose, e.g., the remaining dose, comprises about 60% of the total dose (e.g., about 1.2×106 cells/kg) of the CD19 CAR-expressing cells. In some embodiments, a third percentage of the total dose, e.g., third partial dose, is administered, e.g., delivered or infused, on the third day, e.g., third consecutive day.


In some embodiments, the first partial dose of the CD22 CAR-expressing cells and the first partial dose of the CD19 CAR-expressing cells are administered on the same day, e.g., the first day. In some embodiments, the first partial dose of the CD22 CAR-expressing cells and the first partial dose of the CD19 CAR-expressing cells are administered consecutively, e.g., without any lapse in time between each administration, e.g., infusion.


In some embodiments, the second partial dose of the CD22 CAR-expressing cells and the second partial dose of the CD19 CAR-expressing cells are administered on the same day, e.g., the second day, e.g., the second consecutive day. In some embodiments, the second partial dose of the CD22 CAR-expressing cells and the second partial dose of the CD19 CAR-expressing cells are administered consecutively, e.g., without any lapse in time between each administration, e.g., infusion.


In some embodiments, the third partial dose of the CD22 CAR-expressing cells and the third partial dose of the CD19 CAR-expressing cells are administered on the same day, e.g., the third day, e.g., the third consecutive day. In some embodiments, the third partial dose of the CD22 CAR-expressing cells and the third partial dose of the CD19 CAR-expressing cells are administered consecutively, e.g., without any lapse in time between each administration, e.g., infusion.


In one embodiment, the total dose of CD22 CAR-expressing cells comprising three separate administrations of a partial dose, e.g., a first percentage of total dose comprising a first partial dose, a second percentage of total dose comprising a second partial dose, and a third percentage of a total dose comprising a third partial dose of CD22-CAR expressing cells is administered prior to the administration of CD19 CAR-expressing cells.


In an embodiment, the CD19 CAR-expressing cell is administered after the administration of all three partial doses of the CD22 CAR-expressing cell, e.g., according to a dose fractionation regimen described herein.


In one embodiment, the therapy described herein (e.g., a CD22 CAR therapy) is administered to a subject as a first line treatment for a disease, e.g., cancer, e.g., a cancer described herein, e.g., a hematological cancer, e.g., ALL, e.g., B cell ALL, e.g., relapsed and/or refractory ALL. In another embodiment, the therapy described herein (e.g., a CD22 CAR therapy) is administered to a subject as a second, third, fourth, or fifth line treatment for a disease, e.g., a cancer, e.g., a cancer described herein, e.g., a hematological cancer, e.g., ALL, e.g., B cell ALL, e.g., relapsed and/or refractory ALL. In some embodiments, the subject has relapsed and/or is refractory to a prior line of treatment (e.g., as described herein), e.g., a first, second, or third line of treatment prior to administration of a CAR therapy described herein.


In one embodiment, a population of cells described herein, e.g., a population of cells expressing a CAR, e.g., a CD19 CAR, or a CD22 CAR is administered, e.g., delivered or infused. In some embodiments the population of cells is isolated or purified.


In one embodiment, the method includes administering a population of cells, a plurality of which comprise a CAR molecule described herein. In some embodiments, the population of CAR-expressing cells comprises a mixture of cells expressing different CARs. For example, in one embodiment, the population of CAR-expressing cells can include a first cell expressing a CAR having an anti-CD19 binding domain described herein, and a second cell expressing a CAR having an anti-CD22 binding domain. In embodiments, the first and second cell populations are T cells. In embodiments, the first and second populations of T cells are the same, e.g., the same isotype, e.g., are both CD4+ T cells, or are both CD8+ T cells. In other embodiments, the first and second populations of T cells are different, e.g., are of different isotypes, e.g., the first population comprises CD4+ T cells and the second population comprises CD8+ T cells. In embodiments, the first and second populations of T cells are cell types described in WO2012/129514, which is herein incorporated by reference in its entirety. As another example, a population of cells can comprise a single cell type that expresses both a CAR having an anti-CD19 binding domain described herein and a CAR having a an anti-CD22 antigen binding domain. As another example, the population of CAR-expressing cells can include a first cell expressing a CAR that includes an anti-CD19 binding domain, e.g., as described herein, and a second cell expressing a CAR that includes an anti-CD22 antigen binding domain. In one embodiment, the population of CAR-expressing cells includes, e.g., a first cell expressing a CAR that includes a primary intracellular signaling domain, and a second cell expressing a CAR that includes a secondary intracellular signaling domain. In one embodiment, the population of CAR-expressing cells includes, e.g., a first cell expressing a CAR that includes an intracellular signaling domain, and a second cell expressing a CAR that also includes an intracellular signaling domain, e.g., a same or different intracellular signaling domain. In one embodiment, the population of CAR-expressing cells includes, e.g., a first cell expressing a CAR that includes a first secondary signaling domain, and a second cell expressing a CAR that includes a secondary signaling domain different from the first secondary signaling domain.


In an embodiment, when the first CAR is a CD19 CAR and the first cell is a CD19 CAR-expressing cell, and the second CAR is a CD22-CAR and the second cell is a CD22 CAR-expressing cell, the first CAR and second CAR may be expressed by the same cell type or different types. For instance, in some embodiments, the cell expressing a CD19 CAR is a CD4+ T cell and the cell expressing a CD22 CAR is a CD8+ T cell, or the cell expressing a CD19 CAR is a CD8+ T cell and the cell expressing a CD22 CAR is a CD4+ T cell. In other embodiments, the cell expressing a CD19 CAR is a T cell and the cell expressing a CD22 CAR is a NK cell, or the cell expressing a CD19 CAR is a NK cell and the cell expressing a CD22 CAR is a T cell. In other embodiments, the cell expressing a CD19 CAR and the cell expressing a CD22 CAR are both NK cells or are both T cells, e.g., are both CD4+ T cells, or are both CD8+ T cells. In yet other embodiments, a single cell expresses the CD19 CAR and CD22 CAR, and this cell is, e.g., a NK cell or a T cell such as a CD4+ T cell or CD8+ T cell. The first CAR and second CAR can comprise the same or different intracellular signaling domains. For instance, in some embodiments the CD19 CAR comprises a CD3 zeta signaling domain and the CD22 CAR comprises a costimulatory domain, e.g., a 41BB, CD27 or CD28 costimulatory domain, while in some embodiments, the CD19 CAR comprises a costimulatory domain, e.g., a 41BB, CD27 or CD28 costimulatory domain and the CD22 CAR comprises a CD3 zeta signaling domain. In other embodiments, each of the CD19 CAR and the CD22 CAR comprises the same type of primary signaling domain, e.g., a CD3 zeta signaling domain, but the CD19 CAR and the CD22 CAR comprise different costimulatory domains, e.g., (1) the CD19 CAR comprises a 41BB costimulatory domain and the CD22 CAR comprises a different costimulatory domain e.g., a CD27 costimulatory domain, (2) the CD19 CAR comprises a CD27 costimulatory domain and the CD22 CAR comprises a different costimulatory domain e.g., a 41BB costimulatory domain, (3) the CD19 CAR comprises a 41BB costimulatory domain and the CD22 CAR comprises a CD28 costimulatory domain, (4) the CD19 CAR comprises a CD28 costimulatory domain and the CD22 CAR comprises a different costimulatory domain e.g., a 41BB costimulatory domain, (5) the CD19 CAR comprises a CD27 costimulatory domain and the CD22 CAR comprises a CD28 costimulatory domain, or (6) the CD19 CAR comprises a CD28 costimulatory domain and the CD22 CAR comprises a CD27 costimulatory domain. In another embodiment, a cell comprises a CAR that comprises both a CD19 antigen-binding domain and a CD22 antigen-binding domain.


In one embodiment, the 4-1BB costimulatory domain comprises a sequence of SEQ ID NO: 16. In one embodiment, the 4-1BB costimulatory domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO: 16, or a sequence with at least 95, 96, 97, 98 or 99% identity to an amino acid sequence of SEQ ID NO:16. In one embodiment, the 4-1BB costimulatory domain is encoded by a nucleic acid sequence of SEQ ID NO:60, or a sequence with at least 95, 96, 97, 98 or 99% identity thereof.


In one embodiment, the CD27 costimulatory domain comprises a sequence of SEQ ID NO: 16. In one embodiment, the CD27 costimulatory domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO: 16, or a sequence with at least 95, 96, 97, 98 or 99% identity to an amino acid sequence of SEQ ID NO:16. In one embodiment, the CD27 costimulatory domain is encoded by a nucleic acid sequence of SEQ ID NO:17, or a sequence with at least 95, 96, 97, 98 or 99% identity thereof.


In one embodiment, the CD28 costimulatory domain comprises a sequence of SEQ ID NO: 1317. In one embodiment, the CD28 costimulatory domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO: 1317, or a sequence with at least 95, 96, 97, 98 or 99% identity to an amino acid sequence of SEQ ID NO:1317. In one embodiment, the CD28 costimulatory domain is encoded by a nucleic acid sequence of SEQ ID NO:1318, or a sequence with at least 95, 96, 97, 98 or 99% identity thereof.


In one embodiment, the wild-type ICOS costimulatory domain comprises a sequence of SEQ ID NO: 1319. In one embodiment, the wild-type ICOS costimulatory domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO: 1319, or a sequence with at least 95, 96, 97, 98 or 99% identity to an amino acid sequence of SEQ ID NO: 1319. In one embodiment, the wild-type ICOS costimulatory domain is encoded by a nucleic acid sequence of SEQ ID NO: 1320, or a sequence with at least 95, 96, 97, 98 or 99% identity thereof.


In one embodiment, the Y to F mutant ICOS costimulatory domain comprises a sequence of SEQ ID NO: 1321. In one embodiment, the Y to F mutant ICOS costimulatory domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO: 1321, or a sequence with at least 95, 96, 97, 98 or 99% identity to an amino acid sequence of SEQ ID NO: 1321. In one embodiment, the Y to F mutant ICOS costimulatory domain is encoded by a nucleic acid sequence with at least 95, 96, 97, 98 or 99% identity to a nucleic acid sequence of SEQ ID NO:1320 (wherein SEQ ID NO: 1320 encodes wild-type ICOS).


In embodiments, the primary signaling domain comprises a functional signaling domain of CD3 zeta. In embodiments, the functional signaling domain of CD3 zeta comprises SEQ ID NO: 17 (mutant CD3 zeta) or SEQ ID NO: 43 (wild-type human CD3 zeta).


In one embodiment, the method includes administering a population of cells wherein at least one cell in the population expresses a CAR, e.g., having an anti-CD19 domain described herein and/or an anti-CD22 binding domain described herein, and an agent which enhances the activity of a CAR-expressing cell, e.g., a second cell expressing the agent which enhances the activity of a CAR-expressing cell. For example, in one embodiment, the agent can be an agent which inhibits an immune inhibitory molecule. Examples of immune inhibitory molecules include PD1, PD-L1, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and TGFR beta. In one embodiment, the agent that inhibits an immune inhibitory molecule comprises a first polypeptide, e.g., an inhibitory molecule, associated with a second polypeptide that provides a positive signal to the cell, e.g., an intracellular signaling domain described herein. In one embodiment, the agent comprises a first polypeptide, e.g., of an inhibitory molecule such as PD1, PD-L1, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 or TGFR beta, or a fragment of any of these (e.g., at least a portion of an extracellular domain of any of these), and a second polypeptide which is an intracellular signaling domain described herein (e.g., comprising a costimulatory domain (e.g., 41BB, CD27 or CD28, e.g., as described herein) and/or a primary signaling domain (e.g., a CD3 zeta signaling domain described herein). In one embodiment, the agent comprises a first polypeptide of PD1 or a fragment thereof (e.g., at least a portion of the extracellular domain of PD1), and a second polypeptide of an intracellular signaling domain described herein (e.g., a CD28 signaling domain described herein and/or a CD3 zeta signaling domain described herein).


In an embodiment, the method further comprises transplanting a cell, e.g., a hematopoietic stem cell, or a bone marrow cell, into the mammal.


In another aspect, the invention pertains to a cell expressing a CAR molecule described herein, e.g., a CD19 CAR molecule or a CD22 CAR molecule, for use as a medicament.


In another aspect, the invention pertains to a cell expressing a CAR molecule described herein, e.g., a CD19 CAR molecule or a CD22 CAR molecule, for use in the manufacture of a medicament for treating a disease, e.g., a cancer, (e.g., a hematological cancer, e.g., ALL, e.g., relapsed and/or refractory ALL) or a disease associated with expression of CD19 and/or CD22.


In another aspect, the invention pertains to a cell expressing a CAR molecule described herein, e.g., a CD19 CAR molecule or a CD22 CAR molecule for use in the treatment of a disease e.g., a cancer, (e.g., a hematological cancer, e.g., ALL, e.g., relapsed and/or refractory ALL) or a disease associated with expression of CD19 and/or CD22.


In one embodiment, the method includes administering a population of cells wherein at least one cell in the population expresses a therapy herein (e.g., a CD22 CAR, or a CD19 CAR) and an agent which enhances the activity of a CAR-expressing cell, wherein the agent is a cytokine, e.g., IL-7, IL-15 (e.g., hetIL-15), IL-21, or a combination thereof. The cytokine can be delivered in combination with, e.g., simultaneously or shortly after, administration of the CAR-expressing cell(s). Alternatively, the cytokine can be delivered after a prolonged period of time after administration of the CAR-expressing cell(s), e.g., after assessment of the subject's response to the CAR-expressing cell(s). Related compositions for use and methods of making a medicament are also provided.


In one embodiment, the cells described herein (e.g., cells expressing a CD22 CAR molecule, or cells expressing a CD19 CAR molecule) are administered in combination with an agent that increases the efficacy of a cell expressing a CAR molecule or one of the inhibitors, e.g., an agent described herein.


In one embodiment, the cells described herein (e.g., cells expressing a CD22 CAR molecule, or expressing a CD19 CAR molecule) are administered in combination with an agent that ameliorates one or more side effect associated with administration of a cell expressing a CAR molecule or one of the inhibitors, e.g., an agent described herein.


In another aspect, the invention features a composition comprising a cell expressing a Chimeric Antigen Receptor (CAR) molecule that binds CD19, in combination with a cell expressing a CAR molecule that binds CD22. The CD19 CAR-expressing cell and the CD22 CAR-expressing cell can be present in a single dose form, or as two or more dose forms.


In an embodiment, the composition is a pharmaceutically acceptable composition.


In embodiments, the compositions disclosed herein (e.g., nucleic acids, vectors, or cells) are for use as a medicament.


In embodiments, the compositions disclosed herein are use in the treatment of a disease associated with expression of a B-cell antigen (e.g., CD19 or CD22), e.g., a B-cell leukemia or lymphoma, e.g., B-cell ALL, e.g., relapsed or refractory B-cell ALL.


CD22 CARs

In some aspects, the present disclosure provides a CD22 CAR molecule comprising an anti-CD22 binding domain, e.g., a CD22 binding domain as described herein. The disclosure also provides a nucleic acid encoding the CD22 binding domain, e.g., encoding a CAR comprising the CD22 binding domain. The composition may also comprise a second agent, e.g., an anti-CD19 CAR-expressing cell or a CD19 binding domain. The agents may be, e.g., encoded by a single nucleic acid or different nucleic acids.


In some aspects, a CD22 CAR comprising an anti-CD22 binding domain, e.g., a CD22 CAR-expressing cell, is administered as a monotherapy. In some aspects, the CD22 CAR comprising an anti-CD22 binding domain, e.g., a CD22 CAR-expressing cell, is administered in combination with a second agent such as an anti-CD19 CAR-expressing cell.


In another aspect, the invention pertains to a CD22 binding domain, or a CAR molecule, comprising the amino acid sequence of the heavy chain variable domain (VH) of CD22-65sKD, e.g., comprising the amino acid sequence of SEQ ID NO: 839; and/or the amino acid sequence of the light chain variable domain (VL) of CD22-65sKD, e.g., comprising the amino acid sequence of SEQ ID NO: 840. In embodiments, the VH and VL sequences are connected directly, e.g., without a linker. In embodiments, the VH and VL sequences are connected via a linker. In some embodiments, the linker is a (Gly4-Ser)n linker, wherein n is 0, 1, 2, 3, 4, 5, or 6 (SEQ ID NO: 53). In some embodiments, there is no linker between the VH region of CD22-65sKD and the VL region of CD22-65KD, e.g., n is 0. In one embodiment, the linker is a (Gly4-Ser)n linker, wherein n is 1 (SEQ ID NO: 18). In some embodiments, the CD22 binding domain comprises the amino acid sequence of CD22-65sKD scFv, e.g., comprising the amino acid sequence of SEQ ID NO: 837.


In another aspect, the invention pertains to a CD22 binding domain, or a CAR molecule, comprising the amino acid sequence of an scFv of CD22-65s (a (Gly4-Ser)n linker, wherein n is 1 (SEQ ID NO: 18)) or CD22-65ss (no linker). In some embodiments, the CD22 binding domain comprises the scFv of SEQ ID NO: 835. In some embodiments, the CD22 binding domain comprises the scFv of SEQ ID NO: 836.


In an aspect, the invention pertains to a CD22 binding domain, or a CAR molecule, comprising the amino acid sequence of the light chain variable region (VL) of murine anti-CD22 antibody m971, e.g., comprising the amino acid sequence of SEQ ID NO: 1333, or an amino acid sequence with at least 95% identity thereto; and/or the heavy chain variable region (VH) of murine anti-CD22 antibody m971, e.g., comprising the amino acid sequence of SEQ ID NO: 1332, or an amino acid sequence with at least 95% identity thereto.


In some embodiments, a CD22 CAR molecule described herein comprises a CD22 binding domain comprising the light chain variable region of SEQ ID NO: 1333, or an amino acid sequence with at least 95% identity thereto; and/or the heavy chain variable region of SEQ ID NO: 1332, or an amino acid sequence with at least 95% identity thereto.


The invention also pertains to nucleic acid molecules, vectors, cells and uses comprising any of the foregoing aspects or embodiments.


In one embodiment, the CD22 binding domain is an anti-CD22 antibody or fragment thereof. In an embodiment, the antibody is a monospecific antibody and in another embodiment the antibody is a bispecific antibody. In an embodiment, the antibody is a monospecific antibody, optionally conjugated to a second agent such as a chemotherapeutic agent. For instance, in an embodiment the antibody is an anti-CD22 monoclonal antibody-MMAE conjugate (e.g., DCDT2980S). In an embodiment, the antibody is an scFv of an anti-CD22 antibody, e.g., an scFv of antibody RFB4. This scFv can be fused to all of or a fragment of Pseudomonas exotoxin-A (e.g., BL22). In an embodiment, the antibody is a humanized anti-CD22 monoclonal antibody (e.g., epratuzumab). In an embodiment, the antibody or fragment thereof comprises the Fv portion of an anti-CD22 antibody, which is optionally covalently fused to all or a fragment or (e.g., a 38 KDa fragment of) Pseudomonas exotoxin-A (e.g., moxetumomab pasudotox). In an embodiment, the anti-CD22 antibody is an anti-CD19/CD22 bispecific antibody, optionally conjugated to a toxin. For instance, in one embodiment, the anti-CD22 antibody comprises an anti-CD19/CD22 bispecific portion, (e.g., two scFv ligands, recognizing human CD19 and CD22) optionally linked to all of or a portion of diphtheria toxin (DT), e.g., first 389 amino acids of diphtheria toxin (DT), DT 390, e.g., a ligand-directed toxin such as DT2219ARL). In another embodiment, the bispecific portion (e.g., anti-CD19/anti-CD22) is linked to a toxin such as deglycosylated ricin A chain (e.g., Combotox).


In one embodiment, the CD22 CAR molecule is an anti-CD22 expressing cell, e.g., a CD22 CART or CD22-expressing NK cell.


In one aspect, the present disclosure provides a population of CAR-expressing cells, e.g., CART cells, comprising a mixture of cells expressing CD19 CARs and CD22 CARs. For example, in one embodiment, the population of CART cells can include a first cell expressing a CD19 CAR and a second cell expressing a CD22 CAR. As another example, the population of CAR T cells can include a single population expressing more than one, e.g., 2, 3, 4, 5, or 6 or more, CARs, e.g., a CD19 CAR and a CD22 CAR.


In some embodiments, the CD22-CAR comprises an optional leader sequence (e.g., an optional leader sequence described herein), an extracellular antigen binding domain, a hinge (e.g., hinge described herein), a transmembrane domain (e.g., transmembrane domain described herein), and an intracellular stimulatory domain (e.g., intracellular stimulatory domain described herein). In one embodiment, an exemplary CD22 CAR construct comprises an optional leader sequence (e.g., a leader sequence described herein), an extracellular antigen binding domain, a hinge, a transmembrane domain, an intracellular costimulatory domain (e.g., an intracellular costimulatory domain described herein) and an intracellular stimulatory domain.


In one embodiment, the CD22 binding domain comprises one or more (e.g., all three) light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of a CD22 binding domain described herein, and/or one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of a CD22 binding domain described herein, e.g., a CD22 binding domain comprising one or more, e.g., all three, LC CDRs and one or more, e.g., all three, HC CDRs. These CDRs may be, e.g., one or more CDRs of Table 6-10. In one embodiment, the CD22 binding domain comprises one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of a CD22 binding domain described herein, e.g., the CD22 binding domain has two variable heavy chain regions, each comprising a HC CDR1, a HC CDR2 and a HC CDR3 described herein. In one embodiment, the CD22 binding domain comprises a light chain variable region described herein (e.g., in Table 6 or 10) and/or a heavy chain variable region described herein (e.g., in Table 6 or 9). In one embodiment, the CD22 binding domain comprises a heavy chain variable region described herein (e.g., in Table 6 or 9), e.g., at least two heavy chain variable regions described herein (e.g., in Table 6 or 9). In one embodiment, the CD22 binding domain is a scFv comprising a light chain and a heavy chain of an amino acid sequence of Tables 6-10. In an embodiment, the CD22 binding domain (e.g., an scFv) comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 6 or 10, or a sequence with 95-99% identity with an amino acid sequence of Table 6 or 10; and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 6 or 9, or a sequence with 95-99% identity to an amino acid sequence of Table 6 or 9. The CD22 binding domain may be part of, e.g., an antibody molecule or a CAR molecule.


In one embodiment, the CAR molecule comprises an anti-CD22 binding domain that includes one or more (e.g., 2, 3, 4, 5, or 6) LC CDR1, LC CDR2, LC CDR3, HC CDR1, HC CDR2, and HC CDR3 of a construct of Table 6-10, e.g., CD22-65s, CD22-65ss, CD22-65sKD, CD22-65, CD22-57, CD22-58, CD22-59, CD22-60, CD22-61, CD22-62, CD22-63, or CD22-64, CD22 m971 or a sequence with at least 95% identity thereto.


In one embodiment, the CAR molecule comprises an anti-CD22 binding domain that includes a VL and/or VH of a construct of Table 6-10, e.g., CD22-65s, CD22-65ss, CD22-65sKD, CD22-65, CD22-57, CD22-58, CD22-59, CD22-60, CD22-61, CD22-62, CD22-63, or CD22-64, CD22 m971 or a sequence with at least 95% identity thereto.


The scFv may be preceded by an optional leader sequence such as provided in SEQ ID NO: 13, and followed by an optional hinge sequence such as provided in SEQ ID NO: 14 or SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49, a transmembrane region such as provided in SEQ ID NO:15, an intracellular signalling domain that includes SEQ ID NO:16 or SEQ ID NO:51 and a CD3 zeta sequence that includes SEQ ID NO:17 or SEQ ID NO:43, e.g., wherein the domains are contiguous with and in the same reading frame to form a single fusion protein.


Further embodiments include a nucleotide sequence that encodes a polypeptide of any of Tables 6-10. Further embodiments include a nucleotide sequence that encodes a polypeptide of any of Tables 6-10, and each of the domains of SEQ ID NOS: 13, 14, 15, 16, 17, and optionally 51.


In one embodiment, the CD22 binding domain is characterized by particular functional features or properties of an antibody or antibody fragment. For example, in one embodiment, the portion of a CAR composition of the invention that comprises an antigen binding domain specifically binds human CD22 or a fragment thereof.


In one embodiment, the CD22 binding domain is a fragment, e.g., a single chain variable fragment (scFv). In one embodiment, the CD22 binding domain is a Fv, a Fab, a (Fab′)2, or a bi-functional (e.g. bi-specific) hybrid antibody (e.g., Lanzavecchia et al., Eur. J. Immunol. 17, 105 (1987)). In one aspect, the antibodies and fragments thereof of the invention binds a CD22 protein or a fragment thereof with wild-type or enhanced affinity. In some instances, a human scFv can be derived from a display library.


In one embodiment, the CD22 binding domain, e.g., scFv comprises at least one mutation such that the mutated scFv confers improved stability to the CART22 construct. In another embodiment, the CD22 binding domain, e.g., scFv comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 mutations arising, e.g., from the humanization process such that the mutated scFv confers improved stability to the CART22 construct.


In one embodiment, the present disclosure provides a population of CAR-expressing cells, e.g., CART cells, comprising a mixture of cells expressing CD19 CARs and CD22 CARs. For example, in one embodiment, the population of CART cells can include a first cell expressing a CD19 CAR and a second cell expressing a CD22 CAR.


In some aspects, a binding domain or antibody molecule described herein binds the same (or substantially the same) or an overlapping (or substantially overlapping) epitope with a second antibody molecule to CD22, wherein the second antibody molecule is an antibody molecule described herein, e.g., an antibody molecule chosen from Tables 6-10. In some embodiments, a binding domain or antibody molecule described herein competes for binding, and/or binds the same (or substantially the same) or overlapping (or substantially overlapping) epitope, with a second antibody molecule to CD22, wherein the second antibody molecule is an antibody molecule described herein, e.g., an antibody molecule chosen from Tables 6-10. In some embodiments, a biparatopic CD22 binding domain binds a first epitope, e.g., an epitope bound by an antibody molecule chosen from Tables 6-10, and the biparatopic binding domain also binds a second epitope, e.g., a second epitope bound by an antibody molecule chosen from Tables 6-10. In some aspects, the present disclosure provides a method of treatment comprising administering a first CD22 binding domain that binds a first epitope, e.g., an epitope bound by an antibody molecule chosen from Tables 6-10 and a second CD22 binding domain that binds a second epitope, e.g., a second epitope bound by an antibody molecule chosen from Tables 6-10. In some embodiments, the CD22 binding domains are part of CAR molecules, e.g., expressed by a CAR-expressing cell.


In some embodiments, a CD22 binding domain binds to one or more of Ig-like domains 1, 2, 3, 4, 5, 6, or 7 of CD22. In some embodiments, the CD22 binding domain binds to domains 1 and 2; to domains 3 and 4; or to domains 5, 6, and 7.


In some aspects, this disclosure provides a method of treating a CD19-negative cancer, e.g., a leukemia, e.g., an ALL, e.g., B-ALL, comprising administering a CD22 inhibitor, e.g., a CD22 binding domain or CD22 CAR-expressing cell described herein. In some embodiments, the method includes a step of determining whether the cancer is CD19-negative. In some embodiments, the subject has received a CD19 inhibitor, e.g., a CD19 CAR-expressing cell, and is resistant, relapsed, or refractory to the CD19 inhibitor.


CD19 CARs

In one embodiment, the cell expresses a CAR molecule comprising an anti-CD19 binding domain (e.g., a murine or humanized antibody or antibody fragment that specifically binds to CD19), a transmembrane domain, and an intracellular signaling domain (e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain). In one embodiment, the CAR comprises an antibody or antibody fragment which includes an anti-CD19 binding domain described herein (e.g., a murine or humanized antibody or antibody fragment that specifically binds to CD19 as described herein), a transmembrane domain described herein, and an intracellular signaling domain described herein (e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain described herein).


In one embodiment, the CAR molecule comprises an anti-CD19 binding domain comprising one or more (e.g., all three) light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of an anti-CD19 binding domain described herein (e.g., one or more (e.g., all three) LC CDRs from Table 5), and one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of an anti-CD19 binding domain described herein (e.g., one or more (e.g., all three) HC CDRs from Table 4), e.g., an anti-CD19 binding domain comprising one or more, e.g., all three, LC CDRs and one or more, e.g., all three, HC CDRs. In one embodiment, the anti-CD19 binding domain comprises one or more (e.g., all three) HC CDR1, HC CDR2, and HC CDR3 of an anti-CD19 binding domain described herein, e.g., the anti-CD19 binding domain has two variable heavy chain regions, each comprising a HC CDR1, a HC CDR2 and a HC CDR3 described herein. In one embodiment, the anti-CD19 binding domain comprises a murine light chain variable region described herein (e.g., in Table 3, e.g., the murine light chain variable region of SEQ ID NO:59) and/or a murine heavy chain variable region described herein (e.g., in Table 3, e.g., the murine heavy chain variable region of SEQ ID NO:59). In one embodiment, the anti-CD19 binding domain is a scFv comprising a murine light chain and a murine heavy chain of an amino acid sequence of Table 3, e.g., the scFv of SEQ ID NO:59. In an embodiment, the anti-CD19 binding domain (e.g., an scFv) comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 3 (e.g., the murine light chain variable region of SEQ ID NO:59), or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity with an amino acid sequence of Table 3 (e.g., the murine light chain variable region of SEQ ID NO:59); and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 3 (e.g., the murine heavy chain variable region of SEQ ID NO:59), or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to an amino acid sequence of Table 3 (e.g., the heavy chain variable region of SEQ ID NO:59). In one embodiment, the anti-CD19 binding domain comprises a sequence of SEQ ID NO:59, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereof. In one embodiment, the anti-CD19 binding domain is a scFv, and a light chain variable region comprising an amino acid sequence described herein, e.g., in Table 3, is attached to a heavy chain variable region comprising an amino acid sequence described herein, e.g., in Table 3, via a linker, e.g., a linker described herein. In one embodiment, the anti-CD19 binding domain includes a (Gly4-Ser)n linker, wherein n is 1, 2, 3, 4, 5, or 6, e.g., 3 or 4 (SEQ ID NO: 53). The light chain variable region and heavy chain variable region of a scFv can be, e.g., in any of the following orientations: light chain variable region-linker-heavy chain variable region or heavy chain variable region-linker-light chain variable region.


In one embodiment, the CAR molecule comprises a humanized anti-CD19 binding domain that includes one or more (e.g., all three) light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of a humanized anti-CD19 binding domain described herein (e.g., one or more (e.g., all three) LC CDRs from Table 5), and one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of a humanized anti-CD19 binding domain described herein (e.g., one or more (e.g., all three) HC CDRs from Table 4), e.g., a humanized anti-CD19 binding domain comprising one or more, e.g., all three, LC CDRs and one or more, e.g., all three, HC CDRs. In one embodiment, the humanized anti-CD19 binding domain comprises at least HC CDR2. In one embodiment, the humanized anti-CD19 binding domain comprises one or more (e.g., all three) HC CDR1, HC CDR2, and HC CDR3 of a humanized anti-CD19 binding domain described herein, e.g., the humanized anti-CD19 binding domain has two variable heavy chain regions, each comprising a HC CDR1, a HC CDR2 and a HC CDR3 described herein. In one embodiment, the humanized anti-CD19 binding domain comprises at least HC CDR2. In one embodiment, the light chain variable region comprises one, two, three or all four framework regions of VK3_L25 germline sequence. In one embodiment, the light chain variable region has a modification (e.g., substitution, e.g., a substitution of one or more amino acid found in the corresponding position in the murine light chain variable region of SEQ ID NO: 58, e.g., a substitution at one or more of positions 71 and 87). In one embodiment, the heavy chain variable region comprises one, two, three or all four framework regions of VH4_4-59 germline sequence. In one embodiment, the heavy chain variable region has a modification (e.g., substitution, e.g., a substitution of one or more amino acid found in the corresponding position in the murine heavy chain variable region of SEQ ID NO: 58, e.g., a substitution at one or more of positions 71, 73 and 78). In one embodiment, the humanized anti-CD19 binding domain comprises a light chain variable region described herein (e.g., in Table 2, e.g., any of the light chain variable regions of SEQ ID NOs:1-12, e.g., the light chain variable region of SEQ ID NO:2) and/or a heavy chain variable region described herein (e.g., in Table 2, e.g., any of the heavy chain variable regions of SEQ ID NOs:1-12, e.g., the heavy chain variable region of SEQ ID NO:2). In one embodiment, the humanized anti-CD19 binding domain is a scFv comprising a light chain and a heavy chain of an amino acid sequence of Table 2 e.g., any of the scFvs of SEQ ID NOs:1-12, e.g., the scFv of SEQ ID NO:2). In an embodiment, the humanized anti-CD19 binding domain (e.g., an scFv) comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 2 (e.g., any of the light chain variable regions of SEQ ID NOs:1-12, e.g., the light chain variable region of SEQ ID NO:2), or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity with an amino acid sequence of Table 2 (e.g., any of the light chain variable regions of SEQ ID NOs:1-12, e.g., the light chain variable region of SEQ ID NO:2); and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 2 e.g., any of the heavy chain variable regions of SEQ ID NOs:1-12, e.g., the heavy chain variable region of SEQ ID NO:2), or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to an amino acid sequence of Table 2 (e.g., any of the heavy chain variable regions of SEQ ID NOs:1-12, e.g., the heavy chain variable region of SEQ ID NO:2). In one embodiment, the humanized anti-CD19 binding domain comprises a sequence selected from a group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO: 4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11 and SEQ ID NO:12, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereof. In one embodiment, the humanized anti-CD19 binding domain is a scFv, and a light chain variable region comprising an amino acid sequence described herein, e.g., in Table 2, is attached to a heavy chain variable region comprising an amino acid sequence described herein, e.g., in Table 2, via a linker, e.g., a linker described herein. In one embodiment, the humanized anti-CD19 binding domain includes a (Gly4-Ser)n linker, wherein n is 1, 2, 3, 4, 5, or 6, e.g., 3 or 4 (SEQ ID NO: 53). The light chain variable region and heavy chain variable region of a scFv can be, e.g., in any of the following orientations: light chain variable region-linker-heavy chain variable region or heavy chain variable region-linker-light chain variable region.


In one embodiment, the CAR molecule comprises an anti-CD19 binding domain that includes one or more (e.g., 2, 3, 4, 5, or 6) LC CDR1, LC CDR2, LC CDR3, HC CDR1, HC CDR2, and HC CDR3 of a construct of Table 4 and 5, e.g., murine_CART19, humanized_CART19 a, humanized_CART19 b, or humanized_CART19 c.


In one embodiment, the CD19 CAR molecule comprises an anti-CD19 binding domain comprising a heavy chain variable region and/or a light chain variable region, e.g., as described in Table 2. In one embodiment, the CD19 CAR molecule comprises an anti-CD19 binding domain comprising the amino acid sequence of SEQ ID NO: 2, or an amino acid sequence having at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to the amino acid sequence of SEQ ID NO: 2.


In one embodiment, the CD19 CAR molecule comprises an anti-CD19 binding domain comprising a heavy chain variable region and/or a light chain variable region, e.g., as described in Table 3. In one embodiment, the CD19 CAR molecule comprises an anti-CD19 binding domain comprising the amino acid sequence of SEQ ID NO: 59, or an amino acid sequence having at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to the amino acid sequence of SEQ ID NO: 59.


In one embodiment, the CAR molecule comprises a leader sequence, e.g., a leader sequence described herein, e.g., a leader sequence of SEQ ID NO: 13, or having 95-99% identity thereof; an anti-CD19 binding domain described herein, e.g., an anti-CD19 binding domain comprising a LC CDR1, a LC CDR2, a LC CDR3, a HC CDR1, a HC CDR2 and a HC CDR3 described herein, e.g., a murine anti-CD19 binding domain described in Table 3, a humanized anti-CD19 binding domain described in Table 2, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereof; a hinge region, e.g., a hinge region described herein, e.g., a hinge region of SEQ ID NO:14 or having at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereof; a transmembrane domain, e.g., a transmembrane domain described herein, e.g., a transmembrane domain having a sequence of SEQ ID NO:15 or a sequence having at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereof; an intracellular signaling domain, e.g., an intracellular signaling domain described herein (e.g., an intracellular signaling domain comprising a costimulatory domain and/or a primary signaling domain). In one embodiment, the intracellular signaling domain comprises a costimulatory domain, e.g., a costimulatory domain described herein, e.g., a 4-1BB costimulatory domain having a sequence of SEQ ID NO:16 or SEQ ID NO:51, or having at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereof, and/or a primary signaling domain, e.g., a primary signaling domain described herein, e.g., a CD3 zeta stimulatory domain having a sequence of SEQ ID NO:17 or SEQ ID NO:43, or having at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereof.


In one embodiment, the CAR molecule comprises (e.g., consists of) an amino acid sequence of SEQ ID NO:58, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41 or SEQ ID NO:42, or an amino acid sequence having at least one, two, three, four, five, 10, 15, 20 or 30 modifications (e.g., substitutions) but not more than 60, 50 or 40 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO:58, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41 or SEQ ID NO:42, or an amino acid sequence having 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to an amino acid sequence of SEQ ID NO:58, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41 or SEQ ID NO:42. In one embodiment, the CAR molecule comprises the amino acid sequence of SEQ ID NO: 32, of an amino acid sequence having at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to the amino acid sequence of SEQ ID NO: 32.


Car Molecules

The binding domains described herein (e.g., binding domains against one or more of CD19, or CD22) may further comprise one or more additional amino acid sequences.


In one embodiment, the CAR molecule comprises a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154. In one embodiment, the transmembrane domain comprises a sequence of SEQ ID NO: 15. In one embodiment, the transmembrane domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO: 15, or a sequence with 95-99% identity to an amino acid sequence of SEQ ID NO: 15.


In one embodiment, the binding domain is connected to the transmembrane domain by a hinge region, e.g., a hinge region described herein. In one embodiment, the encoded hinge region comprises SEQ ID NO:14 or SEQ ID NO:45, or a sequence with 95-99% identity thereof.


In one embodiment, the CAR molecule further comprises a sequence encoding a costimulatory domain, e.g., a costimulatory domain described herein. In one embodiment, the costimulatory domain comprises a functional signaling domain of a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). In one embodiment, the costimulatory domain comprises a sequence of SEQ ID NO: 16. In one embodiment, the costimulatory domain comprises a sequence of SEQ ID NO:51. In one embodiment, the costimulatory domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO: 16 or SEQ ID NO:51, or a sequence with 95-99% identity to an amino acid sequence of SEQ ID NO: 16 or SEQ ID NO:51. In one embodiment, the costimulatory domain comprises a functional signaling domain of a protein selected from the group consisting of MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, 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, NKG2D, NKG2C, 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, CD19a, and a ligand that specifically binds with CD83. In embodiments, the costimulatory domain comprises 4-1BB, CD27, CD28, or ICOS.


In one embodiment, the CAR molecule further comprises a sequence encoding an intracellular signaling domain, e.g., an intracellular signaling domain described herein. In one embodiment, the intracellular signaling domain comprises a functional signaling domain of 4-1BB and/or a functional signaling domain of CD3 zeta. In one embodiment, the intracellular signaling domain comprises the sequence of SEQ ID NO: 16 and/or the sequence of SEQ ID NO:17. In one embodiment, the intracellular signaling domain comprises the sequence of SEQ ID NO:16 and/or the sequence of SEQ ID NO:43. In one embodiment, the intracellular signaling domain comprises a functional signaling domain of CD27 and/or a functional signaling domain of CD3 zeta. In one embodiment, the intracellular signaling domain comprises the sequence of SEQ ID NO: 51 and/or the sequence of SEQ ID NO:17. In one embodiment, the intracellular signaling domain comprises the sequence of SEQ ID NO:51 and/or the sequence of SEQ ID NO:43. In one embodiment, the intracellular signaling domain comprises an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 20, 10 or 5 modifications (e.g., substitutions) of an amino acid sequence of SEQ ID NO:16 or SEQ ID NO:51 and/or an amino acid sequence of SEQ ID NO:17 or SEQ ID NO:43, or a sequence with 95-99% identity to an amino acid sequence of SEQ ID NO:16 or SEQ ID NO:51 and/or an amino acid sequence of SEQ ID NO:17 or SEQ ID NO:43. In one embodiment, the intracellular signaling domain comprises the sequence of SEQ ID NO:16 or SEQ ID NO:51 and the sequence of SEQ ID NO: 17 or SEQ ID NO:43, wherein the sequences comprising the intracellular signaling domain are expressed in the same frame and as a single polypeptide chain.


In one embodiment, the CAR molecule further comprises a leader sequence, e.g., a leader sequence described herein. In one embodiment, the leader sequence comprises an amino acid sequence of SEQ ID NO: 13, or a sequence with 95-99% identity to an amino acid sequence of SEQ ID NO:13.


In one aspect, the CAR (e.g., a CD19 CAR, or a CD22 CAR) comprises an optional leader sequence (e.g., an optional leader sequence described herein), an extracellular antigen binding domain, a hinge (e.g., hinge described herein), a transmembrane domain (e.g., transmembrane domain described herein), and an intracellular stimulatory domain (e.g., intracellular stimulatory domain described herein). In one aspect an exemplary CAR construct comprises an optional leader sequence (e.g., a leader sequence described herein), an extracellular antigen binding domain, a hinge, a transmembrane domain, an intracellular costimulatory domain (e.g., an intracellular costimulatory domain described herein) and an intracellular stimulatory domain.


It was found that CAR molecules comprising a short or no linker between the variable domains (e.g., VH and VL) of the antigen binding domain showed equal to, or greater, activity than longer versions of the linker. For example, in some embodiments, CD22-65s (having (Gly4-Ser)n linker, wherein n is 1 (SEQ ID NO: 18)) shows comparable or greater activity and/or efficacy in a tumor model, compared to CD22-65 (having (Gly4-Ser)n linker, wherein n is 3 (SEQ ID NO: 107)).


Accordingly, any of the antigen binding domains or CAR molecules described herein, e.g., CD19 CAR molecule or a CD22 CAR molecule, can have a linker connecting the variable domains of the antigen binding domain of varying lengths, including for example, a short linker of about 3 to 6 amino acids, 4 to 5 amino acids, or about 5 amino acids. In some embodiments, a longer linker can be used, e.g., about 6 to 35 amino acids, e.g., 8 to 32 amino acids, 10 to 30 amino acids, 10 to 20 amino acids. For example, a (Gly4-Ser)n linker, wherein n is 0, 1, 2, 3, 4, 5, or 6 (SEQ ID NO: 53) can be used. In one embodiment, the variable domains are not connected via a linker, e.g., (Gly4-Ser)n linker, n=0 (“Gly4-Ser” disclosed as SEQ ID NO: 18). In some embodiments, the variable domains are connected via a short linker, e.g., (Gly4-Ser)n linker, n=1 (SEQ ID NO: 18). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=2 (SEQ ID NO: 49). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=3 (SEQ ID NO: 107). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=4 (SEQ ID NO: 106). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=5 (SEQ ID NO: 1341). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=6 (SEQ ID NO: 1342). The order of the variable domain, e.g., in which the VL and VH domains appear in the antigen binding domain, e.g., scFv, can be varied (i.e., VL-VH, or VH-VL orientation). In another embodiment, the antigen binding domain binds to CD22, e.g., a CD22 antigen binding domain as described herein. In another embodiment, the antigen binding domain binds to CD19, e.g., a CD19 antigen binding domain as described herein.


The invention also pertains to nucleic acid molecules, vectors, cells and uses comprising any of the foregoing aspects or embodiments.


Bispecific Antibodies

A bispecific antibody molecule (which can be, e.g., administered alone or as a portion of a CAR) can comprise two VH regions and two VL regions. In some embodiments, the upstream antibody or portion thereof (e.g. scFv) is arranged with its VH (VH1) upstream of its VL (VL1) and the downstream antibody or portion thereof (e.g. scFv) is arranged with its VL (VL2) upstream of its VH (VH2), such that the overall bispecific antibody molecule has the arrangement VH1-VL1-VL2-VH2. In other embodiments, the upstream antibody or portion thereof (e.g. scFv) is arranged with its VL (VL1) upstream of its VH (VH1) and the downstream antibody or portion thereof (e.g. scFv) is arranged with its VH (VH2) upstream of its VL (VL2), such that the overall bispecific antibody molecule has the arrangement VL1-VH1-VH2-VL2.


mTOR Inhibitors


In one embodiment, the cells expressing a CAR molecule, e.g., a CD19 CAR molecule, or a CD22 CAR molecule e.g., a CAR molecule described herein, are co-administered with a low, immune enhancing dose of an mTOR inhibitor. While not wishing to be bound by theory, it is believed that treatment with a low, immune enhancing, dose (e.g., a dose that is insufficient to completely suppress the immune system but sufficient to improve immune function) is accompanied by a decrease in PD-1 positive T cells or an increase in PD-1 negative cells. PD-1 positive T cells, but not PD-1 negative T cells, can be exhausted by engagement with cells which express a PD-1 ligand, e.g., PD-L1 or PD-L2.


In an embodiment this approach can be used to optimize the performance of CAR cells described herein in the subject. While not wishing to be bound by theory, it is believed that, in an embodiment, the performance of endogenous, non-modified immune effector cells, e.g., T cells, is improved. While not wishing to be bound by theory, it is believed that, in an embodiment, the performance of a CAR expressing cell is improved. In other embodiments, cells, e.g., T cells, which have, or will be engineered to express a CAR, can be treated ex vivo by contact with an amount of an mTOR inhibitor that increases the number of PD1 negative immune effector cells, e.g., T cells or increases the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells.


In an embodiment, administration of a low, immune enhancing, dose of an mTOR inhibitor, e.g., an allosteric inhibitor, e.g., RAD001, or a catalytic inhibitor, is initiated prior to administration of an CAR expressing cell described herein, e.g., T cells. In an embodiment, the CAR cells are administered after a sufficient time, or sufficient dosing, of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, has been, at least transiently, increased.


In an embodiment, the cell, e.g., T cell, to be engineered to express a CAR, is harvested after a sufficient time, or after sufficient dosing of the low, immune enhancing, dose of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, in the subject or harvested from the subject has been, at least transiently, increased.


Additional features or embodiments of the compositions or methods described herein include one or more of the following:


In embodiments, the one or more cells that express a CAR molecule that binds CD19 are administered concurrently with, before, or after the cells that express a CAR molecule that binds CD19.


In embodiments, the subject has or is identified as having a difference between a determined characteristic compared to a reference characteristic, in a characteristic of CD19, e.g., a mutation causing a frameshift or a premature stop codon or both, in a biological sample.


In embodiments, the subject has or is identified as having a difference, e.g., a statistically significant difference, between a determined level compared to a reference level of Treg cells in a biological sample.


In embodiments, the subject has or is identified as having an increase, e.g., a statistically significant increase, between a determined level and to a reference level of Treg cells in a biological sample.


In embodiments, the subject has relapsed or is identified as having relapsed after treatment with the one or more cells that express a CAR molecule that binds CD19, e.g., a CD19 CAR.


In embodiments, in accordance with a method described herein, e.g., a method of providing anti-tumor immunity to a mammal, or method of treating a mammal, a mammal is a non-responder, partial responder, or complete responder to a previously administered cancer therapy, e.g., a CD19 CAR therapy or a cancer therapy other than a CD19 CAR-expressing cell. In embodiments, the mammal is a non-relapser, partial relapse, or complete relapse to a previously administered cancer therapy, e.g., a CD19 CAR therapy or a cancer therapy other than a CD19 CAR-expressing cell. In embodiments, the mammal comprises a CD19-negative cancer cell or a CD19-positive cancer cell. In embodiments, the mammal further comprises a CD22-positive cancer cell. In embodiments, the mammal has a relapsed and/or refractory ALL cancer. In embodiments, the mammal was previously administered a CD19 CAR-expressing cell and is refractory to CD19 CAR treatment.


In an embodiment, the method further comprises administering a checkpoint inhibitor. In embodiments, the subject receives a pre-treatment of with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor, prior to the initiation of a CART therapy. In embodiments, the subject receives concurrent treatment with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor. In embodiments, the subject receives treatment with an agent, e.g., an mTOR inhibitor, and/or a checkpoint inhibitor, post-CART therapy.


In embodiments, the determined level or determined characteristic is acquired before, at the same time, or during a course of CART therapy.


In embodiments, the cell expresses an inhibitory molecule that comprises a first polypeptide that comprises at least a portion of an inhibitory molecule, associated with a second polypeptide that comprises a positive signal from an intracellular signaling domain. In embodiments, the inhibitory molecule comprise first polypeptide that comprises at least a portion of PD1 and a second polypeptide comprising a costimulatory domain and primary signaling domain.


In embodiments, the method comprises assaying a gene signature that indicates whether a subject treated with the cell is likely to relapse, or has relapsed. In embodiments, the method comprises assaying the gene signature in the cell prior to infusion into the subject. In embodiments, the method further comprises decreasing the TREG signature of a population of cells comprising the transduced cell. In embodiments, decreasing the TREG signature comprises performing CD25-depletion on the population of cells.


In embodiments, the subject is a mammal, e.g., a human, e.g., a pediatric subject, a young adult or an adult.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein (e.g., sequence database reference numbers) are incorporated by reference in their entirety. For example, all GenBank, Unigene, and Entrez sequences referred to herein, e.g., in any Table herein, are incorporated by reference. Unless otherwise specified, the sequence accession numbers specified herein, including in any Table herein, refer to the database entries current as of Apr. 8, 2015. When one gene or protein references a plurality of sequence accession numbers, all of the sequence variants are encompassed.


In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.


Headings, sub-headings or numbered or lettered elements, e.g., (a), (b), (i) etc, are presented merely for ease of reading. The use of headings or numbered or lettered elements in this document does not require the steps or elements be performed in alphabetical order or that the steps or elements are necessarily discrete from one another.


Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-1B show the in vivo anti-leukemia activity of CART22 in a primary ALL model. FIG. 1A shows the experimental design. NSG mice were injected with one million luciferase-positive JH331-CBG primary ALL cells (CD19+CD22). On day 14 mice were randomized based on tumor burden to receive two million UTD cells, two million CART22 cells or two million CART19 cells. Bioluminescence imaging (BLI) measurements were obtained prior to engraftment, and on days 11, 18, 35 and 42. FIG. 1B shows a graph with the experimental data. Untreated (UTD) mice showed rapid progression while CART22 treated mice showed complete remission and long-term tumor control (similar to CART19).



FIG. 2 is a graph showing tumor burden measured by serial bioluminescence imaging. NSG mice were injected with one million Nalm6 leukemia cells followed one week later by administration of one million T cells expressing the various CAR constructs as indicated. Control mice were left untreated (UTD).



FIG. 3 depicts the study design as described in Example 2.





DETAILED DESCRIPTION

The disclosure provides, inter alia, a method of treating a hematological cancer, comprising administering cells that express a CAR molecule that binds CD22, e.g., a CD22 CAR as described herein, wherein the CD22 CAR is administered according to a fractionated dosing regimen, e.g., split-dosing regimen, e.g., as described herein. In some embodiments, the fractionated dosing regimen comprises a total dose administered in at least 1, 2, 3, 4 or more doses, e.g., partial doses. In some embodiments, the CD22 CAR-expressing cells are administered in combination with an additional therapy, e.g., a CD19 CAR-expressing cell therapy. In some embodiments, the CD22 CAR-expressing cells are administered before, after or concurrently with the CD19 CAR-expressing cells. In some embodiments, the hematological cancer is ALL, e.g., relapsed and/or refractory ALL. In some embodiments, the subject is a young adult or a pediatric subject, e.g., aged about 1-29 years. Also described herein are compositions comprising CD22-expressing cells and methods of manufacturing the same.


Definitions

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


The term “a” and “an” refers 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.


The term “about” when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ±20% or in some instances ±10%, or in some instances ±5%, or in some instances ±1%, or in some instances ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.


The term “dose fractionation” as used herein refers to a dose, e.g., a total dose, e.g., a clinical dose, that is administered in at least two partial doses or sub-doses. In an embodiment, the partial doses or sub-doses are equal. In an embodiment, the partial doses or sub-doses are fractions of the total dose. In an embodiment, the dose is fractionated, e.g., split, into three partial doses, e.g., a first partial dose, a second partial dose and a third partial dose. In an embodiment the first partial dose is 10% of the total dose. In an embodiment, the second partial dose is 30% of the total dose. In an embodiment, the third partial dose is 60% of the total dose. In an embodiment, the partial doses or sub-doses are administered over a period of time, e.g., over at least 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 2 days, 3 days, 4 days, 5 days, 6 days or 7 days, 2 weeks, 3 weeks, 4 weeks, 2 months, 3 months, 4 months, 6 months or 1 year. In an embodiment, a dose fractionation is a split-dose.


The term “apheresis” as used herein refers to the art-recognized extracorporeal process by which the blood of a donor or patient is removed from the donor or patient and passed through an apparatus that separates out selected particular constituent(s) and returns the remainder to the circulation of the donor or patient, e.g., by retransfusion. Thus, “an apheresis sample” refers to a sample obtained using apheresis.


The term “bioequivalent” refers to an amount of an agent other than the reference compound (e.g., RAD001), required to produce an effect equivalent to the effect produced by the reference dose or reference amount of the reference compound (e.g., RAD001). In an embodiment the effect is the level of mTOR inhibition, e.g., as measured by P70 S6 kinase inhibition, e.g., as evaluated in an in vivo or in vitro assay, e.g., as measured by an assay described herein, e.g., the Boulay assay, or measurement of phosphorylated S6 levels by western blot. In an embodiment, the effect is alteration of the ratio of PD-1 positive/PD-1 negative T cells, as measured by cell sorting. In an embodiment a bioequivalent amount or dose of an mTOR inhibitor is the amount or dose that achieves the same level of P70 S6 kinase inhibition as does the reference dose or reference amount of a reference compound. In an embodiment, a bioequivalent amount or dose of an mTOR inhibitor is the amount or dose that achieves the same level of alteration in the ratio of PD-1 positive/PD-1 negative T cells as does the reference dose or reference amount of a reference compound.


The term “inhibition” or “inhibitor” includes a reduction in a certain parameter, e.g., an activity, of a given molecule, e.g., CD19, or CD22. For example, inhibition of an activity, e.g., an activity of CD19, or CD22, of at least 5%, 10%, 20%, 30%, 40%, or more is included by this term. Thus, inhibition need not be 100%. Activities for the inhibitors can be determined as described herein or by assays known in the art. A “B-cell inhibitor” is a molecule, e.g., a small molecule, antibody, CAR or cell comprising a CAR, which causes the reduction in a certain parameter, e.g., an activity, e.g., growth or proliferation, of a B-cell, or which causes a reduction in a certain parameter, e.g., an activity, of a molecule associated with a B cell.


The term “Chimeric Antigen Receptor” or alternatively a “CAR” refers to a set of polypeptides, typically two in the simplest embodiments, which when in an immune effector cell, provides the cell with specificity for a target cell, typically a cancer cell, and with intracellular signal generation. In some embodiments, a CAR comprises at least an extracellular antigen binding domain, a transmembrane domain and a cytoplasmic signaling domain (also referred to herein as “an intracellular signaling domain”) comprising a functional signaling domain derived from a stimulatory molecule and/or costimulatory molecule as defined below. In some embodiments, the set of polypeptides are in the same polypeptide chain, e.g., comprise a chimeric fusion protein. In some embodiments, the set of polypeptides are not contiguous with each other, e.g., are in different polypeptide chains. In some embodiments, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signaling domain. In one aspect, the stimulatory molecule of the CAR is the zeta chain associated with the T cell receptor complex (e.g., CD3 zeta). In one aspect, the cytoplasmic signaling domain comprises a primary signaling domain (e.g., a primary signaling domain of CD3-zeta). In one aspect, the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one costimulatory molecule as defined below. In one aspect, the costimulatory molecule is chosen from the costimulatory molecules described herein, e.g., 4-1BB (i.e., CD137), CD27, and/or CD28. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a costimulatory molecule and a functional signaling domain derived from a stimulatory molecule. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In one aspect, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising at least two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In one aspect the CAR comprises an optional leader sequence at the amino-terminus (N-ter) of the CAR fusion protein. In one aspect, the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen binding domain, wherein the leader sequence is optionally cleaved from the antigen binding domain (e.g., a scFv) during cellular processing and localization of the CAR to the cellular membrane.


The phrase “disease associated with expression of CD22” as used herein includes but is not limited to, a disease associated with expression of CD22 (e.g., wild-type or mutant CD22) or condition associated with cells which express, or at any time expressed, CD22 (e.g., wild-type or mutant CD22) including, e.g., a proliferative disease such as a cancer or malignancy or a precancerous condition such as a myelodysplasia, a myelodysplastic syndrome or a preleukemia; or a noncancer related indication associated with cells which express CD22 (e.g., wild-type or mutant CD22). For the avoidance of doubt, a disease associated with expression of CD22 may include a condition associated with cells which do not presently express CD22, e.g., because CD22 expression has been downregulated, e.g., due to treatment with a molecule targeting CD22, e.g., a CD22 CAR, but which at one time expressed CD22. In one aspect, a cancer associated with expression of CD22 is a hematological cancer. In one aspect, a hematological cancer includes but is not limited to AML, myelodysplastic syndrome, ALL, hairy cell leukemia, Prolymphocytic leukemia, Chronic myeloid leukemia, Hodgkin lymphoma, Blastic plasmacytoid dendritic cell neoplasm, and the like. In an embodiment, a hematological cancer associated with expression of CD22 is ALL, e.g., relapsed or refractory ALL. Further disease associated with expression of CD22 expression include, but are not limited to, e.g., atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases associated with expression of CD22. Non-cancer related indications associated with expression of CD22 may also be included. In some embodiments, the CD22-expressing cells express, or at any time expressed, CD22 mRNA. In an embodiment, the CD22-expressing cells produce a CD22 protein (e.g., wild-type or mutant), and the CD22 protein may be present at normal levels or reduced levels. In an embodiment, the CD22-expressing cells produced detectable levels of a CD22 protein at one point, and subsequently produced substantially no detectable CD22 protein.


As used herein, unless otherwise specified, the terms “prevent,” “preventing” and “prevention” refer to an action that occurs before the subject begins to suffer from the condition, or relapse of the condition. Prevention need not result in a complete prevention of the condition; partial prevention or reduction of the condition or a symptom of the condition, or reduction of the risk of developing the condition, is encompassed by this term.


Administered “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject during the course of the subject's affliction with the disorder, e.g., the two or more treatments are delivered after the subject has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons. In some embodiments, the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referred to herein as “simultaneous” or “concurrent delivery”. In other embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration. For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive. The delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered. In one embodiment, the CAR-expressing cell is administered at a dose and/or dosing schedule described herein, and the B-cell inhibitor, or agent that enhances the activity of the CD19 CAR-expressing cell is administered at a dose and/or dosing schedule described herein.


“Derived from” as that term is used herein, indicates a relationship between a first and a second molecule. It generally refers to structural similarity between the first molecule and a second molecule and does not connote or include a process or source limitation on a first molecule that is derived from a second molecule. For example, in the case of an intracellular signaling domain that is derived from a CD3zeta molecule, the intracellular signaling domain retains sufficient CD3zeta structure such that is has the required function, namely, the ability to generate a signal under the appropriate conditions. It does not connote or include a limitation to a particular process of producing the intracellular signaling domain, e.g., it does not mean that, to provide the intracellular signaling domain, one must start with a CD3zeta sequence and delete unwanted sequence, or impose mutations, to arrive at the intracellular signaling domain.


The term “signaling domain” refers to the functional portion of a protein which acts by transmitting information within the cell to regulate cellular activity via defined signaling pathways by generating second messengers or functioning as effectors by responding to such messengers.


As used herein, the term “CD19” refers to the Cluster of Differentiation 19 protein, which is an antigenic determinant detectable on leukemia precursor cells. The human and murine amino acid and nucleic acid sequences can be found in a public database, such as GenBank, UniProt and Swiss-Prot. For example, the amino acid sequence of human CD19 can be found as UniProt/Swiss-Prot Accession No. P15391 and the nucleotide sequence encoding of the human CD19 can be found at Accession No. NM_001178098. As used herein, “CD19” includes proteins comprising mutations, e.g., point mutations, fragments, insertions, deletions and splice variants of full length wild-type CD19. CD19 is expressed on most B lineage cancers, including, e.g., acute lymphoblastic leukemia, chronic lymphocyte leukemia and non-Hodgkin lymphoma. Other cells with express CD19 are provided below in the definition of “disease associated with expression of CD19.” It is also an early marker of B cell progenitors. See, e.g., Nicholson et al. Mol. Immun. 34 (16-17): 1157-1165 (1997). In one aspect the antigen-binding portion of the CART recognizes and binds an antigen within the extracellular domain of the CD19 protein. In one aspect, the CD19 protein is expressed on a cancer cell.


The term “antibody,” as used herein, refers to a protein, or polypeptide sequence derived from an immunoglobulin molecule which specifically binds with an antigen. Antibodies can be polyclonal or monoclonal, multiple or single chain, or intact immunoglobulins, and may be derived from natural sources or from recombinant sources. Antibodies can be tetramers of immunoglobulin molecules.


The term “antibody fragment” refers to at least one portion of an antibody, that retains the ability to specifically interact with (e.g., by binding, steric hindrance, stabilizing/destabilizing, spatial distribution) an epitope of an antigen. Examples of antibody fragments include, but are not limited to, Fab, Fab′, F(ab′)2, Fv fragments, scFv antibody fragments, disulfide-linked Fvs (sdFv), a Fd fragment consisting of the VH and CH1 domains, linear antibodies, single domain antibodies such as sdAb (either VL or VH), camelid VHH domains, multi-specific antibodies formed from antibody fragments such as a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, and an isolated CDR or other epitope binding fragments of an antibody. An antigen binding fragment can also be incorporated into single domain antibodies, maxibodies, minibodies, nanobodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR and bis-scFv (see, e.g., Hollinger and Hudson, Nature Biotechnology 23:1126-1136, 2005). Antigen binding fragments can also be grafted into scaffolds based on polypeptides such as a fibronectin type III (Fn3)(see U.S. Pat. No. 6,703,199, which describes fibronectin polypeptide minibodies).


The term “scFv” refers to a fusion protein comprising at least one antibody fragment comprising a variable region of a light chain and at least one antibody fragment comprising a variable region of a heavy chain, wherein the light and heavy chain variable regions are contiguously linked, e.g., via a synthetic linker, e.g., a short flexible polypeptide linker, and capable of being expressed as a single chain polypeptide, and wherein the scFv retains the specificity of the intact antibody from which it is derived. Unless specified, as used herein an scFv may have the VL and VH variable regions in either order, e.g., with respect to the N-terminal and C-terminal ends of the polypeptide, the scFv may comprise VL-linker-VH or may comprise VH-linker-VL.


The term “complementarity determining region” or “CDR,” as used herein, refers to the sequences of amino acids within antibody variable regions which confer antigen specificity and binding affinity. For example, in general, there are three CDRs in each heavy chain variable region (e.g., HCDR1, HCDR2, and HCDR3) and three CDRs in each light chain variable region (LCDR1, LCDR2, and LCDR3). The precise amino acid sequence boundaries of a given CDR can be determined using any of a number of well-known schemes, including those described by Kabat et al. (1991), “Sequences of Proteins of Immunological Interest,” 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (“Kabat” numbering scheme), Al-Lazikani et al., (1997) JMB 273,927-948 (“Chothia” numbering scheme), or a combination thereof. Under the Kabat numbering scheme, in some embodiments, the CDR amino acid residues in the heavy chain variable domain (VH) are numbered 31-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the light chain variable domain (VL) are numbered 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3). Under the Chothia numbering scheme, in some embodiments, the CDR amino acids in the VH are numbered 26-32 (HCDR1), 52-56 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the VL are numbered 26-32 (LCDR1), 50-52 (LCDR2), and 91-96 (LCDR3). In a combined Kabat and Chothia numbering scheme, in some embodiments, the CDRs correspond to the amino acid residues that are part of a Kabat CDR, a Chothia CDR, or both. For instance, in some embodiments, the CDRs correspond to amino acid residues 26-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3) in a VH, e.g., a mammalian VH, e.g., a human VH; and amino acid residues 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3) in a VL, e.g., a mammalian VL, e.g., a human VL.


As used herein, the term “binding domain” or “antibody molecule” refers to a protein, e.g., an immunoglobulin chain or fragment thereof, comprising at least one immunoglobulin variable domain sequence. The term “binding domain” or “antibody molecule” encompasses antibodies and antibody fragments. In an embodiment, an antibody molecule is a multispecific antibody molecule, e.g., it comprises a plurality of immunoglobulin variable domain sequences, wherein a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope and a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope. In an embodiment, a multispecific antibody molecule is a bispecific antibody molecule. A bispecific antibody has specificity for no more than two antigens. A bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope.


The portion of the CAR of the invention comprising an antibody or antibody fragment thereof may exist in a variety of forms where the antigen binding domain is expressed as part of a contiguous polypeptide chain including, for example, a single domain antibody fragment (sdAb), a single chain antibody (scFv), a humanized antibody, or bispecific antibody (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). In one aspect, the antigen binding domain of a CAR composition of the invention comprises an antibody fragment. In a further aspect, the CAR comprises an antibody fragment that comprises a scFv.


The term “antibody heavy chain,” refers to the larger of the two types of polypeptide chains present in antibody molecules in their naturally occurring conformations, and which normally determines the class to which the antibody belongs.


The term “antibody light chain,” refers to the smaller of the two types of polypeptide chains present in antibody molecules in their naturally occurring conformations. Kappa (κ) and lambda (λ) light chains refer to the two major antibody light chain isotypes.


The term “recombinant antibody” refers to an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage or yeast expression system. The term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using recombinant DNA or amino acid sequence technology which is available and well known in the art.


The term “antigen” or “Ag” refers to a molecule that provokes an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both. The skilled artisan will understand that any macromolecule, including virtually all proteins or peptides, can serve as an antigen. Furthermore, antigens can be derived from recombinant or genomic DNA. A skilled artisan will understand that any DNA, which comprises a nucleotide sequences or a partial nucleotide sequence encoding a protein that elicits an immune response therefore encodes an “antigen” as that term is used herein. Furthermore, one skilled in the art will understand that an antigen need not be encoded solely by a full length nucleotide sequence of a gene. It is readily apparent that the present invention includes, but is not limited to, the use of partial nucleotide sequences of more than one gene and that these nucleotide sequences are arranged in various combinations to encode polypeptides that elicit the desired immune response. Moreover, a skilled artisan will understand that an antigen need not be encoded by a “gene” at all. It is readily apparent that an antigen can be generated synthesized or can be derived from a biological sample, or might be macromolecule besides a polypeptide. Such a biological sample can include, but is not limited to a tissue sample, a tumor sample, a cell or a fluid with other biological components.


The terms “compete” or “cross-compete” are used interchangeably herein to refer to the ability of an antibody molecule to interfere with binding of an antibody molecule, e.g., an anti-CD19 or CD22 antibody molecule provided herein, to a target, e.g., human CD19 or CD22. The interference with binding can be direct or indirect (e.g., through an allosteric modulation of the antibody molecule or the target). The extent to which an antibody molecule is able to interfere with the binding of another antibody molecule to the target, and therefore whether it can be said to compete, can be determined using a competition binding assay, e.g., as described herein. In some embodiments, a competition binding assay is a quantitative competition assay. In some embodiments, a first antibody molecule is said to compete for binding to the target with a second antibody molecule when the binding of the first antibody molecule to the target is reduced by 10% or more, e.g., 20% or more, 30% or more, 40% or more, 50% or more, 55% or more, 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more, 98% or more, 99% or more in a competition binding assay (e.g., a competition assay described herein).


As used herein, the term “epitope” refers to the moieties of an antigen (e.g., human CD19 or CD22) that specifically interact with an antibody molecule. Such moieties, referred to herein as epitopic determinants, typically comprise, or are part of, elements such as amino acid side chains or sugar side chains. An epitopic determinate can be defined, e.g., by methods known in the art or disclosed herein, e.g., by crystallography or by hydrogen-deuterium exchange. At least one or some of the moieties on the antibody molecule, that specifically interact with an epitopic determinant, are typically located in a CDR(s). Typically an epitope has a specific three dimensional structural characteristics. Typically an epitope has specific charge characteristics. Some epitopes are linear epitopes while others are conformational epitopes.


The term “anti-cancer effect” refers to a biological effect which can be manifested by various means, including but not limited to, e.g., a decrease in tumor volume, a decrease in the number of cancer cells, a decrease in the number of metastases, an increase in life expectancy, decrease in cancer cell proliferation, decrease in cancer cell survival, or amelioration of various physiological symptoms associated with the cancerous condition. An “anti-cancer effect” can also be manifested by the ability of the peptides, polynucleotides, cells and antibodies described herein in prevention of the occurrence of cancer in the first place. The term “anti-tumor effect” refers to a biological effect which can be manifested by various means, including but not limited to, e.g., a decrease in tumor volume, a decrease in the number of tumor cells, a decrease in tumor cell proliferation, or a decrease in tumor cell survival.


The term “autologous” refers to any material derived from the same individual to whom it is later to be re-introduced into the individual.


The term “allogeneic” refers to any material derived from a different animal of the same species as the individual to whom the material is introduced. Two or more individuals are said to be allogeneic to one another when the genes at one or more loci are not identical. In some aspects, allogeneic material from individuals of the same species may be sufficiently unlike genetically to interact antigenically


The term “xenogeneic” refers to a graft derived from an animal of a different species.


The term “cancer” refers to a disease characterized by the 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 are described herein and include but are not limited to, 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. The terms “tumor” and “cancer” are used interchangeably herein, e.g., both terms encompass solid and liquid, e.g., diffuse or circulating, tumors. As used herein, the term “cancer” or “tumor” includes premalignant, as well as malignant cancers and tumors.


The terms “cancer associated antigen” or “tumor antigen” or “proliferative disorder antigen” or “antigen associated with a proliferative disorder” interchangeably refers to a molecule (typically protein, carbohydrate or lipid) that is preferentially expressed on the surface of a cancer cell, either entirely or as a fragment (e.g., MHC/peptide), in comparison to a normal cell, and which is useful for the preferential targeting of a pharmacological agent to the cancer cell. In some embodiments, a tumor antigen is a marker expressed by both normal cells and cancer cells, e.g., a lineage marker, e.g., CD19 or CD22 on B cells. In certain aspects, the tumor antigens of the present invention are derived from, cancers including but not limited to primary or metastatic melanoma, thymoma, lymphoma, sarcoma, lung cancer, liver cancer, non-Hodgkin lymphoma, Hodgkin lymphoma, leukemias, uterine cancer, cervical cancer, bladder cancer, kidney cancer and adenocarcinomas such as breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, and the like. In some embodiments, the tumor antigen is an antigen that is common to a specific proliferative disorder. In some embodiments, a cancer-associated antigen is a cell surface molecule that is overexpressed in a cancer cell in comparison to a normal cell, for instance, 1-fold over expression, 2-fold overexpression, 3-fold overexpression or more in comparison to a normal cell. In some embodiments, a cancer-associated antigen is a cell surface molecule that is inappropriately synthesized in the cancer cell, for instance, a molecule that contains deletions, additions or mutations in comparison to the molecule expressed on a normal cell. In some embodiments, a cancer-associated antigen will be expressed exclusively on the cell surface of a cancer cell, entirely or as a fragment (e.g., MHC/peptide), and not synthesized or expressed on the surface of a normal cell. In some embodiments, the CARs of the present invention includes CARs comprising an antigen binding domain (e.g., antibody or antibody fragment) that binds to a MHC presented peptide. Normally, peptides derived from endogenous proteins fill the pockets of Major histocompatibility complex (MHC) class I molecules, and are recognized by T cell receptors (TCRs) on CD8+ T lymphocytes. The MHC class I complexes are constitutively expressed by all nucleated cells. In cancer, virus-specific and/or tumor-specific peptide/MHC complexes represent a unique class of cell surface targets for immunotherapy. TCR-like antibodies targeting peptides derived from viral or tumor antigens in the context of human leukocyte antigen (HLA)-A1 or HLA-A2 have been described (see, e.g., Sastry et al., J Virol. 2011 85(5):1935-1942; Sergeeva et al., Bood, 2011 117(16):4262-4272; Verma et al., J Immunol 2010 184(4):2156-2165; Willemsen et al., Gene Ther 2001 8(21):1601-1608; Dao et al., Sci Transl Med 2013 5(176):176ra33; Tassev et al., Cancer Gene Ther 2012 19(2):84-100). For example, TCR-like antibody can be identified from screening a library, such as a human scFv phage displayed library.


The phrase “disease associated with expression of CD19” includes, but is not limited to, a disease associated with expression of CD19 (e.g., wild-type or mutant CD19) or condition associated with cells which express, or at any time expressed, CD19 (e.g., wild-type or mutant CD19) including, e.g., proliferative diseases such as a cancer or malignancy or a precancerous condition such as a myelodysplasia, a myelodysplastic syndrome or a preleukemia; or a noncancer related indication associated with cells which express CD19. For the avoidance of doubt, a disease associated with expression of CD19 may include a condition associated with cells which do not presently express CD19, e.g., because CD19 expression has been downregulated, e.g., due to treatment with a molecule targeting CD19, e.g., a CD19 CAR, but which at one time expressed CD19. In one aspect, a cancer associated with expression of CD19 is a hematological cancer. In one aspect, the hematological cancer is a leukemia or a lymphoma. In one aspect, a cancer associated with expression of CD19 includes cancers and malignancies including, but not limited to, e.g., one or more acute leukemias including but not limited to, e.g., B-cell acute Lymphoid Leukemia (BALL), T-cell acute Lymphoid Leukemia (TALL), acute lymphoid leukemia (ALL); one or more chronic leukemias including but not limited to, e.g., chronic myelogenous leukemia (CML), Chronic Lymphoid Leukemia (CLL). Additional cancers or hematologic conditions associated with expression of CD19 comprise, but are not limited to, e.g., B cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma, Follicular lymphoma, Hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), Marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells, and the like. Further diseases associated with expression of CD19 expression include, but not limited to, e.g., atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases associated with expression of CD19. Non-cancer related indications associated with expression of CD19 include, but are not limited to, e.g., autoimmune disease, (e.g., lupus), inflammatory disorders (allergy and asthma) and transplantation. In some embodiments, the CD19-expressing cells express, or at any time expressed, CD19 mRNA. In an embodiment, the CD19-expressing cells produce a CD19 protein (e.g., wild-type or mutant), and the CD19 protein may be present at normal levels or reduced levels. In an embodiment, the CD19-expressing cells produced detectable levels of a CD19 protein at one point, and subsequently produced substantially no detectable CD19 protein.


The term “conservative sequence modifications” refers to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody or antibody fragment containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody or antibody fragment of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within a CAR of the invention can be replaced with other amino acid residues from the same side chain family and the altered CAR can be tested using the functional assays described herein.


The term “stimulation,” refers to a primary response induced by binding of a stimulatory molecule (e.g., a TCR/CD3 complex or CAR) with its cognate ligand (or tumor antigen in the case of a CAR) thereby mediating a signal transduction event, such as, but not limited to, signal transduction via the TCR/CD3 complex or signal transduction via the appropriate NK receptor or signaling domains of the CAR. Stimulation can mediate altered expression of certain molecules.


The term “stimulatory molecule,” refers to a molecule expressed by an immune cell, e.g., T cell, NK cell, or B cell) that provides the cytoplasmic signaling sequence(s) that regulate activation of the immune cell in a stimulatory way for at least some aspect of the immune cell signaling pathway. In one aspect, the signal is a primary signal that is initiated by, for instance, binding of a TCR/CD3 complex with an MHC molecule loaded with peptide, and which leads to mediation of a T cell response, including, but not limited to, proliferation, activation, differentiation, and the like. A primary cytoplasmic signaling sequence (also referred to as a “primary signaling domain”) that acts in a stimulatory manner may contain a signaling motif which is known as immunoreceptor tyrosine-based activation motif or ITAM. Examples of an ITAM containing cytoplasmic signaling sequence that is of particular use in the invention includes, but is not limited to, those derived from CD3 zeta, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon R1b), CD3 gamma, CD3 delta, CD3 epsilon, CD79a, CD79b, DAP10, and DAP12. In a specific CAR of the invention, the intracellular signaling domain in any one or more CARS of the invention comprises an intracellular signaling sequence, e.g., a primary signaling sequence of CD3-zeta. In a specific CAR of the invention, the primary signaling sequence of CD3-zeta is the sequence provided as SEQ ID NO:17, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In a specific CAR of the invention, the primary signaling sequence of CD3-zeta is the sequence as provided in SEQ ID NO:43, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.


The term “antigen presenting cell” or “APC” refers to an immune system cell such as an accessory cell (e.g., a B-cell, a dendritic cell, and the like) that displays a foreign antigen complexed with major histocompatibility complexes (MHC's) on its surface. T-cells may recognize these complexes using their T-cell receptors (TCRs). APCs process antigens and present them to T-cells.


“Immune effector cell,” as that term is used herein, refers to a cell that is involved in an immune response, e.g., in the promotion of an immune effector response. Examples of immune effector cells include T cells, e.g., alpha/beta T cells and gamma/delta T cells, B cells, natural killer (NK) cells, natural killer T (NK-T) cells, mast cells, and myeloid-derived phagocytes.


“Immune effector function or immune effector response,” as that term is used herein, refers to function or response, e.g., of an immune effector cell, that enhances or promotes an immune attack of a target cell. E.g., an immune effector function or response refers a property of a T or NK cell that promotes killing or the inhibition of growth or proliferation, of a target cell. In the case of a T cell, primary stimulation and co-stimulation are examples of immune effector function or response.


The term “effector function” refers to a specialized function of a cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines.


An “intracellular signaling domain,” as the term is used herein, refers to an intracellular portion of a molecule. The intracellular signaling domain can generate a signal that promotes an immune effector function of the CAR containing cell, e.g., a CART cell. Examples of immune effector function, e.g., in a CART cell, include cytolytic activity and helper activity, including the secretion of cytokines. In embodiments, the intracellular signal domain is the portion of the protein which transduces the effector function signal and directs the cell to perform a specialized function. While the entire intracellular signaling domain can be employed, in many cases it is not necessary to use the entire chain. To the extent that a truncated portion of the intracellular signaling domain is used, such truncated portion may be used in place of the intact chain as long as it transduces the effector function signal. The term intracellular signaling domain is thus meant to include any truncated portion of the intracellular signaling domain sufficient to transduce the effector function signal.


In an embodiment, the intracellular signaling domain can comprise a primary intracellular signaling domain. Exemplary primary intracellular signaling domains include those derived from the molecules responsible for primary stimulation, or antigen dependent simulation. In an embodiment, the intracellular signaling domain can comprise a costimulatory intracellular domain. Exemplary costimulatory intracellular signaling domains include those derived from molecules responsible for costimulatory signals, or antigen independent stimulation. For example, in the case of a CART, a primary intracellular signaling domain can comprise a cytoplasmic sequence of a T cell receptor, and a costimulatory intracellular signaling domain can comprise cytoplasmic sequence from co-receptor or costimulatory molecule.


A primary intracellular signaling domain can comprise a signaling motif which is known as an immunoreceptor tyrosine-based activation motif or ITAM. Examples of ITAM containing primary cytoplasmic signaling sequences include, but are not limited to, those derived from CD3 zeta, FcR gamma, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon R1b), CD3 gamma, CD3 delta, CD3 epsilon, CD22, CD79a, CD79b, CD278 (“ICOS”), FcεRI, CD66d, CD32, DAP10 and DAP12.


The term “zeta” or alternatively “zeta chain”, “CD3-zeta” or “TCR-zeta” is defined as the protein provided as GenBank Acc. No. BAG36664.1, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like, and a “zeta stimulatory domain” or alternatively a “CD3-zeta stimulatory domain” or a “TCR-zeta stimulatory domain” is defined as the amino acid residues from the cytoplasmic domain of the zeta chain, or functional derivatives thereof, that are sufficient to functionally transmit an initial signal necessary for T cell activation. In one aspect the cytoplasmic domain of zeta comprises residues 52 through 164 of GenBank Acc. No. BAG36664.1 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like, that are functional orthologs thereof. In one aspect, the “zeta stimulatory domain” or a “CD3-zeta stimulatory domain” is the sequence provided as SEQ ID NO:17. In one aspect, the “zeta stimulatory domain” or a “CD3-zeta stimulatory domain” is the sequence provided as SEQ ID NO:43.


The term “costimulatory molecule” refers to the cognate binding partner on a T cell that specifically binds with a costimulatory ligand, thereby mediating a costimulatory response by the T cell, such as, but not limited to, proliferation. Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that contribute to an efficient immune response. Costimulatory molecules include, but are not limited to an MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signalling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, 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, NKG2D, NKG2C, 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, CD19a, and a ligand that specifically binds with CD83.


A costimulatory intracellular signaling domain refers to the intracellular portion of a costimulatory molecule. The intracellular signaling domain can comprise the entire intracellular portion, or the entire native intracellular signaling domain, of the molecule from which it is derived, or a functional fragment or derivative thereof.


The term “4-1BB” refers to a member of the TNFR superfamily with an amino acid sequence provided as GenBank Acc. No. AAA62478.2, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like; and a “4-1BB costimulatory domain” is defined as amino acid residues 214-255 of GenBank Acc. No. AAA62478.2, or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like. In one aspect, the “4-1BB costimulatory domain” is the sequence provided as SEQ ID NO:16 or the equivalent residues from a non-human species, e.g., mouse, rodent, monkey, ape and the like.


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 templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (e.g., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene, cDNA, or RNA, 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 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.


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 a 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 “effective amount” or “therapeutically effective amount” are used interchangeably herein, and refer to an amount of a compound, formulation, material, or composition, as described herein effective to achieve a particular biological result.


The term “endogenous” refers to any material from or produced inside an organism, cell, tissue or system.


The term “exogenous” refers to any material introduced from or produced outside an organism, cell, tissue or system.


The term “expression” refers to the transcription and/or translation of a particular nucleotide sequence driven by a promoter.


The term “transfer vector” refers to a composition of matter which 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, but not limited to, linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses. Thus, the term “transfer vector” includes an autonomously replicating plasmid or a virus. The term should also be construed to further include non-plasmid and non-viral compounds which facilitate transfer of nucleic acid into cells, such as, for example, a polylysine compound, liposome, and the like. Examples of viral transfer vectors include, but are not limited to, adenoviral vectors, adeno-associated virus vectors, retroviral vectors, lentiviral vectors, and the like.


The term “expression vector” refers to a vector comprising a recombinant polynucleotide comprising expression control sequences operatively linked to a nucleotide sequence to be expressed. An expression vector comprises 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, including 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.


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. HIV, SIV, and FIV are all examples of lentiviruses.


The term “lentiviral vector” refers to a vector derived from at least a portion of a lentivirus genome, including especially a self-inactivating lentiviral vector as provided in Milone et al., Mol. Ther. 17(8): 1453-1464 (2009). Other examples of lentivirus vectors that may be used in the clinic, include but are not limited to, e.g., the LENTIVECTOR® gene delivery technology from Oxford BioMedica, the LENTIMAX™ vector system from Lentigen and the like. Nonclinical types of lentiviral vectors are also available and would be known to one skilled in the art.


The term “homologous” or “identity” refers to the subunit sequence identity between two polymeric molecules, e.g., between two nucleic acid molecules, such as, two DNA molecules or two RNA molecules, or between two polypeptide molecules. When a subunit position in both of the two molecules is occupied by the same monomeric subunit; e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous or identical at that position. The homology between two sequences is a direct function of the number of matching or homologous positions; e.g., if half (e.g., five positions in a polymer ten subunits in length) of the positions in two sequences are homologous, the two sequences are 50% homologous; if 90% of the positions (e.g., 9 of 10), are matched or homologous, the two sequences are 90% homologous.


“Humanized” forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′)2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies and antibody fragments thereof are human immunoglobulins (recipient antibody or antibody fragment) in which residues from a complementarity-determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, a humanized antibody/antibody fragment can comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications can further refine and optimize antibody or antibody fragment performance. In general, the humanized antibody or antibody fragment thereof will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or a significant portion of the FR regions are those of a human immunoglobulin sequence. The humanized antibody or antibody fragment can also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al., Nature, 321: 522-525, 1986; Reichmann et al., Nature, 332: 323-329, 1988; Presta, Curr. Op. Struct. Biol., 2: 593-596, 1992.


“Fully human” refers to an immunoglobulin, such as an antibody or antibody fragment, where the whole molecule is of human origin or consists of an amino acid sequence identical to a human form of the antibody or immunoglobulin.


The term “isolated” means altered or removed from the natural state. For example, a nucleic acid or a peptide naturally present in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is “isolated.” An isolated nucleic acid or protein can exist in substantially purified form, or can exist in a non-native environment such as, for example, a host cell.


In the context of the present invention, 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.


The term “operably linked” or “transcriptional control” refers to functional linkage between a regulatory sequence and a heterologous nucleic acid sequence resulting in expression of the latter. For example, a first nucleic acid sequence is operably linked with a second nucleic acid sequence when the first nucleic acid sequence is placed in a functional relationship with the second nucleic acid sequence. For instance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Operably linked DNA sequences can be contiguous with each other and, e.g., where necessary to join two protein coding regions, are in the same reading frame.


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


The term “nucleic acid” or “polynucleotide” refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single- or double-stranded form. The term “nucleic acid” includes a gene, cDNA, or an mRNA. In one embodiment, the nucleic acid molecule is synthetic (e.g., chemically synthesized) or recombinant. Unless specifically limited, the term encompasses nucleic acids containing analogues or derivatives of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementarity sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)).


The terms “peptide,” “polypeptide,” and “protein” are used interchangeably, and refer to a compound comprised of amino acid residues covalently linked by peptide bonds. A protein or peptide must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a protein's or peptide's sequence. Polypeptides include any peptide or protein comprising two or more amino acids joined to each other by peptide bonds. As used herein, the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types. “Polypeptides” include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion proteins, among others. A polypeptide includes a natural peptide, a recombinant peptide, or a combination thereof.


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 “promoter/regulatory sequence” refers to a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulatory sequence. In some instances, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product. The promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.


The term “constitutive” promoter refers to a nucleotide sequence which, when operably linked with a polynucleotide which encodes or specifies a gene product, causes the gene product to be produced in a cell under most or all physiological conditions of the cell.


The term “inducible” promoter refers to a nucleotide sequence which, when operably linked with a polynucleotide which encodes or specifies a gene product, causes the gene product to be produced in a cell substantially only when an inducer which corresponds to the promoter is present in the cell.


The term “tissue-specific” promoter refers to a nucleotide sequence which, when operably linked with a polynucleotide encodes or specified by a gene, causes the gene product to be produced in a cell substantially only if the cell is a cell of the tissue type corresponding to the promoter.


The term “flexible polypeptide linker” or “linker” as used in the context of a scFv refers to a peptide linker that consists of amino acids such as glycine and/or serine residues used alone or in combination, to link variable heavy and variable light chain regions together. In one embodiment, the flexible polypeptide linker is a Gly/Ser linker and comprises the amino acid sequence (Gly-Gly-Gly-Ser)n, where n is a positive integer equal to or greater than 1. For example, n=1, n=2, n=3. n=4, n=5, n=6, n=7, n=8, n=9 and n=10 (SEQ ID NO:105). In one embodiment, the flexible polypeptide linkers include, but are not limited to, (Gly4 Ser)4 (SEQ ID NO:106) or (Gly4 Ser)3 (SEQ ID NO:107). In another embodiment, the linkers include multiple repeats of (Gly2Ser), (GlySer) or (Gly3Ser) (SEQ ID NO:108). Also included within the scope of the invention are linkers described in WO2012/138475, incorporated herein by reference.


As used herein, a 5′ cap (also termed an RNA cap, an RNA 7-methylguanosine cap or an RNA m7G cap) is a modified guanine nucleotide that has been added to the “front” or 5′ end of a eukaryotic messenger RNA shortly after the start of transcription. The 5′ cap consists of a terminal group which is linked to the first transcribed nucleotide. Its presence is important for recognition by the ribosome and protection from RNases. Cap addition is coupled to transcription, and occurs co-transcriptionally, such that each influences the other. Shortly after the start of transcription, the 5′ end of the mRNA being synthesized is bound by a cap-synthesizing complex associated with RNA polymerase. This enzymatic complex catalyzes the chemical reactions that are required for mRNA capping. Synthesis proceeds as a multi-step biochemical reaction. The capping moiety can be modified to modulate functionality of mRNA such as its stability or efficiency of translation.


As used herein, “in vitro transcribed RNA” refers to RNA, e.g., mRNA, that has been synthesized in vitro. Generally, the in vitro transcribed RNA is generated from an in vitro transcription vector. The in vitro transcription vector comprises a template that is used to generate the in vitro transcribed RNA.


As used herein, a “poly(A)” is a series of adenosines attached by polyadenylation to the mRNA. In some embodiments of a construct for transient expression, the polyA is between 50 and 5000 (SEQ ID NO: 28), e.g., greater than 64, e.g., greater than 100, e.g., than 300 or 400. Poly(A) sequences can be modified chemically or enzymatically to modulate mRNA functionality such as localization, stability or efficiency of translation.


As used herein, “polyadenylation” refers to the covalent linkage of a polyadenylyl moiety, or its modified variant, to a messenger RNA molecule. In eukaryotic organisms, most messenger RNA (mRNA) molecules are polyadenylated at the 3′ end. The 3′ poly(A) tail is a long sequence of adenine nucleotides (often several hundred) added to the pre-mRNA through the action of an enzyme, polyadenylate polymerase. In higher eukaryotes, the poly(A) tail is added onto transcripts that contain a specific sequence, the polyadenylation signal. The poly(A) tail and the protein bound to it aid in protecting mRNA from degradation by exonucleases. Polyadenylation is also important for transcription termination, export of the mRNA from the nucleus, and translation. Polyadenylation occurs in the nucleus immediately after transcription of DNA into RNA, but additionally can also occur later in the cytoplasm. After transcription has been terminated, the mRNA chain is cleaved through the action of an endonuclease complex associated with RNA polymerase. The cleavage site is usually characterized by the presence of the base sequence AAUAAA near the cleavage site. After the mRNA has been cleaved, adenosine residues are added to the free 3′ end at the cleavage site.


As used herein, “transient” refers to expression of a non-integrated transgene for a period of hours, days or weeks, wherein the period of time of expression is less than the period of time for expression of the gene if integrated into the genome or contained within a stable plasmid replicon in the host cell.


The term “signal transduction pathway” refers to the biochemical relationship between a variety of signal transduction molecules that play a role in the transmission of a signal from one portion of a cell to another portion of a cell. The phrase “cell surface receptor” includes molecules and complexes of molecules capable of receiving a signal and transmitting signal across the membrane of a cell.


The term “subject” is intended to include living organisms in which an immune response can be elicited (e.g., mammals, human).


The term, a “substantially purified” cell refers to a cell that is essentially free of other cell types. A substantially purified cell also refers to a cell which has been separated from other cell types with which it is normally associated in its naturally occurring 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 cell that have been separated from the cells with which they are naturally associated in their natural state. In some aspects, the cells are cultured in vitro. In other aspects, the cells are not cultured in vitro.


The term “therapeutic” as used herein means a treatment. A therapeutic effect is obtained by reduction, suppression, remission, or eradication of a disease state.


The term “prophylaxis” as used herein means the prevention of or protective treatment for a disease or disease state.


In the context of the present invention, “tumor antigen” or “hyperproliferative disorder antigen” or “antigen associated with a hyperproliferative disorder” refers to antigens that are common to specific hyperproliferative disorders. In certain aspects, the hyperproliferative disorder antigens of the present invention are derived from, cancers including but not limited to primary or metastatic melanoma, thymoma, lymphoma, sarcoma, lung cancer, liver cancer, non-Hodgkin lymphoma, Hodgkin lymphoma, leukemias, uterine cancer, cervical cancer, bladder cancer, kidney cancer and adenocarcinomas such as breast cancer, prostate cancer, ovarian cancer, pancreatic cancer, and the like.


The term “transfected” or “transformed” or “transduced” refers to a process by which 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 exogenous nucleic acid. The cell includes the primary subject cell and its progeny.


A subject “responds” to treatment if a parameter of a cancer (e.g., a hematological cancer, e.g., cancer cell growth, proliferation and/or survival) in the subject is retarded or reduced by a detectable amount, e.g., about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more as determined by any appropriate measure, e.g., by mass, cell count or volume. In one example, a subject responds to treatment if the subject experiences a life expectancy extended by about 5%, 10%, 20%, 30%, 40%, 50% or more beyond the life expectancy predicted if no treatment is administered. In another example, a subject responds to treatment, if the subject has an increased disease-free survival, overall survival or increased time to progression. Several methods can be used to determine if a patient responds to a treatment including, for example, criteria provided by NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). For example, in the context of B-ALL, a complete response or complete responder, may involve one or more of: <5% BM blast, >1000 neutrophil/ANC (/μL). >100,000 platelets (/μL) with no circulating blasts or extramedullary disease (no lymphadenopathy, splenomegaly, skin/gum infiltration/testicular mass/CNS involvement), Trilineage hematopoiesis, and no recurrence for 4 weeks. A partial responder may involve one or more of >50% reduction in BM blast, >1000 neutrophil/ANC (/μL). >100,000 platelets (/μL). A non-responder can show disease progression, e.g., >25% in BM blasts.


“Refractory” as used herein refers to a disease, e.g., cancer, that does not respond to a treatment. In embodiments, a refractory cancer can be resistant to a treatment before or at the beginning of the treatment. In other embodiments, the refractory cancer can become resistant during a treatment. A refractory cancer is also called a resistant cancer.


The term “relapse” as used herein refers to reappearance of a cancer after an initial period of responsiveness (e.g., complete response or partial response). The initial period of responsiveness may involve the level of cancer cells falling below a certain threshold, e.g., below 20%, 1%, 10%, 5%, 4%, 3%, 2%, or 1%. The reappearance may involve the level of cancer cells rising above a certain threshold, e.g., above 20%, 1%, 10%, 5%, 4%, 3%, 2%, or 1%. For example, e.g., in the context of B-ALL, the reappearance may involve, e.g., a reappearance of blasts in the blood, bone marrow (>5%), or any extramedullary site, after a complete response. A complete response, in this context, may involve <5% BM blast. More generally, in an embodiment, a response (e.g., complete response or partial response) can involve the absence of detectable MRD (minimal residual disease). In an embodiment, the initial period of responsiveness lasts at least 1, 2, 3, 4, 5, or 6 days; at least 1, 2, 3, or 4 weeks; at least 1, 2, 3, 4, 6, 8, 10, or 12 months; or at least 1, 2, 3, 4, or 5 years.


In some embodiments, a therapy that includes a CD19 inhibitor, e.g., a CD19 CAR therapy, may relapse or be refractory to treatment. The relapse or resistance can be caused by CD19 loss (e.g., an antigen loss mutation) or other CD19 alteration that reduces the level of CD19 (e.g., caused by clonal selection of CD19-negative clones). A cancer that harbors such CD19 loss or alteration is referred to herein as a “CD19-negative cancer” or a “CD19-negative relapsed cancer”). It shall be understood that a CD19-negative cancer need not have 100% loss of CD19, but a sufficient reduction to reduce the effectiveness of a CD19 therapy such that the cancer relapses or becomes refractory. In some embodiments, a CD19-negative cancer results from a CD19 CAR therapy.


The term “specifically binds,” refers to an antibody, or a ligand, which recognizes and binds with a binding partner (e.g., a stimulatory tumor antigen) protein present in a sample, but which antibody or ligand does not substantially recognize or bind other molecules in the sample.


As used herein, the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66:1-19.


“Regulatable chimeric antigen receptor (RCAR),” as that term is used herein, refers to a set of polypeptides, typically two in the simplest embodiments, which when in a RCARX cell, provides the RCARX cell with specificity for a target cell, typically a cancer cell, and with regulatable intracellular signal generation or proliferation, which can optimize an immune effector property of the RCARX cell. An RCARX cell relies at least in part, on an antigen binding domain to provide specificity to a target cell that comprises the antigen bound by the antigen binding domain. In an embodiment, an RCAR includes a dimerization switch that, upon the presence of a dimerization molecule, can couple an intracellular signaling domain to the antigen binding domain.


“Membrane anchor” or “membrane tethering domain”, as that term is used herein, refers to a polypeptide or moiety, e.g., a myristoyl group, sufficient to anchor an extracellular or intracellular domain to the plasma membrane.


“Switch domain,” as that term is used herein, e.g., when referring to an RCAR, refers to an entity, typically a polypeptide-based entity, that, in the presence of a dimerization molecule, associates with another switch domain. The association results in a functional coupling of a first entity linked to, e.g., fused to, a first switch domain, and a second entity linked to, e.g., fused to, a second switch domain. A first and second switch domain are collectively referred to as a dimerization switch. In embodiments, the first and second switch domains are the same as one another, e.g., they are polypeptides having the same primary amino acid sequence, and are referred to collectively as a homodimerization switch. In embodiments, the first and second switch domains are different from one another, e.g., they are polypeptides having different primary amino acid sequences, and are referred to collectively as a heterodimerization switch. In embodiments, the switch is intracellular. In embodiments, the switch is extracellular. In embodiments, the switch domain is a polypeptide-based entity, e.g., FKBP or FRB-based, and the dimerization molecule is small molecule, e.g., a rapalogue. In embodiments, the switch domain is a polypeptide-based entity, e.g., an scFv that binds a myc peptide, and the dimerization molecule is a polypeptide, a fragment thereof, or a multimer of a polypeptide, e.g., a myc ligand or multimers of a myc ligand that bind to one or more myc scFvs. In embodiments, the switch domain is a polypeptide-based entity, e.g., myc receptor, and the dimerization molecule is an antibody or fragments thereof, e.g., myc antibody.


“Dimerization molecule,” as that term is used herein, e.g., when referring to an RCAR, refers to a molecule that promotes the association of a first switch domain with a second switch domain. In embodiments, the dimerization molecule does not naturally occur in the subject, or does not occur in concentrations that would result in significant dimerization. In embodiments, the dimerization molecule is a small molecule, e.g., rapamycin or a rapalogue, e.g., RAD001.


The term “low, immune enhancing, dose” when used in conjunction with an mTOR inhibitor, e.g., an allosteric mTOR inhibitor, e.g., RAD001 or rapamycin, or a catalytic mTOR inhibitor, refers to a dose of mTOR inhibitor that partially, but not fully, inhibits mTOR activity, e.g., as measured by the inhibition of P70 S6 kinase activity. Methods for evaluating mTOR activity, e.g., by inhibition of P70 S6 kinase, are discussed herein. The dose is insufficient to result in complete immune suppression but is sufficient to enhance the immune response. In an embodiment, the low, immune enhancing, dose of mTOR inhibitor results in a decrease in the number of PD-1 positive T cells and/or an increase in the number of PD-1 negative T cells, or an increase in the ratio of PD-1 negative T cells/PD-1 positive T cells. In an embodiment, the low, immune enhancing, dose of mTOR inhibitor results in an increase in the number of naive T cells. In an embodiment, the low, immune enhancing, dose of mTOR inhibitor results in one or more of the following:


an increase in the expression of one or more of the following markers: CD62Lhigh, CD127high, CD27+, and BCL2, e.g., on memory T cells, e.g., memory T cell precursors;


a decrease in the expression of KLRG1, e.g., on memory T cells, e.g., memory T cell precursors; and


an increase in the number of memory T cell precursors, e.g., cells with any one or combination of the following characteristics: increased CD62Lhigh, increased CD127high, increased CD27+, decreased KLRG1, and increased BCL2;


wherein any of the changes described above occurs, e.g., at least transiently, e.g., as compared to a non-treated subject.


Ranges: throughout this disclosure, various aspects of the invention 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 invention. 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. As another example, a range such as 95-99% identity, includes something with 95%, 96%, 97%, 98% or 99% identity, and includes subranges such as 96-99%, 96-98%, 96-97%, 97-99%, 97-98% and 98-99% identity. This applies regardless of the breadth of the range.


CAR Therapies

The CAR-expressing cells described herein, e.g., CAR-expressing cells directed against CD19, and/or, CD22, may comprise one or more of the compositions described herein, e.g., a transmembrane domain, intracellular signaling domain, costimulatory domain, leader sequence, or hinge.


In one aspect, the present invention encompasses a recombinant nucleic acid construct comprising a transgene encoding a CAR. In some embodiments, the nucleic acid molecule comprises a nucleic acid sequence encoding an anti-CD19 binding domain selected from one or more of SEQ ID NOS:61-72, wherein the sequence is contiguous with and in the same reading frame as the nucleic acid sequence encoding an intracellular signaling domain. An exemplary intracellular signaling domain that can be used in the CAR includes, but is not limited to, one or more intracellular signaling domains of, e.g., CD3-zeta, CD28, 4-1BB, and the like. In some instances, the CAR can comprise any combination of CD3-zeta, CD28, 4-1BB, and the like.


In one aspect, the present invention contemplates modifications of the starting antibody or fragment (e.g., scFv) amino acid sequence that generate functionally equivalent molecules. For example, the VH or VL of an antigen binding domain, e.g., scFv, comprised in the CAR can be modified to retain at least about 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity of the starting VH or VL framework region of the antigen binding domain, e.g., scFv. The present invention contemplates modifications of the entire CAR construct, e.g., modifications in one or more amino acid sequences of the various domains of the CAR construct in order to generate functionally equivalent molecules. The CAR construct can be modified to retain at least about 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity of the starting CAR construct. The present invention also contemplates modifications of CDRs, e.g., modifications in one or more amino acid sequences of one or more CDRs of a CAR construct in order to generate functionally equivalent molecules. For instance, the CDR may have, e.g., up to and including 1, 2, 3, 4, 5, or 6 alterations (e.g., substitutions) relative to a CDR sequence provided herein.


The nucleic acid sequences coding for the desired molecules can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques. Alternatively, the nucleic acid of interest can be produced synthetically, rather than cloned.


The present invention includes, among other things, retroviral and lentiviral vector constructs expressing a CAR that can be directly transduced into a cell.


The present invention also includes an RNA construct that can be directly transfected into a cell. A method for generating mRNA for use in transfection involves in vitro transcription (IVT) of a template with specially designed primers, followed by polyA addition, to produce a construct containing 3′ and 5′ untranslated sequence (“UTR”), a 5′ cap and/or Internal Ribosome Entry Site (IRES), the nucleic acid to be expressed, and a polyA tail, typically 50-2000 bases in length (SEQ ID NO:118). RNA so produced can efficiently transfect different kinds of cells. In one embodiment, the template includes sequences for the CAR. In an embodiment, an RNA CAR vector is transduced into a T cell by electroporation.


Antigen Binding Domain

In one aspect, the CAR of the invention comprises a target-specific binding element otherwise referred to as an antigen binding domain. The choice of moiety depends upon the type and number of ligands that define the surface of a target cell. For example, the antigen binding domain may be chosen to recognize a ligand that acts as a cell surface marker on target cells associated with a particular disease state. Thus examples of cell surface markers that may act as ligands for the antigen binding domain in a CAR of the invention include those associated with viral, bacterial and parasitic infections, autoimmune disease and cancer cells. The antigen-binding domain can bind, e.g., CD19, and/or CD22.


In one aspect, the CAR-mediated T-cell response can be directed to an antigen of interest by way of engineering an antigen binding domain that specifically binds a desired antigen into the CAR.


The antigen binding domain (e.g., an antigen-binding domain that binds CD19, and/or CD22) can be any domain that binds to the antigen including but not limited to a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a murine antibody, a human antibody, a humanized antibody, and a functional fragment thereof, including but not limited to a single-domain antibody such as a heavy chain variable domain (VH), a light chain variable domain (VL) and a variable domain (VHH) of camelid derived nanobody, and to an alternative scaffold known in the art to function as antigen binding domain, such as a recombinant fibronectin domain, and the like.


In some instances, it is beneficial for the antigen binding domain to be derived from the same species in which the CAR will ultimately be used in. For example, for use in humans, it may be beneficial for the antigen binding domain (e.g., an antigen-binding domain that binds CD19, and/or CD22) of the CAR to comprise human or humanized residues for the antigen binding domain of an antibody or antibody fragment.


A humanized antibody can be produced using a variety of techniques known in the art, including but not limited to, CDR-grafting (see, e.g., European Patent No. EP 239,400; International Publication No. WO 91/09967; and U.S. Pat. Nos. 5,225,539, 5,530,101, and 5,585,089, each of which is incorporated herein in its entirety by reference), veneering or resurfacing (see, e.g., European Patent Nos. EP 592,106 and EP 519,596; Padlan, 1991, Molecular Immunology, 28(4/5):489-498; Studnicka et al., 1994, Protein Engineering, 7(6):805-814; and Roguska et al., 1994, PNAS, 91:969-973, each of which is incorporated herein by its entirety by reference), chain shuffling (see, e.g., U.S. Pat. No. 5,565,332, which is incorporated herein in its entirety by reference), and techniques disclosed in, e.g., U.S. Patent Application Publication No. US2005/0042664, U.S. Patent Application Publication No. US2005/0048617, U.S. Pat. Nos. 6,407,213, 5,766,886, International Publication No. WO 9317105, Tan et al., J. Immunol., 169:1119-25 (2002), Caldas et al., Protein Eng., 13(5):353-60 (2000), Morea et al., Methods, 20(3):267-79 (2000), Baca et al., J. Biol. Chem., 272(16):10678-84 (1997), Roguska et al., Protein Eng., 9(10):895-904 (1996), Couto et al., Cancer Res., 55 (23 Supp):5973s-5977s (1995), Couto et al., Cancer Res., 55(8):1717-22 (1995), Sandhu J S, Gene, 150(2):409-10 (1994), and Pedersen et al., J. Mol. Biol., 235(3):959-73 (1994), each of which is incorporated herein in its entirety by reference. Often, framework residues in the framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, for example improve, antigen binding. These framework substitutions are identified by methods well-known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No. 5,585,089; and Riechmann et al., 1988, Nature, 332:323, which are incorporated herein by reference in their entireties.)


A humanized antibody or antibody fragment has one or more amino acid residues remaining in it from a source which is nonhuman. These nonhuman amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. As provided herein, humanized antibodies or antibody fragments comprise one or more CDRs from nonhuman immunoglobulin molecules and framework regions wherein the amino acid residues comprising the framework are derived completely or mostly from human germline. Multiple techniques for humanization of antibodies or antibody fragments are well-known in the art and can essentially be performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody, i.e., CDR-grafting (EP 239,400; PCT Publication No. WO 91/09967; and U.S. Pat. Nos. 4,816,567; 6,331,415; 5,225,539; 5,530,101; 5,585,089; 6,548,640, the contents of which are incorporated herein by reference herein in their entirety). In such humanized antibodies and antibody fragments, substantially less than an intact human variable domain has been substituted by the corresponding sequence from a nonhuman species. Humanized antibodies are often human antibodies in which some CDR residues and possibly some framework (FR) residues are substituted by residues from analogous sites in rodent antibodies. Humanization of antibodies and antibody fragments can also be achieved by veneering or resurfacing (EP 592,106; EP 519,596; Padlan, 1991, Molecular Immunology, 28(4/5):489-498; Studnicka et al., Protein Engineering, 7(6):805-814 (1994); and Roguska et al., PNAS, 91:969-973 (1994)) or chain shuffling (U.S. Pat. No. 5,565,332), the contents of which are incorporated herein by reference herein in their entirety.


The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is to reduce antigenicity. According to the so-called “best-fit” method, the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the rodent is then accepted as the human framework (FR) for the humanized antibody (Sims et al., J. Immunol., 151:2296 (1993); Chothia et al., J. Mol. Biol., 196:901 (1987), the contents of which are incorporated herein by reference herein in their entirety). Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (see, e.g., Nicholson et al. Mol. Immun. 34 (16-17): 1157-1165 (1997); Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993), the contents of which are incorporated herein by reference herein in their entirety). In some embodiments, the framework region, e.g., all four framework regions, of the heavy chain variable region are derived from a VH4_4-59 germline sequence. In one embodiment, the framework region can comprise, one, two, three, four or five modifications, e.g., substitutions, e.g., from the amino acid at the corresponding murine sequence (e.g., of SEQ ID NO:59). In one embodiment, the framework region, e.g., all four framework regions of the light chain variable region are derived from a VK3_1.25 germline sequence. In one embodiment, the framework region can comprise, one, two, three, four or five modifications, e.g., substitutions, e.g., from the amino acid at the corresponding murine sequence (e.g., of SEQ ID NO:59).


In some aspects, the portion of a CAR composition of the invention that comprises an antibody fragment is humanized with retention of high affinity for the target antigen and other favorable biological properties. According to one aspect of the invention, humanized antibodies and antibody fragments are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, e.g., the analysis of residues that influence the ability of the candidate immunoglobulin to bind the target antigen. In this way, FR residues can be selected and combined from the recipient and import sequences so that the desired antibody or antibody fragment characteristic, such as increased affinity for the target antigen, is achieved. In general, the CDR residues are directly and most substantially involved in influencing antigen binding.


A humanized antibody or antibody fragment may retain a similar antigenic specificity as the original antibody, e.g., in the present invention, the ability to bind human CD19, or CD22. In some embodiments, a humanized antibody or antibody fragment may have improved affinity and/or specificity of binding to human CD19, or CD22.


In one aspect, the binding domain (e.g., an antigen-binding domain that binds CD19, or CD22) is a fragment, e.g., a single chain variable fragment (scFv). In one aspect, the binding domain is a Fv, a Fab, a (Fab′)2, or a bi-functional (e.g. bi-specific) hybrid antibody (e.g., Lanzavecchia et al., Eur. J. Immunol. 17, 105 (1987)). In one aspect, the antibodies and fragments thereof of the invention binds a CD19, or CD22 protein with wild-type or enhanced affinity.


In some instances, scFvs can be prepared according to method known in the art (see, for example, Bird et al., (1988) Science 242:423-426 and Huston et al., (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). ScFv molecules can be produced by linking VH and VL regions together using flexible polypeptide linkers. The scFv molecules comprise a linker (e.g., a Ser-Gly linker) with an optimized length and/or amino acid composition. The linker length can greatly affect how the variable regions of a scFv fold and interact. In fact, if a short polypeptide linker is employed (e.g., between 5-10 amino acids) intrachain folding is prevented. Interchain folding is also required to bring the two variable regions together to form a functional epitope binding site. For examples of linker orientation and size see, e.g., Hollinger et al. 1993 Proc Natl Acad. Sci. U.S.A. 90:6444-6448, U.S. Patent Application Publication Nos. 2005/0100543, 2005/0175606, 2007/0014794, and PCT publication Nos. WO2006/020258 and WO2007/024715, is incorporated herein by reference.


An scFv can comprise a linker of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more amino acid residues between its VL and VH regions. The linker sequence may comprise any naturally occurring amino acid. In some embodiments, the linker sequence comprises amino acids glycine and serine. In another embodiment, the linker sequence comprises sets of glycine and serine repeats such as (Gly4Ser)n, where n is a positive integer equal to or greater than 1 (SEQ ID NO:18). In one embodiment, the linker can be (Gly4Ser)4 (SEQ ID NO:106) or (Gly4Ser)3(SEQ ID NO:107). Variation in the linker length may retain or enhance activity, giving rise to superior efficacy in activity studies.


In some embodiments, the amino acid sequence of the antigen binding domain (e.g., an antigen-binding domain that binds CD19, or CD22) or other portions or the entire CAR) can be modified, e.g., an amino acid sequence described herein can be modified, e.g., by a conservative substitution. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).


Percent identity in the context of two or more nucleic acids or polypeptide sequences, refers to two or more sequences that are the same. Two sequences are “substantially identical” if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (e.g., 60% identity, optionally 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity over a specified region, or, when not specified, over the entire sequence), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Optionally, the identity exists over a region that is at least about 50 nucleotides (or 10 amino acids) in length, or over a region that is 100 to 500 or 1000 or more nucleotides (or 20, 50, 200 or more amino acids) in length.


For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman, (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch, (1970) J. Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman, (1988) Proc. Nat'l. Acad. Sci. USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection (see, e.g., Brent et al., (2003) Current Protocols in Molecular Biology).


Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al., (1977) Nuc. Acids Res. 25:3389-3402; and Altschul et al., (1990) J. Mol. Biol. 215:403-410, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.


The percent identity between two amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller, (1988) Comput. Appl. Biosci. 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (1970) J. Mol. Biol. 48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package (available at www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.


In one aspect, the present invention contemplates modifications of the starting antibody or fragment (e.g., scFv) amino acid sequence that generate functionally equivalent molecules. For example, the VH or VL of a binding domain (e.g., an antigen-binding domain that binds CD19, or CD22), e.g., scFv, comprised in the CAR can be modified to retain at least about 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity of the starting VH or VL framework region of the anti-CD19 binding domain, e.g., scFv. In an aspect, the VH or VL of a CD22 antigen binding domain, e.g., scFv, comprised in the CAR can be modified to retain at least about 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity of the starting VH or VL framework region of the anti-CD22 antigen binding domain, e.g., scFv. The present invention contemplates modifications of the entire CAR construct, e.g., modifications in one or more amino acid sequences of the various domains of the CAR construct in order to generate functionally equivalent molecules. The CAR construct can be modified to retain at least about 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity of the starting CAR construct.


Chimeric TCR

In one aspect, the antibodies and antibody fragments disclosed herein (e.g., those directed against CD19, or CD22) can be grafted to one or more constant domain of a T cell receptor (“TCR”) chain, for example, a TCR alpha or TCR beta chain, to create an chimeric TCR that binds specifically to a cancer associated antigen. Without being bound by theory, it is believed that chimeric TCRs will signal through the TCR complex upon antigen binding. For example, an scFv as disclosed herein, can be grafted to the constant domain, e.g., at least a portion of the extracellular constant domain, the transmembrane domain and the cytoplasmic domain, of a TCR chain, for example, the TCR alpha chain and/or the TCR beta chain. As another example, an antibody fragment, for example a VL domain as described herein, can be grafted to the constant domain of a TCR alpha chain, and an antibody fragment, for example a VH domain as described herein, can be grafted to the constant domain of a TCR beta chain (or alternatively, a VL domain may be grafted to the constant domain of the TCR beta chain and a VH domain may be grafted to a TCR alpha chain). As another example, the CDRs of an antibody or antibody fragment, e.g., the CDRs of an antibody or antibody fragment as described in any of the Tables herein may be grafted into a TCR alpha and/or beta chain to create a chimeric TCR that binds specifically to a cancer associated antigen. For example, the LC CDRs disclosed herein may be grafted into the variable domain of a TCR alpha chain and the HC CDRs disclosed herein may be grafted to the variable domain of a TCR beta chain, or vice versa. Such chimeric TCRs may be produced by any appropriate method (For example, Willemsen R A et al, Gene Therapy 2000; 7: 1369-1377; Zhang T et al, Cancer Gene Ther 2004; 11: 487-496; Aggen et al, Gene Ther. 2012 April; 19(4):365-74).


Linkers for Antigen Binding Domains

It was found that CAR molecules comprising a short or no linker between the variable domains (e.g., VH and VL) of the antigen binding domain showed equal to, or greater, activity than longer versions of the linker. For example, in some embodiments, CD22-65s (having (Gly4-Ser)n linker, wherein n is 1 (SEQ ID NO: 18)) shows comparable or greater activity and/or efficacy in a tumor model, compared to CD22-65 (having (Gly4-Ser)n linker, wherein n is 3 (SEQ ID NO: 107)). Accordingly, any of the antigen binding domains or CAR molecules described herein can have a linker connecting the variable domains of the antigen binding domain of varying lengths, including for example, a short linker of about 3 to 6 amino acids, 4 to 5 amino acids, or about 5 amino acids. In some embodiments, a longer linker can be used, e.g., about 6 to 35 amino acids, e.g., 8 to 32 amino acids, 10 to 30 amino acids, 10 to 20 amino acids. For example, a (Gly4-Ser)n linker, wherein n is 0, 1, 2, 3, 4, 5, or 6 (SEQ ID NO: 53) can be used. In one embodiment, the variable domains are not connected via a linker, e.g., (Gly4-Ser)n linker, n=0 (“Gly4-Ser” is disclosed as SEQ ID NO: 18). In some embodiments, the variable domains are connected via a short linker, e.g., (Gly4-Ser)n linker, n=1 (SEQ ID NO: 18). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=2 (SEQ ID NO: 49). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=3 (SEQ ID NO: 107). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=4 (SEQ ID NO: 106). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=5 (SEQ ID NO: 1341). In some embodiments, the variable domains are connected via a (Gly4-Ser)n linker, n=6 (SEQ ID NO: 1342). The order of the variable domain, e.g., in which the VL and VH domains appear in the antigen binding domain, e.g., scFv, can be varied (i.e., VL-VH, or VH-VL orientation). In one embodiment, the antigen binding domain binds to CD20, e.g., a CD20 antigen binding domain as described herein. In another embodiment, the antigen binding domain binds to CD22, e.g., a CD22 antigen binding domain as described herein. In another embodiment, the antigen binding domain binds to CD19, e.g., a CD19 antigen binding domain as described herein.


Non-Antibody Scaffolds

In embodiments, the antigen binding domain comprises a non antibody scaffold, e.g., a fibronectin, ankyrin, domain antibody, lipocalin, small modular immuno-pharmaceutical, maxybody, Protein A, or affilin. The non antibody scaffold has the ability to bind to target antigen on a cell. In embodiments, the antigen binding domain is a polypeptide or fragment thereof of a naturally occurring protein expressed on a cell. In some embodiments, the antigen binding domain comprises a non-antibody scaffold. A wide variety of non-antibody scaffolds can be employed so long as the resulting polypeptide includes at least one binding region which specifically binds to the target antigen on a target cell.


Non-antibody scaffolds include: fibronectin (Novartis, Mass.), ankyrin (Molecular Partners AG, Zurich, Switzerland), domain antibodies (Domantis, Ltd., Cambridge, Mass., and Ablynx nv, Zwijnaarde, Belgium), lipocalin (Pieris Proteolab AG, Freising, Germany), small modular immuno-pharmaceuticals (Trubion Pharmaceuticals Inc., Seattle, Wash.), maxybodies (Avidia, Inc., Mountain View, Calif.), Protein A (Affibody AG, Sweden), and affilin (gamma-crystallin or ubiquitin) (Scil Proteins GmbH, Halle, Germany).


Fibronectin scaffolds can be based on fibronectin type III domain (e.g., the tenth module of the fibronectin type III (10 Fn3 domain)). The fibronectin type III domain has 7 or 8 beta strands which are distributed between two beta sheets, which themselves pack against each other to form the core of the protein, and further containing loops (analogous to CDRs) which connect the beta strands to each other and are solvent exposed. There are at least three such loops at each edge of the beta sheet sandwich, where the edge is the boundary of the protein perpendicular to the direction of the beta strands (see U.S. Pat. No. 6,818,418). Because of this structure, this non-antibody scaffold mimics antigen binding properties that are similar in nature and affinity to those of antibodies. These scaffolds can be used in a loop randomization and shuffling strategy in vitro that is similar to the process of affinity maturation of antibodies in vivo.


The ankyrin technology is based on using proteins with ankyrin derived repeat modules as scaffolds for bearing variable regions which can be used for binding to different targets. The ankyrin repeat module is a 33 amino acid polypeptide consisting of two anti-parallel α-helices and a β-turn. Binding of the variable regions is mostly optimized by using ribosome display.


Avimers are derived from natural A-domain containing protein such as HERS. These domains are used by nature for protein-protein interactions and in human over 250 proteins are structurally based on A-domains. Avimers consist of a number of different “A-domain” monomers (2-10) linked via amino acid linkers. Avimers can be created that can bind to the target antigen using the methodology described in, for example, U.S. Patent Application Publication Nos. 20040175756; 20050053973; 20050048512; and 20060008844.


Affibody affinity ligands are small, simple proteins composed of a three-helix bundle based on the scaffold of one of the IgG-binding domains of Protein A. Protein A is a surface protein from the bacterium Staphylococcus aureus. This scaffold domain consists of 58 amino acids, 13 of which are randomized to generate affibody libraries with a large number of ligand variants (See e.g., U.S. Pat. No. 5,831,012). Affibody molecules mimic antibodies, they have a molecular weight of 6 kDa, compared to the molecular weight of antibodies, which is 150 kDa. In spite of its small size, the binding site of affibody molecules is similar to that of an antibody.


Protein epitope mimetics (PEM) are medium-sized, cyclic, peptide-like molecules (MW 1-2 kDa) mimicking beta-hairpin secondary structures of proteins, the major secondary structure involved in protein-protein interactions. Antigen binding domains, e.g., those comprising scFv, single domain antibodies, or camelid antibodies, can be directed to any target receptor/ligand described herein, e.g., the PD1 receptors, PD-L1 or PD-L2.


In an embodiment the antigen binding domain comprises the extracellular domain, or a counter-ligand binding fragment thereof, of molecule that binds a counterligand on the surface of a target cell.


An antigen binding domain can comprise the extracellular domain of an inhibitory receptor. Engagement with a counterligand of the coinhibitory molecule is redirected into an optimization of immune effector response.


An antigen binding domain can comprise the extracellular domain of a costimulatory molecule, referred to as a Costimulatory ECD domain, Engagement with a counter ligand of the costimulatory molecule results in optimization of immune effector response.


Transmembrane Domain

With respect to the transmembrane domain, in various embodiments, a CAR can be designed to comprise a transmembrane domain that is attached to the extracellular domain of the CAR. A transmembrane domain can include one or more additional amino acids adjacent to the transmembrane region, e.g., one or more amino acid associated with the extracellular region of the protein from which the transmembrane was derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the extracellular region) and/or one or more additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the intracellular region). In one aspect, the transmembrane domain is one that is associated with one of the other domains of the CAR, e.g., in one embodiment, the transmembrane domain may be from the same protein that the signaling domain, costimulatory domain or the hinge domain is derived from. In another aspect, the transmembrane domain is not derived from the same protein that any other domain of the CAR is derived from. In some instances, the transmembrane domain can be selected or modified by amino acid substitution to avoid binding of such domains to the transmembrane domains of the same or different surface membrane proteins, e.g., to minimize interactions with other members of the receptor complex. In one aspect, the transmembrane domain is capable of homodimerization with another CAR on the cell surface of a CAR-expressing cell. In a different aspect the amino acid sequence of the transmembrane domain may be modified or substituted so as to minimize interactions with the binding domains of the native binding partner present in the same CAR-expressing cell.


The transmembrane domain may be derived either from a natural or from a recombinant source. Where the source is natural, the domain may be derived from any membrane-bound or transmembrane protein. In one aspect the transmembrane domain is capable of signaling to the intracellular domain(s) whenever the CAR has bound to a target. A transmembrane domain of particular use in this invention may include at least the transmembrane region(s) of e.g., the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154. In some embodiments, a transmembrane domain may include at least the transmembrane region(s) of, e.g., KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, IL2R beta, IL2R gamma, IL7R a, ITGA1, 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, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKG2D, NKG2C, or CD19.


In some instances, the transmembrane domain can be attached to the extracellular region of the CAR, e.g., the antigen binding domain of the CAR, via a hinge, e.g., a hinge from a human protein. For example, in one embodiment, the hinge can be a human Ig (immunoglobulin) hinge, e.g., an IgG4 hinge, an IgD hinge, a GS linker (e.g., a GS linker described herein), a KIR2DS2 hinge, or a CD8a hinge. In one embodiment, the hinge or spacer comprises (e.g., consists of) the amino acid sequence of SEQ ID NO:14. In one aspect, the transmembrane domain comprises (e.g., consists of) a transmembrane domain of SEQ ID NO: 15.


In one aspect, the hinge or spacer comprises an IgG4 hinge. For example, in one embodiment, the hinge or spacer comprises a hinge of the amino acid sequence ESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWY VDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTI SKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGKM (SEQ ID NO:45). In some embodiments, the hinge or spacer comprises a hinge encoded by a nucleotide sequence of









(SEQ ID NO: 46)


GAGAGCAAGTACGGCCCTCCCTGCCCCCCTTGCCCTGCCCCCGAGTTCC





TGGGCGGACCCAGCGTGTTCCTGTTCCCCCCCAAGCCCAAGGACACCCT





GATGATCAGCCGGACCCCCGAGGTGACCTGTGTGGTGGTGGACGTGTCC





CAGGAGGACCCCGAGGTCCAGTTCAACTGGTACGTGGACGGCGTGGAGG





TGCACAACGCCAAGACCAAGCCCCGGGAGGAGCAGTTCAATAGCACCTA





CCGGGTGGTGTCCGTGCTGACCGTGCTGCACCAGGACTGGCTGAACGGC





AAGGAATACAAGTGTAAGGTGTCCAACAAGGGCCTGCCCAGCAGCATCG





AGAAAACCATCAGCAAGGCCAAGGGCCAGCCTCGGGAGCCCCAGGTGTA





CACCCTGCCCCCTAGCCAAGAGGAGATGACCAAGAACCAGGTGTCCCTG





ACCTGCCTGGTGAAGGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGG





AGAGCAACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCTGTGCT





GGACAGCGACGGCAGCTTCTTCCTGTACAGCCGGCTGACCGTGGACAAG





AGCCGGTGGCAGGAGGGCAACGTCTTTAGCTGCTCCGTGATGCACGAGG





CCCTGCACAACCACTACACCCAGAAGAGCCTGAGCCTGTCCCTGGGCAA





GATG.






In one aspect, the hinge or spacer comprises an IgD hinge. For example, in one embodiment, the hinge or spacer comprises a hinge of the amino acid sequence RWPESPKAQASSVPTAQPQAEGSLAKATTAPATTRNTGRGGEEKKKEKEKEEQEERET KTPECPSHTQPLGVYLLTPAVQDLWLRDKATFTCFVVGSDLKDAHLTWEVAGKVPTG GVEEGLLERHSNGSQSQHSRLTLPRSLWNAGTSVTCTLNHPSLPPQRLMALREPAAQAP VKLSLNLLASSDPPEAASWLLCEVSGFSPPNILLMWLEDQREVNTSGFAPARPPPQPGST TFWAWSVLRVPAPPSPQPATYTCVVSHEDSRTLLNASRSLEVSYVTDH (SEQ ID NO:47). In some embodiments, the hinge or spacer comprises a hinge encoded by a nucleotide sequence of









(SEQ ID NO: 48)


AGGTGGCCCGAAAGTCCCAAGGCCCAGGCATCTAGTGTTCCTACTGCAC





AGCCCCAGGCAGAAGGCAGCCTAGCCAAAGCTACTACTGCACCTGCCAC





TACGCGCAATACTGGCCGTGGCGGGGAGGAGAAGAAAAAGGAGAAAGAG





AAAGAAGAACAGGAAGAGAGGGAGACCAAGACCCCTGAATGTCCATCCC





ATACCCAGCCGCTGGGCGTCTATCTCTTGACTCCCGCAGTACAGGACTT





GTGGCTTAGAGATAAGGCCACCTTTACATGTTTCGTCGTGGGCTCTGAC





CTGAAGGATGCCCATTTGACTTGGGAGGTTGCCGGAAAGGTACCCACAG





GGGGGGTTGAGGAAGGGTTGCTGGAGCGCCATTCCAATGGCTCTCAGAG





CCAGCACTCAAGACTCACCCTTCCGAGATCCCTGTGGAACGCCGGGACC





TCTGTCACATGTACTCTAAATCATCCTAGCCTGCCCCCACAGCGTCTGA





TGGCCCTTAGAGAGCCAGCCGCCCAGGCACCAGTTAAGCTTAGCCTGAA





TCTGCTCGCCAGTAGTGATCCCCCAGAGGCCGCCAGCTGGCTCTTATGC





GAAGTGTCCGGCTTTAGCCCGCCCAACATCTTGCTCATGTGGCTGGAGG





ACCAGCGAGAAGTGAACACCAGCGGCTTCGCTCCAGCCCGGCCCCCACC





CCAGCCGGGTTCTACCACATTCTGGGCCTGGAGTGTCTTAAGGGTCCCA





GCACCACCTAGCCCCCAGCCAGCCACATACACCTGTGTTGTGTCCCATG





AAGATAGCAGGACCCTGCTAAATGCTTCTAGGAGTCTGGAGGTTTCCTA





CGTGACTGACCATT.






In one aspect, the transmembrane domain may be recombinant, in which case it will comprise predominantly hydrophobic residues such as leucine and valine. In one aspect a triplet of phenylalanine, tryptophan and valine can be found at each end of a recombinant transmembrane domain.


Optionally, a short oligo- or polypeptide linker, between 2 and 10 amino acids in length may form the linkage between the transmembrane domain and the cytoplasmic region of the CAR. A glycine-serine doublet provides a particularly suitable linker. For example, in one aspect, the linker comprises the amino acid sequence of GGGGSGGGGS (SEQ ID NO:49). In some embodiments, the linker is encoded by a nucleotide sequence of











(SEQ ID NO: 50)



GGTGGCGGAGGTTCTGGAGGTGGAGGTTCC.






In one aspect, the hinge or spacer comprises a KIR2DS2 hinge.


Cytoplasmic Domain

The cytoplasmic domain or region of the CAR includes an intracellular signaling domain. An intracellular signaling domain is generally responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR has been introduced.


Examples of intracellular signaling domains for use in the CAR of the invention include the cytoplasmic sequences of the T cell receptor (TCR) and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivative or variant of these sequences and any recombinant sequence that has the same functional capability.


It is known that signals generated through the TCR alone are insufficient for full activation of the T cell and that a secondary and/or costimulatory signal is also required. Thus, T cell activation can be said to be mediated by two distinct classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation through the TCR (primary intracellular signaling domains) and those that act in an antigen-independent manner to provide a secondary or costimulatory signal (secondary cytoplasmic domain, e.g., a costimulatory domain).


A primary signaling domain regulates primary activation of the TCR complex either in a stimulatory way, or in an inhibitory way. Primary intracellular signaling domains that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or ITAMs.


Examples of ITAM containing primary intracellular signaling domains that are of particular use in the invention include those of CD3 zeta, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon R1b), CD3 gamma, CD3 delta, CD3 epsilon, CD79a, CD79b, CD278 (also known as “ICOS”), FcεRI, DAP10, DAP12, and CD66d. In one embodiment, a CAR of the invention comprises an intracellular signaling domain, e.g., a primary signaling domain of CD3-zeta.


In one embodiment, a primary signaling domain comprises a modified ITAM domain, e.g., a mutated ITAM domain which has altered (e.g., increased or decreased) activity as compared to the native ITAM domain. In one embodiment, a primary signaling domain comprises a modified ITAM-containing primary intracellular signaling domain, e.g., an optimized and/or truncated ITAM-containing primary intracellular signaling domain. In an embodiment, a primary signaling domain comprises one, two, three, four or more ITAM motifs.


Further examples of molecules containing a primary intracellular signaling domain that are of particular use in the invention include those of DAP10, DAP12, and CD32.


Costimulatory Signaling Domain

The intracellular signalling domain of the CAR can comprise the CD3-zeta signaling domain by itself or it can be combined with any other desired intracellular signaling domain(s) useful in the context of a CAR of the invention. For example, the intracellular signaling domain of the CAR can comprise a CD3 zeta chain portion and a costimulatory signaling domain. The costimulatory signaling domain refers to a portion of the CAR comprising the intracellular domain of a costimulatory molecule. In one embodiment, the intracellular domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of CD28. In one aspect, the intracellular domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of ICOS.


A costimulatory molecule can be a cell surface molecule other than an antigen receptor or its ligands that is required for an efficient response of lymphocytes to an antigen. Examples of such molecules include CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, and a ligand that specifically binds with CD83, and the like. For example, CD27 costimulation has been demonstrated to enhance expansion, effector function, and survival of human CART cells in vitro and augments human T cell persistence and antitumor activity in vivo (Song et al. Blood. 2012; 119(3):696-706). Further examples of such costimulatory molecules include MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, 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, NKG2D, NKG2C, 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, CD19a, and a ligand that specifically binds with CD83.


The intracellular signaling sequences within the cytoplasmic portion of the CAR of the invention may be linked to each other in a random or specified order. Optionally, a short oligo- or polypeptide linker, for example, between 2 and 10 amino acids (e.g., 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) in length may form the linkage between intracellular signaling sequence. In one embodiment, a glycine-serine doublet can be used as a suitable linker. In one embodiment, a single amino acid, e.g., an alanine, a glycine, can be used as a suitable linker.


In one aspect, the intracellular signaling domain is designed to comprise two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains. In an embodiment, the two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains, are separated by a linker molecule, e.g., a linker molecule described herein. In one embodiment, the intracellular signaling domain comprises two costimulatory signaling domains. In some embodiments, the linker molecule is a glycine residue. In some embodiments, the linker is an alanine residue.


In one aspect, the intracellular signaling domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of CD28. In one aspect, the intracellular signaling domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of 4-1BB. In one aspect, the signaling domain of 4-1BB is a signaling domain of SEQ ID NO: 16. In one aspect, the signaling domain of CD3-zeta is a signaling domain of SEQ ID NO: 17.


In one aspect, the intracellular signaling domain is designed to comprise the signaling domain of CD3-zeta and the signaling domain of CD27. In one aspect, the signaling domain of CD27 comprises an amino acid sequence of









(SEQ ID NO: 51)


QRRKYRSNKGESPVEPAEPCRYSCPREEEGSTIPIQEDYRKPEPACSP.







In one aspect, the signalling domain of CD27 is encoded by a nucleic acid sequence of









(SEQ ID NO: 52)


AGGAGTAAGAGGAGCAGGCTCCTGCACAGTGACTACATGAACATGACTC





CCCGCCGCCCCGGGCCCACCCGCAAGCATTACCAGCCCTATGCCCCACC





ACGCGACTTCGCAGCCTATCGCTCC.






Natural Killer Cell Receptor (NKR) CARs

In an embodiment, a CAR molecule described herein comprises one or more components of a natural killer cell receptor (NKR), thereby forming an NKR-CAR. The NKR component can be a transmembrane domain, a hinge domain, or a cytoplasmic domain from any of the following natural killer cell receptors: killer cell immunoglobulin-like receptor (KIR), e.g., KIR2DL1, KIR2DL2/L3, KIR2DL4, KIR2DL5A, KIR2DL5B, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, DIR2DS5, KIR3DL1/S1, KIR3DL2, KIR3DL3, KIR2DP1, and KIR3DP1; natural cytotoxicity receptor (NCR), e.g., NKp30, NKp44, NKp46; signaling lymphocyte activation molecule (SLAM) family of immune cell receptors, e.g., CD48, CD229, 2B4, CD84, NTB-A, CRACC, BLAME, and CD2F-10; Fc receptor (FcR), e.g., CD16, and CD64; and Ly49 receptors, e.g., LY49A, LY49C. The NKR-CAR molecules described herein may interact with an adaptor molecule or intracellular signaling domain, e.g., DAP12. Exemplary configurations and sequences of CAR molecules comprising NKR components are described in International Publication No. WO2014/145252, the contents of which are hereby incorporated by reference.


Strategies for Regulating Chimeric Antigen Receptors

In some embodiments, a regulatable CAR (RCAR) where the CAR activity can be controlled is desirable to optimize the safety and efficacy of a CAR therapy. There are many ways CAR activities can be regulated. For example, inducing apoptosis using, e.g., a caspase fused to a dimerization domain (see, e.g., Di et al., N Engl. J. Med. 2011 Nov. 3; 365(18):1673-1683), can be used as a safety switch in the CAR therapy of the instant invention. In one embodiment, the cells (e.g., T cells or NK cells) expressing a CAR of the present invention further comprise an inducible apoptosis switch, wherein a human caspase (e.g., caspase 9) or a modified version is fused to a modification of the human FKB protein that allows conditional dimerization. In the presence of a small molecule, such as a rapalog (e.g., AP 1903, AP20187), the inducible caspase (e.g., caspase 9) is activated and leads to the rapid apoptosis and death of the cells (e.g., T cells or NK cells) expressing a CAR of the present invention. Examples of a caspase-based inducible apoptosis switch (or one or more aspects of such a switch) have been described in, e.g., US2004040047; US20110286980; US20140255360; WO1997031899; WO2014151960; WO2014164348; WO2014197638; WO2014197638; all of which are incorporated by reference herein.


In another example, CAR-expressing cells can also express an inducible Caspase-9 (iCaspase-9) molecule that, upon administration of a dimerizer drug (e.g., rimiducid (also called AP1903 (Bellicum Pharmaceuticals) or AP20187 (Ariad)) leads to activation of the Caspase-9 and apoptosis of the cells. The iCaspase-9 molecule contains a chemical inducer of dimerization (CID) binding domain that mediates dimerization in the presence of a CID. This results in inducible and selective depletion of CAR-expressing cells. In some cases, the iCaspase-9 molecule is encoded by a nucleic acid molecule separate from the CAR-encoding vector(s). In some cases, the iCaspase-9 molecule is encoded by the same nucleic acid molecule as the CAR-encoding vector. The iCaspase-9 can provide a safety switch to avoid any toxicity of CAR-expressing cells. See, e.g., Song et al. Cancer Gene Ther. 2008; 15(10):667-75; Clinical Trial Id. No. NCT02107963; and Di Stasi et al. N. Engl. J. Med. 2011; 365:1673-83.


Alternative strategies for regulating the CAR therapy of the instant invention include utilizing small molecules or antibodies that deactivate or turn off CAR activity, e.g., by deleting CAR-expressing cells, e.g., by inducing antibody dependent cell-mediated cytotoxicity (ADCC). For example, CAR-expressing cells described herein may also express an antigen that is recognized by molecules capable of inducing cell death, e.g., ADCC or complement-induced cell death. For example, CAR expressing cells described herein may also express a receptor capable of being targeted by an antibody or antibody fragment. Examples of such receptors include EpCAM, VEGFR, integrins (e.g., integrins ανβ3, α4, αI¾β3, α4β7, α5β1, ανβ3, αν), members of the TNF receptor superfamily (e.g., TRAIL-R1, TRAIL-R2), PDGF Receptor, interferon receptor, folate receptor, GPNMB, ICAM-1, HLA-DR, CEA, CA-125, MUC1, TAG-72, IL-6 receptor, 5T4, GD2, GD3, CD2, CD3, CD4, CD5, CD11, CD11 a/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/lgE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin, CD152/CTLA-4, CD154/CD40L, CD195/CCR5, CD319/SLAMF7, and EGFR, and truncated versions thereof (e.g., versions preserving one or more extracellular epitopes but lacking one or more regions within the cytoplasmic domain).


For example, a CAR-expressing cell described herein may also express a truncated epidermal growth factor receptor (EGFR) which lacks signaling capacity but retains the epitope that is recognized by molecules capable of inducing ADCC, e.g., cetuximab (ERBITUX®), such that administration of cetuximab induces ADCC and subsequent depletion of the CAR-expressing cells (see, e.g., WO2011/056894, and Jonnalagadda et al., Gene Ther. 2013; 20(8)853-860). Another strategy includes expressing a highly compact marker/suicide gene that combines target epitopes from both CD32 and CD20 antigens in the CAR-expressing cells described herein, which binds rituximab, resulting in selective depletion of the CAR-expressing cells, e.g., by ADCC (see, e.g., Philip et al., Blood. 2014; 124(8)1277-1287). Other methods for depleting CAR-expressing cells described herein include administration of CAMPATH, a monoclonal anti-CD52 antibody that selectively binds and targets mature lymphocytes, e.g., CAR-expressing cells, for destruction, e.g., by inducing ADCC. In other embodiments, the CAR-expressing cell can be selectively targeted using a CAR ligand, e.g., an anti-idiotypic antibody. In some embodiments, the anti-idiotypic antibody can cause effector cell activity, e.g., ADCC or ADC activities, thereby reducing the number of CAR-expressing cells. In other embodiments, the CAR ligand, e.g., the anti-idiotypic antibody, can be coupled to an agent that induces cell killing, e.g., a toxin, thereby reducing the number of CAR-expressing cells. Alternatively, the CAR molecules themselves can be configured such that the activity can be regulated, e.g., turned on and off, as described below.


In other embodiments, a CAR-expressing cell described herein may also express a target protein recognized by the T cell depleting agent. In one embodiment, the target protein is CD20 and the T cell depleting agent is an anti-CD20 antibody, e.g., rituximab. In such embodiment, the T cell depleting agent is administered once it is desirable to reduce or eliminate the CAR-expressing cell, e.g., to mitigate the CAR induced toxicity. In other embodiments, the T cell depleting agent is an anti-CD52 antibody, e.g., alemtuzumab.


In an aspect, a RCAR comprises a set of polypeptides, typically two in the simplest embodiments, in which the components of a standard CAR described herein, e.g., an antigen binding domain and an intracellular signaling domain, are partitioned on separate polypeptides or members. In some embodiments, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signaling domain. In one embodiment, a CAR of the present invention utilizes a dimerization switch as those described in, e.g., WO2014127261, which is incorporated by reference herein. Additional description and exemplary configurations of such regulatable CARs are provided herein and in International Publication No. WO 2015/090229, hereby incorporated by reference in its entirety.


In some embodiments, an RCAR involves a switch domain, e.g., a FKBP switch domain, as set out SEQ ID NO: 122, or comprise a fragment of FKBP having the ability to bind with FRB, e.g., as set out in SEQ ID NO: 123. In some embodiments, the RCAR involves a switch domain comprising a FRB sequence, e.g., as set out in SEQ ID NO: 124, or a mutant FRB sequence, e.g., as set out in any of SEQ ID Nos. 125-130.











(SEQ ID NO: 122)



D V P D Y A S L G G P S S P K







K K R K V S R G V Q V E T I S








P G D G R T F P K R G Q T C V









V H Y T G M L E D G K K F D S









S R D R N K P F K F M L G K Q









E V I R G W E E G V A Q M S V









G Q R A K L T I S P D Y A Y G









A T G H P G I I P P H A T L V









F D V E L L K L E T S Y








(SEQ ID NO: 123)



V Q V E T I S P G D G R T F P







K R G Q T C V V H Y T G M L E







D G K K F D S S R D R N K P F







K F M L G K Q E V I R G W E E







G V A Q M S V G Q R A K L T I







S P D Y A Y G A T G H P G I I







P P H A T L V F D V E L L K L







E T S







(SEQ ID NO: 124)



ILWHEMWHEG LEEASRLYFG ERNVKGMFEV







LEPLHAMMER GPQTLKETSF NQAYGRDLME







AQEWCRKYMK SGNVKDLTQA WDLYYHVFRR







ISK













TABLE 1







Exemplary mutant FRB having increased


affinity for a dimerization molecule.













SEQ



FRB

ID



mutant
Amino Acid Sequence
NO:







E2032I
ILWHEMWHEGLIEASRLYFG
125



mutant
ERNVKGMFEVLEPLHAMMER





GPQTLKETSFNQAYGRDLME





AQEWCRKYMKSGNVKDLTQA





WDLYYHVFRRISKTS








E2032L
ILWHEMWHEGLLEASRLYFG
126



mutant
ERNVKGMFEVLEPLHAMMER





GPQTLKETSFNQAYGRDLME





AQEWCRKYMKSGNVKDLTQA





WDLYYHVFRRISKTS








T2098L
ILWHEMWHEGLEEASRLYFG
127



mutant
ERNVKGMFEVLEPLHAMMER





GPQTLKETSFNQAYGRDLME





AQEWCRKYMKSGNVKDLLQA





WDLYYHVFRRISKTS








E2032,
ILWHEMWHEGLXEASRLYFG
128



T2098
ERNVKGMFEVLEPLHAMMER




mutant
GPQTLKETSFNQAYGRDLME





AQEWCRKYMKSGNVKDLXQA





WDLYYHVFRRI





SKTS








E2032I,
ILWHEMWHEGLIEASRLYFG
129



T2098L
ERNVKGMFEVLEPLHAMMER




mutant
GPQTLKETSFNQAYGRDLME





AQEWCRKYMKSGNVKDLLQA





WDLYYHVFRRISKTS








E2032L,
ILWHEMWHEGLLEASRLYFG
130



T2098L
ERNVKGMFEVLEPLHAMMER




mutant
GPQTLKETSFNQAYGRDLME





AQEWCRKYMKSGNVKDLLQA





WDLYYHVFRRISKTS










Split CAR

In some embodiments, the CAR-expressing cell uses a split CAR. The split CAR approach is described in more detail in publications WO2014/055442 and WO2014/055657. Briefly, a split CAR system comprises a cell expressing a first CAR having a first antigen binding domain and a costimulatory domain (e.g., 41BB), and the cell also expresses a second CAR having a second antigen binding domain and an intracellular signaling domain (e.g., CD3 zeta). When the cell encounters the first antigen, the costimulatory domain is activated, and the cell proliferates. When the cell encounters the second antigen, the intracellular signaling domain is activated and cell-killing activity begins. Thus, the CAR-expressing cell is only fully activated in the presence of both antigens.


CD22 Inhibitors and Binding Domains

As used herein, the term “CD22,” refers to an antigenic determinant known to be detectable on leukemia precursor cells. The human and murine amino acid and nucleic acid sequences can be found in a public database, such as GenBank, UniProt and Swiss-Prot. For example, the amino acid sequences of isoforms 1-5 human CD22 can be found at Accession Nos. NP 001762.2, NP 001172028.1, NP 001172029.1, NP 001172030.1, and NP 001265346.1, respectively, and the nucleotide sequence encoding variants 1-5 of the human CD22 can be found at Accession No. NM 001771.3, NM 001185099.1, NM 001185100.1, NM 001185101.1, and NM 001278417.1, respectively. As used herein, “CD22” includes proteins comprising mutations, e.g., point mutations, fragments, insertions, deletions and splice variants of full length wild-type CD22. In one aspect the antigen-binding portion of the CAR recognizes and binds an antigen within the extracellular domain of the CD22 protein. In one aspect, the CD22 protein is expressed on a cancer cell.


In some aspects, the present disclosure provides a CD22 inhibitor or binding domain, e.g., a CD22 inhibitor or binding domain as described herein. The disclosure also provides a nucleic acid encoding the CD22 binding domain, or a CAR comprising the CD22 binding domain. A CD22 inhibitor includes but is not limited to a CD22 CAR-expressing cell, e.g., a CD22 CART cell or an anti-CD22 antibody (e.g., an anti-CD22 mono- or bispecific antibody) or a fragment thereof. The composition may also comprise a second agent, e.g., an anti-CD19 CAR-expressing cell or a CD19 binding domain. The agents may be, e.g., encoded by a single nucleic acid or different nucleic acids.


In some aspects, a CD22 inhibitor or binding domain is administered as a monotherapy. In some aspects, the CD22 inhibitor or binding domain is administered in combination with a second agent such as an anti-CD19 CAR-expressing cell. In an embodiment, the CD22 inhibitor is administered in combination with a CD19 inhibitor, e.g., a CD19 CAR-expressing cell, e.g., a CAR-expressing cell described herein e.g., a cell expressing a CAR comprising an antibody binding domain that is murine, human, or humanized.


CD22 CAR-Expressing Cells, e.g., CARTs


In one embodiment, the CD22 inhibitor is a CD22 CAR-expressing cell, e.g., a CD22-CAR that comprises a CD22 binding domain and is engineered into a cell (e.g., T cell or NK cell) for administration in combination with CD19 CAR-expressing cell, e.g., CART, and methods of their use for adoptive therapy.


In another aspect, the present invention provides a population of CAR-expressing cells, e.g., CART cells, comprising a mixture of cells expressing CD19 CARs and CD22 CARs. For example, in one embodiment, the population of CART cells can include a first cell expressing a CD19 CAR and a second cell expressing a CD22 CAR. In one embodiment, the population of CAR-expressing cells includes, e.g., a first cell expressing a CAR (e.g., a CD19 CAR or CD22 CAR) that includes a primary intracellular signaling domain, and a second cell expressing a CAR (e.g., a CD19 CAR or CD22 CAR) that includes a secondary signaling domain.


In one aspect, the CD22-CAR comprises an optional leader sequence (e.g., an optional leader sequence described herein), an extracellular antigen binding domain, a hinge (e.g., hinge described herein), a transmembrane domain (e.g., transmembrane domain described herein), and an intracellular stimulatory domain (e.g., intracellular stimulatory domain described herein). In one aspect an exemplary CD22 CAR construct comprises an optional leader sequence (e.g., a leader sequence described herein), an extracellular antigen binding domain, a hinge, a transmembrane domain, an intracellular costimulatory domain (e.g., an intracellular costimulatory domain described herein) and an intracellular stimulatory domain.


In one aspect, the CAR22 binding domain comprises the scFv portion of an amino acid sequence (or encoded by a nucleotide sequence) provided in Table 6 or an amino acid with at least 95%, 96%, 97%, 98%, 99% or more identity thereto. In an embodiment, the CAR22 binding domain comprises the scFv portion provided in any of SEQ ID NOs: 835-837, 542, 567, 592, 617, 642, 667, 692, 717, 742 763, or an an amino acid with at least 95%, 96%, 97%, 98%, 99% or more identity thereto. In an embodiment, the CAR22 binding domain comprises the scFv portion provided in any of SEQ ID NOs: 835-837, 542, 567, 592, 617, 642, 667, 692, 717, 742 or 763.


In specific aspects, a CAR construct of the invention comprises a scFv domain selected from the group consisting of SEQ ID NOs: 835-837, 542, 567, 592, 617, 642, 667, 692, 717, 742 or 763, wherein the scFv may be preceded by an optional leader sequence such as provided in SEQ ID NO: 13, and followed by an optional hinge sequence such as provided in SEQ ID NO:14 or SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49, a transmembrane region such as provided in SEQ ID NO:15, an intracellular signalling domain that includes SEQ ID NO:16 or SEQ ID NO:51 and a CD3 zeta sequence that includes SEQ ID NO:17 or SEQ ID NO:43, e.g., wherein the domains are contiguous with and in the same reading frame to form a single fusion protein. In some embodiments, the scFv domain is a human scFv domain selected from the group consisting of SEQ ID NOs: 835-837, 542, 567, 592, 617, 642, 667, 692, 717, 742 or 763. In some embodiments, the scFv domain is a murine scFv domain, e.g., comprising SEQ ID NO: 1006 or 1332 and/or SEQ ID NO: 1007 or 1333, or a sequence with at least 95% identity thereto.


Also included in the invention is a nucleotide sequence that encodes the polypeptide of each of the scFv fragments depicted in Table 6. In some embodiments, a nucleotide sequence that encodes the polypeptide of the scFv fragment selected from the group consisting of SEQ ID NOs: 835-837, 542, 567, 592, 617, 642, 667, 692, 717, 742 or 763, or a nucleotide sequence with at least 95%, 96%, 97%, 98%, or 99% identity thereto. Also included in the invention is a nucleotide sequence that encodes the polypeptide of each of the scFv fragments selected from the group consisting of SEQ ID NOs: 835-837, 542, 567, 592, 617, 642, 667, 692, 717, 742 or 763, and each of the domains of SEQ ID NOS: 13-17, plus the encoded CD22 CAR of the invention.


In one aspect, the present invention encompasses a recombinant nucleic acid construct comprising a nucleic acid molecule encoding a CAR, wherein the nucleic acid molecule comprises the nucleic acid sequence encoding a CD22 binding domain, e.g., described herein, e.g., that is contiguous with and in the same reading frame as a nucleic acid sequence encoding an intracellular signaling domain. In one aspect, a CD22 binding domain is selected from a CD22 light chain variable domain (VL), CD22 heavy chain variable domain (VH), or a CD22 scFv listed in Table 6, e.g., one or more of SEQ ID NOS: 835-840, 528, 539, 542, 553, 564, 567, 578, 589, 592, 603, 614, 617, 628, 639, 642, 653, 664, 667, 678, 689, 692, 703, 714, 717, 728, 739, 742, 752, 760, 763, 1006 or 1332 or 1007 or 1333. In one aspect, the present invention encompasses a recombinant nucleic acid construct comprising a nucleic acid molecule encoding a CAR, wherein the nucleic acid molecule comprises a nucleic acid sequence encoding a CD22 binding domain, e.g., wherein the sequence is contiguous with and in the same reading frame as the nucleic acid sequence encoding an intracellular signaling domain. An exemplary intracellular signaling domain that can be used in the CAR includes, but is not limited to, one or more intracellular signaling domains of, e.g., CD3-zeta, CD28, 4-1BB, and the like. In some instances, the CAR can comprise any combination of CD3-zeta, CD28, 4-1BB, and the like.


In one aspect, the nucleic acid sequence of a CAR construct that binds CD22 of the invention comprises the CAR construct of one or more of SEQ ID NOS: 529, 540, 543, 554, 565, 568, 579, 590, 593, 604, 615, 618, 629, 640, 643, 654, 665, 668, 679, 690, 693, 704, 715, 718, 729, 740, 743, 745, 753, or 761, or a nucleic acid sequence listed in Table 6, or a nucleic acid sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto.


In one aspect, the nucleic acid sequence of a CAR construct of the invention comprises an scFv-encoding sequence of one or more of SEQ ID NOs: 835-837, 542, 567, 592, 617, 642, 667, 692, 717, 742 or 763.


In some instances, it is beneficial for the antigen binding domain to be derived from the same species in which the CAR will ultimately be used in. For example, for use in humans, it may be beneficial for the antigen binding domain of the CAR to comprise human or humanized residues for the antigen binding domain of an antibody or antibody fragment. Thus, in one aspect, the antigen binding domain comprises a human antibody or an antibody fragment. In one embodiment, the human CD22 binding domain comprises one or more (e.g., all three) light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of a human CD22 binding domain described herein, and/or one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of a human CD22 binding domain described herein, e.g., a human CD22 binding domain comprising one or more, e.g., all three, LC CDRs and one or more, e.g., all three, HC CDRs. In one embodiment, the human CD22 binding domain comprises one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of a human CD22 binding domain described herein, e.g., the human CD22 binding domain has two variable heavy chain regions, each comprising a HC CDR1, a HC CDR2 and a HC CDR3 described herein. In one embodiment, the human CD22 binding domain comprises a human light chain variable region described herein (e.g., in Table 6, or 10) and/or a human heavy chain variable region described herein (e.g., in Table 6 or 9). In one embodiment, the human CD22 binding domain comprises a human heavy chain variable region described herein (e.g., in Table 6 or 9), e.g., at least two human heavy chain variable regions described herein (e.g., in Table 6 or 9). In one embodiment, the CD22 binding domain is a scFv comprising a light chain and a heavy chain of an amino acid sequence of Table 6, 9 or 10. In an embodiment, the CD22 binding domain (e.g., an scFv) comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 6 or 10, or a sequence with at least 95% identity with an amino acid sequence of Table 6 or 10; and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 6 or 9, or a sequence with at least 95% identity to an amino acid sequence of Table 6 or 9. In one embodiment, the human CD22 binding domain comprises a sequence selected from a group consisting of SEQ ID NOS: 835-840, 528, 539, 542, 553, 564, 567, 578, 589, 592, 603, 614, 617, 628, 639, 642, 653, 664, 667, 678, 689, 692, 703, 714, 717, 728, 739, 742, 752, 760, 763, 1006 or 1332 or 1007 or 1333, or a sequence with at least 95% identity thereof. In one embodiment, the human CD22 binding domain is a scFv, and a light chain variable region comprising an amino acid sequence described herein, e.g., in Table 6 or 10, is attached to a heavy chain variable region comprising an amino acid sequence described herein, e.g., in Table 6 or 9, via a linker, e.g., a linker described herein. In one embodiment, the human CD22 binding domain includes a (Gly4-Ser)n linker, wherein n is 1, 2, 3, 4, 5, or 6, e.g., 3 or 4 (SEQ ID NO:53). The light chain variable region and heavy chain variable region of a scFv can be, e.g., in any of the following orientations: light chain variable region-linker-heavy chain variable region or heavy chain variable region-linker-light chain variable region.


In one aspect, the CD22 binding domain is characterized by particular functional features or properties of an antibody or antibody fragment. For example, in one aspect, the portion of a CAR composition of the invention that comprises an antigen binding domain specifically binds human CD22 or a fragment thereof. In one aspect, the invention relates to an antigen binding domain comprising an antibody or antibody fragment, wherein the antibody binding domain specifically binds to a CD22 protein or fragment thereof, wherein the antibody or antibody fragment comprises a variable heavy chain that includes an amino acid sequence of any of SEQ ID NO:s 839, 528, 553, 578, 603, 628, 653, 678, 703, 728, 752, 1332 and/or a variable light chain that includes an amino acid sequence of any of SEQ ID NOs 840, 539, 564, 589, 614, 639, 664, 689, 714, 739, 760 or 1333. In certain aspects, the scFv is contiguous with and in the same reading frame as a leader sequence. In one aspect the leader sequence is the polypeptide sequence provided as SEQ ID NO:13.


In embodiments, the CAR comprises an antibody or antibody fragment which includes a CD22 binding domain, a transmembrane domain, and an intracellular signaling domain. In embodiments, the CD22 binding domain comprises one or more of light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of any CD22 light chain binding domain amino acid sequence listed in Table 8 or 10, and one or more of heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of any CD22 heavy chain binding domain amino acid sequence listed in Table 7 or 9.


In one aspect, the CD22 binding domain is a fragment, e.g., a single chain variable fragment (scFv). In one aspect, the CD22 binding domain is a Fv, a Fab, a (Fab′)2, or a bi-functional (e.g. bi-specific) hybrid antibody (e.g., Lanzavecchia et al., Eur. J. Immunol. 17, 105 (1987)). In one aspect, the antibodies and fragments thereof of the invention binds a CD22 protein or a fragment thereof with wild-type or enhanced affinity.


In some instances, a human scFv can be derived from a display library.


In one embodiment, the CD22 binding domain, e.g., scFv comprises at least one mutation such that the mutated scFv confers improved stability to the CART22 construct. In another embodiment, the CD22 binding domain, e.g., scFv comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 mutations arising, e.g., from the humanization process such that the mutated scFv confers improved stability to the CART22 construct.


In one aspect, the present invention contemplates modifications of the starting antibody or fragment (e.g., scFv) amino acid sequence that generate functionally equivalent molecules. For example, the VH or VL of a CD22 binding domain, e.g., scFv, comprised in the CAR can be modified to retain at least about 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity of the starting VH or VL framework region of the CD22 binding domain, e.g., scFv. The present invention contemplates modifications of the entire CAR construct, e.g., modifications in one or more amino acid sequences of the various domains of the CAR construct in order to generate functionally equivalent molecules. The CAR construct can be modified to retain at least about 70%, 71%. 72%. 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% identity of the starting CAR construct.


In an embodiment, the CD22 binding domain comprises six CDRs (e.g., one each of a HC CDR1, HC CDR2, HC CDR3, LC CDR1, LC CDR2, and LC CDR3) of any one of CD22-65s, CD22-65ss, CD22-65sKD, CD22-65, CD22-57, CD22-58, CD22-59, CD22-60, CD22-61, CD22-62, CD22-63, CD22-64, or CAR22 M971 (e.g., as described in Table 6, 7 or 8), or a sequence substantially identical thereto. In an embodiment, the CD22 binding domain comprises three CDRs (e.g., one each of a HC CDR1, HC CDR2, and HC CDR3, or one each of a LC CDR1, LC CDR2, and LC CDR3) of any one of CD22-65s, CD22-65ss, CD22-65sKD, CD22-65, CD22-57, CD22-58, CD22-59, CD22-60, CD22-61, CD22-62, CD22-63, CD22-64 or CAR22 M971 (e.g., as described in Table 6, 7 or 8), or a sequence substantially identical thereto.


Further embodiments include a nucleotide sequence that encodes a polypeptide described in this section. For example, further embodiments include a nucleotide sequence that encodes a polypeptide of any of Tables 6-10. For instance, the nucleotide sequence can comprise a CAR construct or scFv of Table 6. The nucleotide may encode a VH of Table 9, a VL or Table 10, or both. The nucleotide may encode one or more of (e.g., two or three of) a VH CDR1, VH CDR2, or VH CDR3 of Table 7 and/or the nucleotide may encode one or more of (e.g., two or three of) a VL CDR1, VL CDR2, or VL CDR3 of Table 8. The nucleotide sequence can also include one or more of, e.g., all of the domains of SEQ ID NOS: 13, 14, 15, 16, 17, and 51.


The CD22 CAR may also comprise one or more of a a transmembrane domain, e.g., a transmembrane domain as described herein, an intracellular signaling domain, e.g., intracellular signaling domain as described herein, a costimulatory domain, e.g., a costimulatory domain as described herein, a leader sequence, e.g. a leader sequence as described herein, or a hinge, e.g., a hinge as described herein.


In one embodiment, the CD22 inhibitor is a CD22 inhibitor described herein. The CD22 inhibitor can be, e.g., an anti-CD22 antibody (e.g., an anti-CD22 mono- or bispecific antibody), a small molecule, or a CD22 CART. In some embodiments the anti-CD22 antibody is conjugated or otherwise bound to a therapeutic agent. Exemplary therapeutic agents include, e.g., microtubule disrupting agents (e.g., monomethyl auristatin E) and toxins (e.g., diphtheria toxin or Pseudomonas exotoxin-A, ricin). In an embodiment, the CD22 inhibitor is administered in combination with a CD19 inhibitor, e.g., a CD19 CAR-expressing cell, e.g., a CAR-expressing cell described herein e.g., a cell expressing a CAR comprising an antibody binding domain that is murine, human, or humanized.


In one embodiment, the anti-CD22 antibody is selected from an anti-CD19/CD22 bispecific ligand-directed toxin (e.g., two scFv ligands, recognizing human CD19 and CD22, linked to the first 389 amino acids of diphtheria toxin (DT), DT 390, e.g., DT2219ARL); anti-CD22 monoclonal antibody-MMAE conjugate (e.g., DCDT2980S); scFv of an anti-CD22 antibody RFB4 fused to a fragment of Pseudomonas exotoxin-A (e.g., BL22); deglycosylated ricin A chain-conjugated anti-CD19/anti-CD22 (e.g., Combotox); humanized anti-CD22 monoclonal antibody (e.g., epratuzumab); or the Fv portion of an anti-CD22 antibody covalently fused to a 38 KDa fragment of Pseudomonas exotoxin-A (e.g., moxetumomab pasudotox).


In one embodiment, the anti-CD22 antibody is an anti-CD19/CD22 bispecific ligand-directed toxin (e.g., DT2219ARL) and the anti-CD19/CD22 bispecific ligand-directed toxin is administered at a dose of about 1 μg/kg, 2 μg/kg, 3 μg/kg, 4 μg/kg, 5 μg/kg, 6 μg/kg, 7 μg/kg, 8 μg/kg, 9 μg/kg, 10 μg/kg, 11 μg/kg, 12 μg/kg, 13 μg/kg, 14 μg/kg, 15 μg/kg, 20 μg/kg, 25 μg/kg, 30 μg/kg, 40 μg/kg, 60 μg/kg, 80 μg/kg, 100 μg/kg, 120 μg/kg, 140 μg/kg, 160 μg/kg, 180 μg/kg, 200 μg/kg, 220 μg/kg, 250 μg/kg, 300 μg/kg, 350 μg/kg, 400 μg/kg, 450 μg/kg, 500 μg/kg, 600 μg/kg, 700 μg/kg, 800 μg/kg, 900 μg/kg, 1 mg·kg (e.g., 30 μg/kg, 40 μg/kg, 60 μg/kg, or 80 μg/kg) for a period of time, e.g., every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more days. In some embodiments, the anti-CD19/CD22 bispecific ligand-directed toxin is administered via intravenous infusion.


In one embodiment, the anti-CD22 antibody is BL22 and BL22 is administered at a dose of about 1 μg/kg, 2 μg/kg, 3 μg/kg, 4 μg/kg, 5 μg/kg, 6 μg/kg, 7 μg/kg, 8 μg/kg, 9 μg/kg, 10 μg/kg, 11 μg/kg, 12 μg/kg, 13 μg/kg, 14 μg/kg, 15 μg/kg, 20 μg/kg, 25 μg/kg, 30 μg/kg, 40 μg/kg, 60 μg/kg, 80 μg/kg, 100 μg/kg, 120 μg/kg, 140 μg/kg, 160 μg/kg, 180 μg/kg, 200 μg/kg, 220 μg/kg, 250 μg/kg, 300 μg/kg, 350 μg/kg, 400 μg/kg, 450 μg/kg, 500 μg/kg, 600 μg/kg, 700 μg/kg, 800 μg/kg, 900 μg/kg, 1 mg·kg (e.g., 3 μg/kg, 30 μg/kg, 40 μg/kg, or 50 μg/kg) for a period of time, e.g., every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more days. In some embodiments, BL22 is administered daily, every other day, every third, day, or every fourth day for a period of time, e.g., for a 4 day cycle, a 6 day cycle, an 8 day cycle, a 10 day cycle, a 12 day cycle, or a 14 day cycle. In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more cycles of BL22 are administered. In some embodiments, BL22 is administered via intravenous infusion.


In one embodiment, the anti-CD22 antibody is a deglycosylated ricin A chain-conjugated anti-CD19/anti-CD22 (e.g., Combotox) and the deglycosylated ricin A chain-conjugated anti-CD19/anti-CD22 is administered at a dose of about 500 μg/m2, 600 μg/m2, 700 μg/m2, 800 μg/m2, 900 μg/m2, 1 mg/m2, 2 mg/m2, 3 mg/m2, 4 mg/m2, 5 mg/m2, 6 mg/m2, or 7 mg/m2 for a period of time, e.g., every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 or more days. In some embodiments, the deglycosylated ricin A chain-conjugated anti-CD19/anti-CD22 is administered daily, every other day, every third, day, or every fourth day for a period of time, e.g., for a 4 day cycle, a 6 day cycle, an 8 day cycle, a 10 day cycle, a 12 day cycle, or a 14 day cycle (e.g., every other day for 6 days). In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more cycles of the deglycosylated ricin A chain-conjugated anti-CD19/anti-CD22 are administered. In some embodiments, the deglycosylated ricin A chain-conjugated anti-CD19/anti-CD22 is administered via intravenous infusion.


In one embodiment, the anti-CD22 antibody is a humanized anti-CD22 monoclonal antibody (e.g., epratuzumab) and the humanized anti-CD22 monoclonal antibody is administered at a dose of about 10 mg/m2/week, 20 mg/m2/week, 50 mg/m2/week, 100 mg/m2/week, 120 mg/m2/week, 140 mg/m2/week, 160 mg/m2/week, 180 mg/m2/week, 200 mg/m2/week, 220 mg/m2/week, 250 mg/m2/week, 260 mg/m2/week, 270 mg/m2/week, 280 mg/m2/week, 290 mg/m2/week, 300 mg/m2/week, 305 mg/m2/week, 310 mg/m2/week, 320 mg/m2/week, 325 mg/m2/week, 330 mg/m2/week, 335 mg/m2/week, 340 mg/m2/week, 345 mg/m2/week, 350 mg/m2/week, 355 mg/m2/week, 360 mg/m2/week, 365 mg/m2/week, 370 mg/m2/week, 375 mg/m2/week, 380 mg/m2/week, 385 mg/m2/week, 390 mg/m2/week, 400 mg/m2/week, 410 mg/m2/week, 420 mg/m2/week, 430 mg/m2/week, 440 mg/m2/week, 450 mg/m2/week, 460 mg/m2/week, 470 mg/m2/week, 480 mg/m2/week, 490 mg/m2/week, 500 mg/m2/week, 600 mg/m2/week, 700 mg/m2/week, 800 mg/m2/week, 900 mg/m2/week, 1 g/m2/week, or 2 g/m2/week (e.g., 360 mg/m2/week or 480 mg/m2/week) for a period of time, e.g., every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more weeks. In some embodiments a first dose is lower than subsequent doses (e.g. a first dose of 360 mg/m2/week followed by subsequent doses of 370 mg/m2/week). In some embodiments, the humanized anti-CD22 monoclonal antibody is administered via intravenous infusion.


In one embodiment, the anti-CD22 antibody is moxetumomab pasudotox and moxetumomab pasudotox is administered at a dose of about 1 μg/kg, 2 μg/kg, 3 μg/kg, 4 μg/kg, 5 μg/kg, 6 μg/kg, 7 μg/kg, 8 μg/kg, 9 μg/kg, 10 μg/kg, 11 μg/kg, 12 μg/kg, 13 μg/kg, 14 μg/kg, 15 μg/kg, 20 μg/kg, 25 μg/kg, 30 μg/kg, 40 μg/kg, 60 μg/kg, 80 μg/kg, 100 μg/kg, 120 μg/kg, 140 μg/kg, 160 μg/kg, 180 μg/kg, 200 μg/kg, 220 μg/kg, 250 μg/kg, 300 μg/kg, 350 μg/kg, 400 μg/kg, 450 μg/kg, 500 μg/kg (e.g., 5 μg/kg, 10 μg/kg, 20 μg/kg, 30 μg/kg, 40 μg/kg, or 50 μg/kg) a period of time, e.g., every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more days. In some embodiments, the moxetumomab pasudotox is administered daily, every other day, every third, day, or every fourth day for a period of time, e.g., for a 4 day cycle, a 6 day cycle, an 8 day cycle, a 10 day cycle, a 12 day cycle, or a 14 day cycle (e.g., every other day for 6 days). In one embodiment, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more cycles of the moxetumomab pasudotox are administered. In some embodiments, the moxetumomab pasudotox is administered via intravenous infusion.


In an embodiment, a CD22 antibody molecule comprises six CDRs (e.g., one each of a HC CDR1, HC CDR2, HC CDR3, LC CDR1, LC CDR2, and LC CDR3) of any one of CD22-65s, CD22-65ss, CD22-65sKD, CD22-65, CD22-57, CD22-58, CD22-59, CD22-60, CD22-61, CD22-62, CD22-63, CD22-64 or CAR22 M971(e.g., as described in Table 6, 7 or 8), or a sequence substantially identical thereto. In an embodiment, a CD22 antibody molecule comprises three CDRs (e.g., one each of a HC CDR1, HC CDR2, and HC CDR3, or one each of a LC CDR1, LC CDR2, and LC CDR3) of any one of CD22-65s, CD22-65ss, CD22-65sKD, CD22-65, CD22-57, CD22-58, CD22-59, CD22-60, CD22-61, CD22-62, CD22-63, CD22-64, or CAR22 M971 (e.g., as described in Table 6, 7 or 8), or a sequence substantially identical thereto. In an embodiment, a CD22 antibody molecule comprises a heavy chain variable region, a light chain variable region, or both of a heavy chain variable region and light chain variable region, or an scFv, as described in Table 6, or a sequence substantially identical thereto. In embodiments, the CD22 antibody molecule is an isolated antibody molecule.


CAR22 Constructs: Design and Function

Fully human anti-CD22 single chain variable fragments were isolated. Anti-CD22 ScFvs were cloned into lentiviral CAR expression vectors with the CD3zeta chain and the 4-1BB costimulatory molecule. CAR-containing plasmids were amplified by bacterial transformation in STBL3 cells, followed by Maxiprep using endotoxin-free Qiagen Plasmid Maki kit. Lentiviral supernatant was produced in 293T cells using standard techniques.


The sequences of the human CARs are provided below in Table 6.


These clones all contained a Q/K residue change in the signal domain of the co-stimulatory domain derived from CD3zeta chain.









TABLE 6 







CD22 CAR Constructs









SEQ ID




NUMBER
Ab region
Sequence





CD22-65s,




ss or KD




SEQ ID NO: 834
Linker
GGGGS





CD22-65S

EVQLQQSGPGLVKPS


SEQ ID NO: 835
scFv (VH-
QTLSLTCAISGDSML



linker-VL) of
SNSDTWNWIRQS



CD22-65s
PSRGLEWLGRTYHRS



(linker shown
TWYDDYASSVRGRVS



by italics
INVDTSRNQYSLQLN



and
AVTPEDTGVYYCARV



underline)
RLQDGNSWSDAFDVW




GQGTMVTVSSGGGGS




QSALTQPASASGSPG




QSVTISCTGTSSDVG




GYNYVSWYQQHPGRA




PRLMIYDVSNRPSGV




SNRFSGSRSGNTASL




TISGLQAEDEADYYC




SSYTSSSTLYVFGTG




TQLTVL





CD22-65ss
scFv (VH-
EVQLQQSGPGLVRPS


SEQ ID NO: 836
VL) of CD22-
QTLSLTCAISGDSML



65ss
SNSDTWNWIRQSPSR



(no linker
GLEWLGRTYHRSTWY



between VH-
DDYASSVRGRVSINV



VL)
DTSRNQYSLQLNAVT




PEDTGVYYCARVRLQ




DGNSWSDAFDVWGQG




TMVTVSSQSALTQPA




SASGSPGQSVTISCT




GTSSDVGGYNYVSWY




QQHPGRAPRLMIYDV




SNRPSGVSNRISGSR




SGNTASLTISGLQAE




DEADYYCSSYTSSST




LYVFGTGTQLTVL





CD22-65sKD
scFv (VH-
EVQLQQSGPGLVKPS


SEQ ID NO: 837
linker-VL) of
QTLSLTCAISGDSML



CD22-65 sRD
SNSDTWNWIRKSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARVRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSQSA




LTOPASASGSPGQSV




TISCTGTSSDVGGYN




YVSWYQDHPGKAPKL




MIYDVSNRPSGVSNR




FSGSKSGNTASLTIS




GLQAEDEADYYCSSY




TSSSTLYVFGTGTQL




TVL





SEQ ID NO: 839
VH of CD22-
EVQLQQSGPGLVRPS



65 sKD
QTLSLTCAISGDSML




SNSDTWNWIRRSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSRNQYSLQLNAVT




PEDTGVYYCARVRLQ




DGNSWSDAFDVWGQG




TMVTVSS





SEQ ID NO: 840
VL of CD22-
QSALTQPASASGSPG



65 sKD
QSVTISCTGTSSDVG




GYNYVSWYQDHPGRA




PRLMIYDVSNRPSGV




SNRFSGSRSGNTASL




TISGLQAEDEADYYC




SSYTSSSTLYVFGTG




TQLTVL





CD22-57




SEQ ID NO: 519
HCDR1
NNNAAWN


(Rabat)







SEQ ID NO: 520
HCDR2
RTYHRSTWYNDYVGS


(Rabat)

VRS





SEQ ID NO: 521
HCDR3
ETDYGDYGAFDI


(Rabat)







SEQ ID NO: 522
HCDR1
GDSVSNNNA


(Chothia)







SEQ ID NO: 523
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 524
HCDR3
ETDYGDYGAFDI


(Chothia)







SEQ ID NO: 525
HCDR1
GDSVSNNNAA


(IMGT)







SEQ ID NO: 526
HCDR2
TYHRSTWYN


(IMGT)







SEQ ID NO: 527
HCDR3
ARETDYGDYGAFDI


(IMGT)







SEQ ID NO: 988
HCDR1
GDSVSNNNAAWN


(Combined




Chothia




and Rabat)







SEQ ID NO: 989
HCDR2
RTYHRSTWYNDYVGS


(Combined

VRS


Chothia




and Rabat)







SEQ ID NO: 990
HCDR3
ETDYGDYGAFDI


(Combined




Chothia




and Rabat)







SEQ ID NO: 528
VH
EVQLQQSGPGLVRPS




QTLSLTCAISGDSVS




NNNAAWNWIRQSPSR




GLEWLGRTYHRSTWY




NDYVGSVRSRITINP




DTSRNQFSLQLNSVT




PEDTAVYYCARETDY




GDYGAFDIWGQGTTV




TVSS





SEQ ID NO: 529
DNA VH
GAAGTCCAATTGCAA




CAATCAGGTCCCGGA




CTCGTGAAACCTTCC




CAAACCCTCTCCCTC




ACTTGCGCGATCAGC




GGAGACTCCGTGTCC




AACAACAATGCTGCC




TGGAACTGGATTAGG




CAGAGCCCTTCAAGA




GGACTGGAATGGCTG




GGACGGACTTACCAC




CGCTCCACCTGGTAC




AACGATTACGTGGGG




TCCGTCAAGTCCCGG




ATCACCATTAACCCG




GACACTTCCAAGAAT




CAGTTCAGCCTGCAA




CTTAACAGCGTGACT




CCCGAGGATACCGCC




GTGTACTACTGTGCC




CGGGAAACCGACTAC




GGGGATTACGGAGCC




TTCGACATCTGGGGA




CAGGGAACCACCGTG




ACCGTGTCCTCG





SEQ ID NO: 530
LCDR1
TGSRNDIGAYESVS


(Rabat)







SEQ ID NO: 531
LCDR2
GVNNRPS


(Rabat)







SEQ ID NO: 532
LCDR3
SSHTTTSTLYV


(Rabat)







SEQ ID NO: 533
LCDR1
SRNDIGAYES


(Chothia)







SEQ ID NO: 534
LCDR2
GVN


(Chothia)







SEQ ID NO: 535
LCDR3
HTTTSTLY


(Chothia)







SEQ ID NO: 536
LCDR1
RNDIGAYES


(IMGT)







SEQ ID NO: 537
LCDR2
GVN


(IMGT)







SEQ ID NO: 538
LCDR3
SSHTTTSTLYV


(IMGT)







SEQ ID NO: 991
LCDR1
TGSRNDIGAYESVS


(Combined




Chothia




and Rabat)







SEQ ID NO: 992
LCDR2
GVNNRPS


(Combined




Chothia




and Rabat)







SEQ ID NO: 993
LCDR3
SSHTTTSTLYV


(Combined




Chothia




and Rabat)







SEQ ID NO: 539
VL
QSALTQPASVSGSPG




QSITISCTGSRNDIG




AYESVSWYQQHPGNA




PRLIIHGVNNRPSGV




FDRFSVSQSGNTASL




TISGLQAEDEADYYC




SSHTTTSTLYVFGTG




TRVTVL





SEQ ID NO: 540
DNA VL
CAGTCGGCCCTGACT




CAGCCGGCCTCCGTG




TCCGGAAGCCCGGGC




CAGTCCATCACCATT




TCGTGCACTGGGTCG




CGCAACGACATCGGC




GCCTACGAATCCGTG




TCGTGGTACCAGCAG




CACCCCGGCAACGCC




CCGAAGCTGATCATC




CATGGCGTCAACAAC




AGACCATCCGGAGTG




TTCGACCGGTTCAGC




GTGTCCCAGTCGGGA




AACACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAG




GCTGACTATTACTGC




TCCTCACACACCACC




ACCTCTACGCTCTAT




GTGTTTGGGACTGGC




ACCAAGGTCACAGTG




CTGGGA





SEQ ID NO: 541
Linker
GGGGSGGGGSGGGGS


SEQ ID NO: 542
scFv (VH-
EVQLQQSGPGLVRPS



linker-VL)
QTLSLTCAISGDSVS




NNNAAWNWIRQS




PSRGLEWLGRTY




HRSTWYNDYVGSVRS




RITINPDTSRNQFSL




QLNSVTPEDTAVYYC




ARETDYGDYGAFDIW




GQGTTVTVSSGGGGS




GGGGSGGGGSQSALT




QPASVSGSPGQSITI




SCTGSRNDI




GAYESVSWYQQHPGN




APKLIIHGVNNRPSG




VFDRFSVSQSGNTAS




LTISGLQAEDEADYY




CSSHTTTSTLYVFGT




GTKVTVL





SEQ ID NO: 543
DNA scFv
GAAGTCCAATTGCAA



(VH-Iinker-
CAATCAGGTCCCGGA



VL)
CTCGTGAAACCTTCC




CAAACCCTCTCCCTC




ACTTGCGCGATCAGC




GGAGACTCCGTGTCC




AACAACAATGCTGCC




TGGAACTGGATTAGG




CAGAGCCCTTCAAGA




GGACTGGAATGGCTG




GGACGGACTTACCAC




CGCTCCACCTGGTAC




AACGATTACGTGGGG




TCCGTCAAGTCCCGG




ATCACCATTAACCCG




GACACTTCCAAGAAT




CAGTTCAGCCTGCAA




CTTAACAGCGTGACT




CCCGAGGATACCGCC




GTGTACTACTGTGCC




CGGGAAACCGACTAC




GGGGATTACGGAGCC




TTCGACATCTGGGGA




CAGGGAACCACCGTG




ACCGTGTCCTCGGGC




GGTGGTGGTTCGGGC




GGCGGGGGATCAGGG




GGCGGAGGAAGCCAG




TCGGCCCTGACTCAG




CCGGCCTCCGTGTCC




GGAAGCCCGGGCCAG




TCCATCACCATTTCG




TGCACTGGGTCGCGC




AACGACATCGGCGCC




TACGAATCCGTGTCG




TGGTACCAGCAGCAC




CCCGGCAACGCCCCG




AAGCTGATCATCCAT




GGCGTCAACAACAGA




CCATCCGGAGTGTTC




GACCGGTTCAGCGTG




TCCCAGTCGGGAAAC




ACCGCATCCCTGACC




ATTAGCGGCCTGCAG




GCGGAGGACGAGGCT




GACTATTACTGCTCC




TCACACACCACCACC




TCTACGCTCTATGTG




TTTGGGACTGGCACC




AAGGTCACAGTGCTG




GGA





CD22-58




SEQ ID NO: 544
HCDR1
SNSAAWN


(Kabat)







SEQ ID NO: 545
HCDR2
RTFYRSKWYNDYAVS


(Kabat)

VKG





SEQ ID NO: 546
HCDR3
GDYYYGLDV


(Kabat)







SEQ ID NO: 547
HCDR1
GDSVSSNSA


(Chothia)







SEQ ID NO: 548
HCDR2
FYRSKWY


(Chothia)







SEQ ID NO: 549
HCDR3
GDYYYGLDV


(Chothia)







SEQ ID NO: 550
HCDR1
GDSVSSNSAA


(IMGT)







SEQ ID NO: 551
HCDR2
TFYRSKWYN


(IMGT)







SEQ ID NO: 552
HCDR3
AGGDYYYGLDV


(IMGT)







SEQ ID NO: 994
HCDR1
GDSVSSNSAAWN


(Combined




Chothia




and Kabat)







SEQ ID NO: 995
HCDR2
RTFYRSKWYNDYAVS


(Combined

VKG


Chothia




and Kabat)







SEQ ID NO: 996
HCDR3
GDYYYGLDV


(Combined




Chothia




and Kabat)







SEQ ID NO: 553
VH
EVQLQQSGPGLVNPS




QTLSITCAISGDSVS




SNSAAWNWIRQSPSR




GLEWLGRTFYRSKWY




NDYAVSVKGRITISP




DTSKNQFSLQLNSVT




PEDTAVYYCAGGDYY




YGLDVWGQGTTVTVS




S





SEQ ID NO: 554
DNA VH
GAAGTCCAGTTGCAA




CAGTCAGGTCCCGGC




CTCGTCAACCCATCC




CAAACCCTTTCCATT




ACCTGTGCCATTAGC




GGGGACAGCGTGTCC




TCCAACTCGGCCGCT




TGGAACTGGATCAGA




CAGAGCCCCAGCCGG




GGTCTGGAGTGGCTG




GGACGGACCTTCTAC




CGCTCAAAGTGGTAC




AACGACTACGCGGTG




TCCGTGAAGGGAAGG




ATTACCATCTCCCCG




GATACATCGAAGAAT




CAGTTCTCCCTGCAA




CTGAACTCTGTGACC




CCTGAGGATACCGCC




GTGTACTACTGCGCG




GGAGGAGACTACTAC




TATGGGCTGGACGTC




TGGGGCCAGGGAACC




ACCGTGACTGTGTCA




AGC





SEQ ID NO: 555
LCDR1
TGSSSDVGGYNSVS


(Kabat)







SEQ ID NO: 556
LCDR2
EVINRPS


(Kabat)







SEQ ID NO: 557
LCDR3
SSYTSSSTYV


(Kabat)







SEQ ID NO: 558
LCDR1
SSSDVGGYNS


(Chothia)







SEQ ID NO: 559
LCDR2
EVI


(Chothia)







SEQ ID NO: 560
LCDR3
YTSSSTY


(Chothia)







SEQ ID NO: 561
LCDR1
SSDVGGYNS


(IMGT)







SEQ ID NO: 562
LCDR2
EVI


(IMGT)







SEQ ID NO: 563
LCDR3
SSYTSSSTYV


(IMGT)







SEQ ID NO: 997
LCDR1
TGSSSDVGGYNSVS


(Combined




Chothia




and Kabat)







SEQ ID NO: 998
LCDR2
EVINRPS


(Combined




Chothia




and Kabat)







SEQ ID NO: 999
LCDR3
SSYTSSSTYV


(Combined




Chothia




and Kabat)







SEQ ID NO: 564
VL
QSALTQPASVSGSPG




QSITISCTGSSSDVG




GYNSVSWYQQHPGKA




PKLMIYEVINRPSGV




SHRFSGSKSGNTASL




TISGLQAEDEADYYC




SSYTSSSTYVFGTGT




KVTVL





SEQ ID NO: 565
DNA VL
CAGAGCGCCCTGACC




CAGCCGGCCAGCGTG




TCCGGGTCGCCGGGC




CAGTCGATCACCATC




AGCTGCACTGGGTCA




TCCTCCGACGTGGGA




GGCTACAACTCCGTG




TCGTGGTACCAGCAG




CACCCGGGGAAGGCT




CCTAAGCTGATGATC




TACGAAGTGATCAAC




CGGCCCTCCGGAGTC




TCGCATCGCTTITCC




GGTTCAAAGTCCGGA




AACACGGCCTCCCTG




ACCATCTCCGGACTC




CAAGCCGAGGATGAA




GCAGACTATTACTGC




TCCTCGTACACTAGC




TCATCCACTTACGTG




TTCGGAACTGGCACC




AAAGTCACTGTGCTC





SEQ ID NO: 566
Linker
GGGGSGGGGSGGGGS





SEQ ID NO: 567
scFv (VH-
EVQLQQSGPGLVNPS



linker-VL)
QTLSITCAISGDSVS




SNSAAWNWIRQSPSR




GLEWLGRTFYRSKWY




NDYAVSVKGRITISP




DTSKNQFSLQLNSVT




PEDTAVYYCAGGDYY




YGLDVWGQGTTVTVS




SGGGGSGGGGSGGGG




SQSALTQPASVSGSP




GQSITISCTGSSSDV




GGYNSVSWYQQHPGK




APKLMIYEVINRPSG




VSHRFSGSKSGNTAS




LTISGLQAEDEADYY




CSSYTSSSTYVFGTG




TKVTVL





SEQ ID NO: 568
DNA scFv
GAAGTCCAGTTGCAA



(VH-linker-
CAGTCAGGTCCCGGC



VL)
CTCGTCAACCCATCC




CAAACCCTTTCCATT




ACCTGTGCCATTAGC




GGGGACAGCGTGTCC




TCCAACTCGGCCGCT




TGGAACTGGATCAGA




CAGAGCCCCAGCCGG




GGTCTGGAGTGGCTG




GGACGGACCTTCTAC




CGCTCAAAGTGGTAC




AACGACTACGCGGTG




TCCGTGAAGGGAAGG




ATTACCATCTCCCCG




GATACATCGAAGAATC




AGTTCTCCCTGCAACT




GAACTCTGTGACCCCT




GAGGATACCGCCGTG




TACTACTGCGCGGGA




GGAGACTACTACTAT




GGGCTGGACGTCTGG




GGCCAGGGAACCACC




GTGACTGTGTCAAGC




GGAGGGGGCGGCTCC




GGTGGAGGAGGCTCG




GGTGGCGGCGGAAGC




CAGAGCGCCCTGACC




CAGCCGGCCAGCGTG




TCCGGGTCGCCGGGC




CAGTCGATCACCATC




AGCTGCACTGGGTCA




TCCTCCGACGTGGGA




GGCTACAACTCCGTG




TCGTGGTACCAGCAG




CACCCGGGGAAGGCT




CCTAAGCTGATGATC




TACGAAGTGATCAAC




CGGCCCTCCGGAGTC




TCGCATCGCTTITCC




GGTTCAAAGTCCGGA




AACACGGCCTCCCTG




ACCATCTCCGGACTC




CAAGCCGAGGATGAA




GCAGACTATTACTGC




TCCTCGTACACTAGC




TCATCCACTTACGTG




TTCGGAACTGGCACC




AAAGTCACTGTGCTC





CD22-59




SEQ ID NO: 569
HCDR1
SNSDTWN


(Kabat)







SEQ ID NO: 570
HCDR2
RTYHRSTWYDDYASS


(Kabat)

VRG





SEQ ID NO: 571
HCDR3
DRLQDGNSWSDAFDV


(Kabat)







SEQ ID NO: 572
HCDR1
GDSVLSNSD


(Chothia)







SEQ ID NO: 573
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 574
HCDR3
DRLQDGNSWSDAFDV


(Chothia)







SEQ ID NO: 575
HCDR1
GDSVLSNSDT


(IMGT)







SEQ ID NO: 576
HCDR2
TYHRSTWYD


(IMGT)







SEQ ID NO: 577
HCDR3
ARDRLQDGNSWSDAF


(IMGT)

DV





SEQ ID NO: 1000
HCDR1
GDSVLSNSDTWN


(Combined




Chothia




and Kabat)







SEQ ID NO: 1001
HCDR2
RTYHRSTWYDDYASS


(Combined

VRG


Chothia




and Kabat)







SEQ ID NO: 1002
HCDR3
DRLQDGNSWSDAFDV


(Combined




Chothia




and Kabat)







SEQ ID NO: 578
VH
EVQLQQSGPGLVKPS




QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSI




NVDTSKNQYSLQLNA




VTPEDTGVYYCARDR




LQDGNSWSDAFDVWG




QGTMVTVSS





SEQ ID NO: 579
DNA VH
GAAGTCCAATTGCAA




CAGTCCGGTCCTGGC




CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGAAACAGCTGG




TCCGATGCCTTCGAT




GTCTGGGGACAGGGT




ACCATGGTCACAGTG




TCCAGC





SEQ ID NO: 580
LCDR1
TGSSSDIGGFNYVS


(Kabat)







SEQ ID NO: 581
LCDR2
EVTNRPS


(Kabat)







SEQ ID NO: 582
LCDR3
SSYASGSPLYV


(Kabat)







SEQ ID NO: 583
LCDR1
SSSDIGGFNY


(Chothia)







SEQ ID NO: 584
LCDR2
EVT


(Chothia)







SEQ ID NO: 585
LCDR3
YASGSPLY


(Chothia)







SEQ ID NO: 586
LCDR1
SSD1GGFNY


(IMGT)







SEQ ID NO: 587
LCDR2
EVT


(IMGT)







SEQ ID NO: 588
LCDR3
SSYASGSPLYV


(IMGT)







SEQ ID NO: 1003
LCDR1
TGSSSDIGGFNYVS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1004
LCDR2
EVTNRPS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1005
LCDR3
SSYASGSPLYV


(Combined




Chothia




and Kabat)







SEQ ID NO: 589
VL
QSALTQPASVSGSPG




QSITISCTGSSSDIG




GFNYVSWYQQHAGEA




PKLMIYEVTNRPSGV




SDRESGSKSDNTASL




TISGLQAEDEADYYC




SSYASGSPLYVFGTG




TKVTVL





SEQ ID NO: 590
DNA VL
CAGTCCGCGCTGACC




CAGCCCGCCTCTGTG




TCCGGATCACCGGGA




CAGTCGATCACGATC




TCCTGCACTGGCTCA




TCGTOCGACATTGGA




GGTTTTAACTACGTG




TCGTGGTACCAGCAG




CATGCAGGAGAAGCC




CCGAAGCTCATGATC




TACGAAGTGACCAAC




CGGCCTTCGGGGGTG




TCAGACAGATTCTCG




GGCTCCAAGTCCGAC




AATACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAA




GCCGACTACTATTGC




TCCTCGTACGCTTCG




GGCTCCCCTCTGTAC




GTGTTCGGCACTGGG




ACCAAAGTCACCGTG




CTC





SEQ ID NO: 591
Linker
GGGGSGGGGSGGGGS





SEQ ID NO: 592
scFv (VH-
EVQLQQSGPGLVKPS



linker-VL)
QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSGGG




GSGGGGSQSALTQPA




SVSGSPGQSITISCT




GSSSDIGGFNYVSWY




QQHAGEAPKLMIYEV




TNRPSGVSDRFSGSK




SDNTASLTISGLQAE




DEADYYCSSYASGSP




LYVFGTGTKVTVL





SEQ ID NO: 593
DNA scEv
GAAGTCCAATTGCAA



(VH-linker-
CAGTCCGGTCCTGGC



VL)
CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTG




CGCGGACGCGTGTCA




ATCAATGTGGACACC




TCCAAGAACCAGTAC




AGCCTGCAACTTAAC




GCTGTGACTCCCGAG




GATACTGGAGTGTAC




TATTGTGCCCGCGAC




CGGCTGCAGGATGGA




AACAGCTGGTCCGAT




GCCTTCGATGTCTGG




GGACAGGGTACCATG




GTCACAGTGTCCAGC




GGGGGGGGCGGATCA




GGCGGCGGTGGCTCC




GGAGGAGGGGGTTCC




CAGTCCGCGCTGACC




CAGCCCGCCTCTGTG




TCCGGATCACCGGGA




CAGTCGATCACGATC




TCCTGCACTGGCTCA




TCGTCCGACATTGGA




GGTTTTAACTACGTG




TCGTGGTACCAGCAG




CATGCAGGAGAAGCC




CCGAAGCTCATGATC




TACGAAGTGACCAAC




CGGCCTTCGGGGGTG




TCAGACAGATTCTCG




GGCTCCAAGTCCGAC




AATACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAA




GCCGACTACTATTGC




TCCTCGTACGCTTCG




GGCTCCCCTCTGTAC




GTGTTCGGCACTGGG




ACCAAAGTCACCGTG




CTC





CD22-60




SEQ ID NO: 594
HCDR1
SNSDTWN


(Kabat)







SEQ ID NO: 595
HCDR2
RTYHRSTWYDDYASS


(Kabat)

VRG





SEQ ID NO: 596
HCDR3
DRLQDGNSWSDAFDV


(Kabat)







SEQ ID NO: 597
HCDR1
GDSVLSNSD


(Chothia)







SEQ ID NO: 598
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 599
HCDR3
DRLQDGNSWSDAFDV


(Chothia)







SEQ ID NO: 600
HCDR1
GDSVLSNSDT


(IMGT)







SEQ ID NO: 601
HCDR2
TYHRSTWYD


(IMGT)







SEQ ID NO: 602
HCDR3
ARDRLQDGNSWSDAF


(IMGT)

DV





SEQ ID NO: 1006
HCDR1
GDSVLSNSDTWN


(Combined Chothia




and Kabat)







SEQ ID NO: 1007
HCDR2
RTYHRSTWYDDYASS


(Combined

VRG


Chothia




and Kabat)







SEQ ID NO: 1008
HCDR3
DRLQDGNSWSDAFDV


(Combined 




Chothia




and Kabat)







SEQ ID NO: 603
VH
EVQLQQSGPGLVKPS




QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAFDVWGQG




TMVTVSS





SEQ ID NO: 604
DNA VH
GAAGTCCAATTGCAA




CAGTCCGGTCCTGGC




CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGAAACAGCTGG




TCCGATGCCTTCGAT




GTCTGGGGACAGGGT




ACCATGGTCACAGTG




TCCAGC





SEQ ID NO: 605
LCDR1
TGTSSDIGGYNYVS


(Kabat)







SEQ ID NO: 606
LCDR2
EVSNRPS


(Kabat)







SEQ ID NO: 607
LCDR3
SSYTSSSTLYV


(Kabat)







SEQ ID NO: 608
LCDR1
TSSDIGGYNY


(Chothia)







SEQ ID NO: 609
LCDR2
EVS


(Chothia)







SEQ ID NO: 610
LCDR3
YTSSSTLY


(Chothia)







SEQ ID NO: 611
LCDR1
SSDIGGYNY


(IMGT)







SEQ ID NO: 612
LCDR2
EVS


(IMGT)







SEQ ID NO: 613
LCDR3
SSYTSSSTLYV


(IMGT)







SEQ ID NO: 1009
LCDR1
TGTSSDIGGYNYVS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1010
LCDR2
EVSNRPS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1011
LCDR3
SSYTSSSTLYV


(Combined




Chothia




and Kabat)







SEQ ID NO: 614
VL
QSALTQPASVSGSPG




QSITFSCTGTSSDIG




GYNYVSWYQQHPG




KAPKLMIYEVSNRPS




GVSNRFSGTKSGNTA




SLTISGLQAEDEA




DYYCSSYTSSSTLYV




FGTGTKLTVL





SEQ ID NO: 615
DNA VL
CAGTCCGCGCTGACC




CAGCCCGCCTCTGTG




TCCGGATCACCGGGA




CAGTCGATCACGTTT




TCCTGCACTGGCACC




TCGTCCGACATCGGA




GGTTACAACTACGTG




TCGTGGTACCAGCAG




CATCCAGGAAAGGCC




CCGAAGCTCATGATC




TACGAAGTGTCAAAC




CGGCCTTCGGGGGTG




TCAAACAGATTCTCG




GGCACCAAGTCCGGA




AATACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAA




GCCGACTACTATTGC




TCCTCGTACACCTCG




AGCTCCACTCTGTAC




GTGTTCGGCACTGGG




ACCAAACTTACCGTG




CTC





SEQ ID NO: 616
Linker
GGGGSGGGGSGSGGS





SEQ ID NO: 617
scFv (VH-
EVQLQQSGPGLVKPS



linker-VL)
QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSGGG




GSGSGGSQSALTQPA




SVSGSPGQSITFSCT




GTSSDIGGYNYVSWY




QQHPGKAPKLMIYEV




SNRPSGVSNRFSGTK




SGNTASLTISGLQAE




DEADYYCSSYTSSST




LYVFGTGTKLTVL





SEQ ID NO: 618
DNA scFv
GAAGTCCAATTGCAA



(VH-linker-
CAGTCCGGTCCTGGC



VL)
CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGA




AACAGCTGGTCCGAT




GCCTTCGATGTCTGG




GGACAGGGTACCATG




GTCACAGTGTCCAGC




GGGGGGGGCGGATCA




GGCGGCGGTGGCTCC




GGATCGGGGGGTTCC




CAGTCCGCGCTGACC




CAGCCCGCCTCTGTG




TCCGGATCACCGGGA




CAGTCGATCACGTTT




TCCTGCACTGGCACC




TCGTCCGACATCGGA




GGTTACAACTACGTG




TCGTGGTACCAGCAG




CATCCAGGAAAGGCC




CCGAAGCTCATGATC




TACGAAGTGTCAAAC




CGGCCTTCGGGGGTG




TCAAACAGATTCTCG




GGCACCAAGTCCGGA




AATACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAA




GCCGACTACTATTGC




TCCTCGTACACCTCG




AGCTCCACTCTGTAC




GTGTTCGGCACTGGG




ACCAAACTTACCGTG




CTC





CD22-61




SEQ ID NO: 619
HCDR1
SNSDTWN


(Kabat)







SEQ ID NO: 620
HCDR2
RTYHRSTWYDDYASS


(Kabat)

VRG





SEQ ID NO: 621
HCDR3
DRLQDGNSWSDAFDV


(Kabat)







SEQ ID NO: 622
HCDR1
GDSVLSNSD


(Chothia)







SEQ ID NO: 623
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 624
HCDR3
DRLQDGNSWSDAFDV


(Chothia)







SEQ ID NO: 625
HCDR1
GDSVLSNSDT


(IMGT)







SEQ ID NO: 626
HCDR2
TYHRSTWYD


(IMGT)







SEQ ID NO: 627
HCDR3
ARDRLQDGNSWSDAF


(IMGT)

DV





SEQ ID NO: 1012
HCDR1
GDSVLSNSDTWN


(Combined




Chothia




and Kabat)







SEQ ID NO: 1013
HCDR2
RTYHRSTWYDDYASS


(Combined

VRG


Chothia




and Kabat)







SEQ ID NO: 1014
HCDR3
DRLQDGNSWSDAFDV


(Combined




Chothia




and Kabat)







SEQ ID NO: 628
VH
QVQLQESGPGLVKPS




QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAEDVWGQG




TMVTVSS





SEQ ID NO: 629
DNA VH
CAAGTCCAATTGCAA




GAATCCGGTCCTGGC




CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGAAACAGCTGG




TCCGATGCCTTCGAT




GTCTGGGGACAGGGT




ACCATGGTCACAGTG




TCCAGC





SEQ ID NO: 630
LCDR1
TGTSSDVGGYNYVS


(Kabat)







SEQ ID NO: 631
LCDR2
EVSNRPS


(Kabat)







SEQ ID NO: 632
LCDR3
SSYTSSSTLYV


(Kabat)







SEQ ID NO: 633
LCDR1
TSSDVGGYNY


(Chothia)







SEQ ID NO: 634
LCDR2
EVS


(Chothia)







SEQ ID NO: 635
LCDR3
YTSSSTLY


(Chothia)







SEQ ID NO: 636
LCDR1
SSDVGGYNY


(IMGT)







SEQ ID NO: 637
LCDR2
EVS


(IMGT)







SEQ ID NO: 638
LCDR3
SSYTSSSTLYV


(IMGT)







SEQ ID NO: 1015
LCDR1
TGTSSDVGGYNYVS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1016
LCDR2
EVSNRPS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1017
LCDR3
SSYTSSSTLYV


(Combined




Chothia




and Kabat)







SEQ ID NO: 639
VL
QSALTQPASVSGSPG




QSITISCTGTSSDVG




GYNYVSWYQQHPGKA




PKLMIYEVSNRPSGV




SNRESGSKSGNTASL




TISGLQAEDEADYYC




SSYTSSSTLYVFGTG




TKVTVL





SEQ ID NO: 640
DNA VL
CAGTCCGCGCTGACC




CAGCCCGCCTCTGTG




TCCGGATCACCGGGA




CAGTCGATCACGATC




TCCTGCACTGGCACC




TCGTCCGACGTGGGA




GGTTACAACTACGTG




TCGTGGTACCAGCAG




CATCCAGGAAAGGCC




CCGAAGCTCATGATC




TACGAAGTGTCAAAC




CGGCCTTCGGGGGTG




TCAAACAGATTCTCG




GGCTCCAAGTCCGGA




AATACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAA




GCCGACTACTATTGC




TCCTCGTACACCTCG




AGCTCCACTCTGTAC




GTGTTCGGCACTGGG




ACCAAAGTCACCGTG




CTC





SEQ ID NO: 641
Linker
GGGGSGGGGSGSGGS





SEQ ID NO: 642
scFv (VH-
QVQLQESGPGLVKPS



linker-VL)
QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSGGG




GSGSGGSQSALTQPA




SVSGSPGQSITISCT




GTSSDVGGYNYVSWY




QQHPGKAPKLMIYEV




SNRPSGVSNRFSGSK




SGNTASLTISGLQAE




DEADYYCSSYTSSST




LYVFGTGTKVTVL





SEQ ID NO: 643
DNA scFv
CAAGTCCAATTGCAAG



(VH-linker-
AATCCGGTCCTGGCC



VL)
TCGTCAAGCCCTCCC




AAACCCTCTCCCTGA




CTTGCGCCATCTCCG




GGGATTCCGTGCTGA




GCAACTCCGACACCT




GGAACTGGATTCGGC




AGAGCCCGTCCAGAG




GCCTGGAGTGGCTGG




GCAGGACCTACCACC




GGAGCACTTGGTACG




ACGACTACGCCAGCT




CCGTGCGCGGACGCG




TGTCAATCAATGTGG




ACACCTCCAAGAACC




AGTACAGCCTGCAAC




TTAACGCTGTGACTC




CCGAGGATACTGGAG




TGTACTATTGTGCCC




GCGACCGGCTGCAGG




ATGGAAACAGCTGGT




CCGATGCCTTCGATG




TCTGGGGACAGGGTA




CCATGGTCACAGTGT




CCAGCGGGGGGGGCG




GATCAGGCGGCGGTG




GCTCCGGATCGGGGG




GTTCCCAGTCCGCGC




TGACCCAGCCCGCCT




CTGTGTCCGGATCAC




CGGGACAGTCGATCA




CGATCTCCTGCACTG




GCACCTCGTCCGACG




TGGGAGGTTACAACT




ACGTGTCGTGGTACC




AGCAGCATCCAGGAA




AGGCCCCGAAGCTCA




TGATCTACGAAGTGT




CAAACCGGCCTTCGG




GGGTGTCAAACAGAT




TCTCGGGCTCCAAGT




CCGGAAATACCGCAT




CCCTGACCAITAGCG




GCCTGCAGGCGGAGG




ACGAAGCCGACTACT




ATTGCTCCTCGTACA




CCTCGAGCTCCACTC




TGTACGTGTTCGGCA




CTGGGACCAAAGTCA




CCGTGCTC





SEQ ID NO: 644
HCDR1
SNSDTWN


(Kabat)







SEQ ID NO: 645
HCDR2
RTYHRSTWYDDYASS


(Kabat)

VRG





SEQ ID NO: 646
HCDR3
DRLQDGNSWSDAFDV


(Kabat)







SEQ ID NO: 647
HCDR1
GDSVLSNSD


(Chothia)







SEQ ID NO: 648
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 649
HCDR3
DRLQDGNSWSDAFDV


(Chothia)







SEQ ID NO: 650
HCDR1
GDSVLSNSDT


(IMGT)







SEQ ID NO: 651
HCDR2
TYHRSTWYD


(IMGT)







SEQ ID NO: 652
HCDR3
ARDRLQDGNSWSDAF


(IMGT)

DV





SEQ ID NO: 1018
HCDR1
GDSVLSNSDTWN


(Combined




Chothia




and Kabat)







SEQ ID NO: 1019
HCDR2
RTYHRSTWYDDYASS


(Combined

VRG


Chothia




and Kabat)







SEQ ID NO: 1020
HCDR3
DRLQDGNSWSDAFDV


(Combined




Chothia




and Kabat)







SEQ ID NO: 653
VH
EVQLQQSGPGLVKPS




QTLSLTCAISGDSVL




SNSDTWNWIRQSP




SRGLEWLGRTYHRST




WYDDYASSVRGRVSI




NVDTSKNQYSLQLNA




VTPEDTGVYYCARDR




LQDGNSWSDAFDVWG




QGTMVTVSS





SEQ ID NO: 654
DNA VH
GAAGTCCAATTGCAA




CAGTCCGGTCCTGGC




CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGAAACAGCTGG




TCCGATGCCTTCGAT




GTCTGGGGACAGGGT




ACCATGGTCACAGTG




TCCAGC





SEQ ID NO: 655
LCDR1
TGTSSDVGGYNYVS


(Kabat)







SEQ ID NO: 656
LCDR2
DVSNRPS


(Kabat)







SEQ ID NO: 657
LCDR3
SSYTSSSTLYV


(Kabat)







SEQ ID NO: 658
LCDR1
TSSDVGGYNY


(Chothia)







SEQ ID NO: 659
LCDR2
DVS


(Chothia)







SEQ ID NO: 660
LCDR3
YTSSSTLY


(Chothia)







SEQ ID NO: 661
LCDR1
SSDVGGYNY


(IMGT)







SEQ ID NO: 662
LCDR2
DVS


(IMGT)







SEQ ID NO: 663
LCDR3
SSYTSSSTLYV


(IMGT)







SEQ ID NO: 1021
LCDR1
TGTSSDVGGYNYVS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1022
LCDR2
DVSNRPS


(Combined Chothia




and Kabat)







SEQ ID NO: 1023
LCDR3
SSYTSSSTLYV


(Combined Chothia




and Kabat)







SEQ ID NO: 664
VL
QSALTQPASVSGSPG




QSITISCTGTSSDVG




GYNYVSWYQQHPGKA




PKLMIYDVSNRPSGV




SNRFSGSKSGNTASL




TISGLQAEDEADYYC




SSYTSSSTLYVFGTG




TKVTVL





SEQ ID NO: 665
DNA VL
CAGTCCGCGCTGACC




CAGCCCGCCTCTGTG




TCCGGATCACCGGGA




CAGTCGATCACGATC




TCCTGCACTGGCACC




TCGTCCGACGTGGGA




GGTTACAACTACGTG




TCGTGGTACCAGCAG




CATCCAGGAAAGGCC




CCGAAGCTCATGATC




TACGACGTGTCAAAC




CGGCCTTCGGGGGTG




TCAAACAGATTCTCG




GGCTCCAAGTCCGGA




AATACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAA




GCCGACTACTAITGC




TCCTCGTACACCTCG




AGCTCCACTCTGTAC




GTGTTCGGCACTGGG




ACCAAAGTCACCGTG




CTC





SEQ ID NO: 666
Linker
GGGGSGGGGSGGGGS





SEQ ID NO: 667
scFv (VH-
EVQLQQSGPGLVKPS



linker-VL)
QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSGGG




GSGGGGSQSALTQPA




SVSGSPGQSITISCT




GTSSDVGGYNYVSWY




QQHPGKAPKLMIYDV




SNRPSGVSNRFSGSK




SGNTASLTISGLQAE




DEADYYCSSYTSSST




LYVFGTGTKVTVL





SEQ ID NO: 668
DNA scFv
GAAGTCCAATTGCAA



(VH-linker-
CAGTCCGGTCCTGGC



VL)
CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGAAACAGCTGG




TCCGATGCCTTCGAT




GTCTGGGGACAGGGT




ACCATGGTCACAGTG




TCCAGCGGGGGGGGC




GGATCAGGCGGCGGT




GGCTCCGGAGGAGGG




GGTTCCCAGTCCGCG




CTGACCCAGCCCGCC




TCTGTGTCCGGATCA




CCGGGACAGTCGATC




ACGATCTCCTGCACT




GGCACCTCGTCCGAC




GTGGGAGGTTACAAC




TACGTGTCGTGGTAC




CAGCAGCATCCAGGA




AAGGCCC




CGAAGCTCATGATCT




ACGACGTGTCAAACC




GGCCTTCGGGGGTGT




CAAACAGATTCTCGG




GCTCCAAGTCCGGAA




ATACCGCATCCCTGA




CCATTAGCGGCCTGC




AGGCGGAGGACGAAG




CCGACTACTATTGCT




CCTCGTACACCTCGA




GCTCCACTCTGTACG




TGTTCGGCACTGGGA




CCAAAGTCACCGTGC




TC





CD22-63




SEQ ID NO: 669
HCDR1
SNSDTWN


(Kabat)







SEQ ID NO: 670
HCDR2
RTYHRSTWYDDYASS


(Kabat)

VRG





SEQ ID NO: 671
HCDR3
DRLQDGNSWSDAFDV


(Kabat)







SEQ ID NO: 672
HCDR1
GDSVLSNSD


(Chothia)







SEQ ID NO: 673
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 674
HCDR3
DRLQDGNSWSDAFDV


(Chothia)







SEQ ID NO: 675
HCDR1
GDSVLSNSDT


(IMGT)







SEQ ID NO: 676
HCDR2
TYHRSTWYD


(IMGT)







SEQ ID NO: 677
HCDR3
ARDRLQDGNSWSDAF


(IMGT)

DV





SEQ ID NO: 1024
HCDR1
GDSVLSNSDTWN


(Combined Chothia




and Kabat)







SEQ ID NO: 1025
HCDR2
RTYHRSTWYDDYASS


(Combined Chothia

VRG


and Kabat)







SEQ ID NO: 1026
HCDR3
DRLQDGNSWSDAFDV


(Combined Chothia




and Kabat)







SEQ ID NO: 678
VH
EVQLQQSGPGLVKPS




QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAFDVWGQG




TMVTVSS





SEQ ID NO: 679
DNA VH
GAAGTCCAATTGCAA




CAGTCCGGTCCTGGC




CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACTCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGAAACAGCTGG




TCCGATGCCTTCGAT




GTCTGGGGACAGGGT




ACCATGGTCACAGTG




TCCAGC





SEQ ID NO: 680
LCDR1
TGTSSDVGGYNYVS


(Kabat)







SEQ ID NO: 681
LCDR2
EVSNRPS


(Kabat)







SEQ ID NO: 682
LCDR3
SSYTSSSTLYI


(Kabat)







SEQ ID NO: 683
LCDR1
TSSDVGGYNY


(Chothia)







SEQ ID NO: 684
LCDR2
EVS


(Chothia)







SEQ ID NO: 685
LCDR3
YTSSSTLY


(Chothia)







SEQ ID NO: 686
LCDR1
SSDVGGYNY


(IMGT)







SEQ ID NO: 687
LCDR2
EVS


(IMGT)







SEQ ID NO: 688
LCDR3
SSYTSSSTLYI


(IMGT)







SEQ ID NO: 1027
LCDR1
TGTSSDVGGYNYVS


(Combined Chothia




and Kabat)







SEQ ID NO: 1028
LCDR2
EVSNRPS


(Combined Chothia




and Kabat)







SEQ ID NO: 1029
LCDR3
SSYTSSSTLYI


(Combined Chothia




and Kabat)







SEQ ID NO: 689
VL
QSALTQPASVSGSPG




QSITISCTGTSSDVG




GYNYVSWYQQHPGKA




PKLMIYEVSNRPSGV




SNRFSGSKSGNTASL




TISGLQAEDEADYYC




SSYTSSSTLYIFGTG




TKVTVL





SEQ ID NO: 690
DNA VL
CAGTCCGCGCTGACC




CAGCCCGCCTCTGTG




TCCGGATCACCGGGA




CAGTCGATCACGATC




TCCTGCACTGGCACC




TCGTCCGACGTGGGA




GGTTACAACTACGTG




TCGTGGTACCAGCAG




CATCCAGGAAAGGCC




CCGAAGCTCATGATC




TACGAAGTGTCAAAC




CGGCCTTCGGGGGTG




TCAAACAGATTCTCG




GGCTCCAAGTCCGGA




AATACCGCATCCCTG




ACCATTAGCGGCCTG




CAGGCGGAGGACGAA




GCCGACTACTATTGC




TCCTCGTACACCTCG




AGCTCCACTCTGTAC




ATTTTCGGCACTGGG




ACCAAAGTCACCGTG




CTC





SEQ ID NO: 691
Linker
GGGGSGGGGSGGGGS





SEQ ID NO: 692
scFv (VH-
EVQLQQSGPGLVKPS



linker-VL)
QTLSLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARDRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSGGG




GSGGGGSQSALTQPA




SVSGSPGQSITISCT




GTSSDVGGYNYVSWY




QQHPGKAPKLMIYEV




SNRPSGVSNRFSGSK




SGNTASLTISGLQAE




DEADYYCSSYTSSST




LYIFGTGTKVTVL





SEQ ID NO: 693
DNA scFv
GAAGTCCAATTGCAA



(VH-linker-
CAGTCCGGTCCTGGC



VL)
CTCGTCAAGCCCTCC




CAAACCCTCTCCCTG




ACTTGCGCCATCTCC




GGGGATTCCGTGCTG




AGCAACFCCGACACC




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGCCTGGAGTGGCTG




GGCAGGACCTACCAC




CGGAGCACTTGGTAC




GACGACTACGCCAGC




TCCGTGCGCGGACGC




GTGTCAATCAATGTG




GACACCTCCAAGAAC




CAGTACAGCCTGCAA




CTTAACGCTGTGACT




CCCGAGGATACTGGA




GTGTACTATTGTGCC




CGCGACCGGCTGCAG




GATGGAAACAGCTGG




TCCGATGCCTTCGAT




GTCTGGGGACAGGGT




ACCATGGTCACAGTG




TCCAGCGGGGGGGGC




GGATCAGGCGGCGGT




GGCTCCGGAGGAGGG




GGTTCCCAGTCCGCG




CTGACCCAGCCCGCC




TCTGTGTCCGGATCA




CCGGGACAGTCGATC




ACGATCTCCTGCACT




GGCACCTCGTCCGAC




GTGGGAGGTTACAAC




TACGTGTCGTGGTAC




CAGCAGCATCCAGGA




AAGGCCCCGAAGCTC




ATGATCTACGAAGTG




TCAAACCGGCCTTCG




GGGGTGTCAAACAGA




TTCTCGGGCTCCAAG




TCCGGAAATACCGCA




TCCCTGACCATTAGC




GGCCTGCAGGCGGAG




GACGAAGCCGACTAC




TATTGCTCCTCGTAC




ACCTCGAGCTCCACT




CTGTACATTTTCGGC




ACTGGGACCAAAGTC




ACCGTGCTC





CD22-64




SEQ ID NO: 694
HCDR1
SNSDTWN


(Kabat)







SEQ ID NO: 695
HCDR2
RTYHRSTWYDDYASS


(Kabat)

VRG





SEQ ID NO: 696
HCDR3
VRLQDGNSWSDAFDV


(Kabat)







SEQ ID NO: 697
HCDR1
GDSVLSNSD


(Chothia)







SEQ ID NO: 698
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 699
HCDR3
VRLQDGNSWSDAFDV


(Chothia)







SEQ ID NO: 700
HCDR1
GDSVLSNSDT


(IMGT)







SEQ ID NO: 701
HCDR2
TYHRSTWYD


(IMGT)







SEQ ID NO: 702
HCDR3
ARVRLQDGNSWSDAF


(IMGT)

DV





SEQ ID NO: 1030
HCDR1
GDSVLSNSDTWN


(Combined




Chothia




and Kabat)







SEQ ID NO: 1031
HCDR2
RTYHRSTWYDDYASS


(Combined

VRG


Chothia




and Kabat)







SEQ ID NO: 1032
HCDR3
VRLQDGNSWSDAFDV


(Combined




Chothia




and Kabat)







SEQ ID NO: 703
VH
EVQLQQSGPGLVKPS




QTLPLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARVRLQ




DGNSWSDAFDVWGQG




TMVTVSS





SEQ ID NO: 704
DNA VH
GAAGTGCAGCTTCAA




CAATCAGGACCCGGA




CTCGTCAAACCATCG




CAGACCCTCCCTCTC




ACTTGCGCCATCTCC




GGGGACTCCGTGCTG




TCCAACTCCGACACT




TGGAACTGGATTCGG




CA




GAGCCCGTCCAGAGG




ATTGGAATGGCTGGG




AAGGACCTATCACCG




GTCCACTTGGTACGA




CGATTACGCCTCGTC




CGTGCGCGGTCGGGT




GTCCATCAACGTGGA




CACCTCCAAGAACCA




GTACTCCCTGCAACT




GAACGCCGTGACCCC




TGAGGACACTGGGGT




GTACTACTGTGCGAG




AGTGCGGCTGCAGGA




TGGGAACTCTTGGTC




CGACGCCTTCGATGT




CTGGGGCCAGGGCAC




CATGGTCACTGTGTC




ATCC





SEQ ID NO: 705
LCDR1
TGTSSDVGGYNYVS


(Kabat)







SEQ ID NO: 706
LCDR2
DVSNRPS


(Kabat)







SEQ ID NO: 707
LCDR3
SSYTSSSTLYV


(Kabat)







SEQ ID NO: 708
LCDR1
TSSDVGGYNY


(Chothia)







SEQ ID NO: 709
LCDR2
DVS


(Chothia)







SEQ ID NO: 710
LCDR3
YTSSSTLY


(Chothia)







SEQ ID NO: 711
LCDR1
SSDVGGYNY


(IMGT)







SEQ ID NO: 712
LCDR2
DVS


(IMGT)







SEQ ID NO: 713
LCDR3
SSYTSSSTLYV


(IMGT)







SEQ ID NO: 1033
LCDR1
TGTSSDVGGYNYVS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1034
LCDR2
DVSNRPS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1035
LCDR3
SSYTSSSTLYV


(Combined




Chothia




and Kabat)







SEQ ID NO: 714
VL
QSALTQPASASGSPG




QSVTISCTGTSSDVG




GYNYVSWYQQHPGKA




PKLMIYDVSNRPSGV




SNRFSGSKSGNTASL




TISGLQAEDEADYYC




SSYTSSSTLYVFGTG




TQLTVL





SEQ ID NO: 715
DNA VL
CAGTCGGCACTGACC




CAGCCTGCCTCAGCC




TCCGGGAGCCCGGGA




CAGTCCGTGACCATT




TCCTGCACCGGGACC




TCCTCCGACGTGGGA




GGCTACAACTACGTG




TCATGGTACCAGCAG




CACCCCGGAAAGGCA




CCGAAGCTGATGATC




TACGACGTGTCCAAC




CGCCCGAGCGGGGTG




TCAAATCGCTTCTCG




GGCTCGAAGTCGGGA




AACACAGCGAGCCTG




ACGATCTCGGGACTG




CAAGCCGAAGATGAG




GCTGACTACTACTGC




TCGTCCTACACTAGC




TCCAGCACCCTCTAC




GTGTTCGGTACTGGT




ACCCAGCTGACCGTC




CTG





SEQ ID NO: 716
Linker
GGGGSGGGGSGGGGP





SEQ ID NO: 717
scFv (VH-
EVQLQQSGPGLVKPS



linker-VL)
QTLPLTCAISGDSVL




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARVRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSGGG




GSGGGGPQSALTQPA




SASGSPGQSVTISCT




GTSSDVGGYNYVSWY




QQHPGKAPKLMIYDV




SNRPSGVSNRFSGSK




SGNTASLTISGLQAE




DEADYYCSSYTSSST




LYVFGTGTQLTVL





SEQ ID NO: 718
DNA scFv
GAAGTGCAGCTTCAA



(VH-linker-
CAATCAGGACCCGGA



VL)
CTCGTCAAACCATCG




CAGACCCTCCCTCTC




ACTTGCGCCATCTCC




GGGGACTCCGTGCTG




TCCAACTCCGACACT




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGATTGGAATGGCTG




GGAAGGACCTATCAC




CGGTCCACTTGGTAC




GACGATTACGCCTCG




TCCGTGCGCGGTCGG




GTGTCCATCAACGTG




GACACCTCCAAGAAC




CAGTACTCCCTGCAA




CTGAACGCCGTGACC




CCTGAGGACACTGGG




GTGTACTACTGTGCG




AGAGTGCGGCFGCAG




GATGGGAACTCTTGG




TCCGACGCCTTCGAT




GTCTGGGGCCAGGGC




ACCATGGTCACTGTG




TCATCCGGCGGTGGT




GGCAGCGGCGGAGGC




GGCAGCGGAGGCGGA




GGACCCCAGTCGGCA




CTGACCCAGCCTGCC




TCAGCCTCCGGGAGC




CCGGGACAGTCCGTG




ACCATTTCCTGCACC




GGGACCTCCTCCGAC




GTGGGAGGCTACAAC




TACGTGTCATGGTAC




CAGCAGCACCCCGGA




AAGGCACCGAAGCTG




ATGATCTACGACGTG




TCCAACCGCCCGAGC




GGGGTGTCAAATCGC




TTCTCGGGCTCGAAG




TCGGGAAACACAGCG




AGCCTGACGATCTCG




GGACTGCAAGCCGAA




GATGAGGCTGACTAC




TACTGCTCGTCCTAC




ACTAGCTCCAGCACC




CTCTACGTGTTCGGT




ACTGGTACCCAGCTG




ACCGTCCTG





CD22-65




SEQ ID NO: 719
HCDR1
SNSDTWN


(Kabat)







SEQ ID NO: 720
HCDR2
RTYHRSTWYDDYASS


(Kabat)

VRG





SEQ ID NO: 721
HCDR3
VRLQDGNSWSDAFDV


(Kabat)







SEQ ID NO: 722
HCDR1
GDSMLSNSD


(Chothia)







SEQ ID NO: 723
HCDR2
YHRSTWY


(Chothia)







SEQ ID NO: 724
HCDR3
VRLQDGNSWSDAFDV


(Chothia)







SEQ ID NO: 725
HCDR1
GDSMLSNSDT


(IMGT)







SEQ ID NO: 726
HCDR2
TYHRSTWYD


(IMGT)







SEQ ID NO: 727
HCDR3
ARVRLQDGNSWSDAF


(IMGT)

DV





SEQ ID NO: 1036
HCDR1
GDSMLSNSDTWN


(Combined




Chothia




and Kabat)







SEQ ID NO: 1037
HCDR2
RTYHRSTWYDDYASS


(Combined

VRG


Chothia




and Kabat)







SEQ ID NO: 1038
HCDR3
VRLQDGNSWSDAFDV


(Combined




Chothia




and Kabat)







SEQ ID NO: 728
VH
EVQLQQSGPGLVKPS




QTLSLTCAISGDSML




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARVRLQ




DGNSWSDAFDVWGQG




TMVTVSS





SEQ ID NO: 729
DNA VH
GAAGTGCAGCTTCAA




CAATCAGGACCCGGA




CTCGTCAAACCATCG




CAGACCCTCAGCCTC




ACTTGCGCCATCTCC




GGGGACTCCATGCTG




TCCAACTCCGACACT




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGATTGGAATGGCTG




GGAAGGACCTATCAC




CGGTCCACTTGGTAC




GACGATTACGCCTCG




TCCGTGCG




CGGTCGGGTGTCCAT




CAACGTGGACACCTC




CAAGAACCAGTACTC




CCTGCAACTGAACGC




CGTGACCCCTGAGGA




CACTGGGGTGTACTA




CTGTGCGAGAGTGCG




GCTGCAGGATGGGAA




CTCTTGGTCCGACGC




CTTCGATGTCTGGGG




CCAGGGCACCATGGT




CACTGTGTCATCC





SEQ ID NO: 730
LCDR1
TGTSSDVGGYNYVS


(Kabat)







SEQ ID NO: 731
LCDR2
DVSNRPS


(Kabat)







SEQ ID NO: 732
LCDR3
SSYTSSSTLYV


(Kabat)







SEQ ID NO: 733
LCDR1
TSSDVGGYNY


(Chothia)







SEQ ID NO: 734
LCDR2
DVS


(Chothia)







SEQ ID NO: 735
LCDR3
YTSSSTLY


(Chothia)







SEQ ID NO: 736
LCDR1
SSDVGGYNY


(IMGT)







SEQ ID NO: 737
LCDR2
DVS


(IMGT)







SEQ ID NO: 738
LCDR3
SSYTSSSTLYV


(IMGT)







SEQ ID NO: 1039
LCDR1
TGTSSDVGGYNYVS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1040
LCDR2
DVSNRPS


(Combined




Chothia




and Kabat)







SEQ ID NO: 1041
LCDR3
SSYTSSSTLYV


(Combined 




Chothia




and Kabat)







SEQ ID NO: 739
VL
QSALTQPASASGSPG




QSVTISCTGTSSDVG




GYNYVSWYQQHPGKA




PKLMIYDVSNRPSGV




SNRFSGSKSGNTASL




TISGLQAEDEADYYC




SSYTSSSTLYVFGTG




TQLTVL





SEQ ID NO: 740
DNA VL
CAGTCGGCACTGACC




CAGCCTGCCTCAGCC




TCCGGGAGCCCGGGA




CAGTCCGTGACCATT




TCCTGCACCGGGACC




TCCTCC




GACGTGGGAGGCTAC




AACTACGTGTCATGG




TACCAGCAGCACCCC




GGAAAGGCACCGAAG




CTGATGATCTACGAC




GTGTCCAACCGCCCG




AGCGGGGTGTCAAAT




CGCTTCTCGGGCTCG




AAGTCGGGAAACACA




GCGAGCCTGACGATC




TCGGGACTGCAAGCC




GAAGATGAGGCTGAC




TACTACTGCTCGTCC




TACACTAGCTCCAGC




ACCCTCTACGTGTTC




GGTACTGGTACCCAG




CTGACCGTCCTG





SEQ ID NO: 741
Linker
GGGGSGGGGSGGGGS





SEQ ID NO: 742
scFv (VH-
EVQLQQSGPGLVKPS



linker-VL)
QTLSLTCAISGDSML




SNSDTWNWIRQSPSR




GLEWLGRTYHRSTWY




DDYASSVRGRVSINV




DTSKNQYSLQLNAVT




PEDTGVYYCARVRLQ




DGNSWSDAFDVWGQG




TMVTVSSGGGGSGGG




GSGGGGSQSALTQPA




SASGSPGQSVTISCT




GTSSDVGGYNYVSWY




QQHPGKAPKLMIYDV




SNRPSGVSNRFSGSK




SGNTASLTISGLQAE




DEADYYCSSYTSSST




LYVFGTGTQLTVL





SEQ ID NO: 743
DNA scFv
GAAGTGCAGCTTCAA



(VH-linker-
CAATCAGGACCCGGA



VL)
CTCGTCAAACCATCG




CAGACCCTCAGCCTC




ACTTGCGCCATCTCC




GGGGACTCCATGCTG




TCCAACTCCGACACT




TGGAACTGGATTCGG




CAGAGCCCGTCCAGA




GGATTGGAATGGCTG




GGAAGGACCTATCAC




CGGTCCACTTGGTAC




GACGATTACGCCTCG




TCCGTGCGCGGTCGG




GTGTCCATCAACGTG




GACACCTCCAAGAAC




CAGTACTCCCTGCAA




CTGAACGCCGTGACC




CCTGAGGACACTGGG




GTGTACTACTGTGCG




AGAGTGCGGCTGCAG




GATGGGAACTCTTGG




TCCGACGCCTTCGAT




GTCTGGGGCCAGGGC




ACCATGGTCACTGTG




TCATCCGGCGGTGGT




GGCAGCGGCGGAGGC




GGCAGCGGAGGCGGA




GGAAGCCAGTCGGCA




CTGACCCAGCCTGCC




TCAGCCTCCGGGAGC




CCGGGACAGTCCGTG




ACCATTTCCTGCACC




GGGACCTCCTCCGAC




GTGGGAGGCTACAAC




TACGTGTCATGGTAC




CAGCAGCACCCCGGA




AAGGCACCGAAGCTG




ATGATCTACGACGTG




TCCAACCGCCCGAGC




GGGGTGTCAAATCGC




TTCTCGGGCTCGAAG




TCGGGAAACACAGCG




AGCCTGACGATCTCG




GGACTGCAAGCCGAA




GATGAGGCTGACTAC




TACTGCTCGTCCTAC




ACTAGCTCCAGCACC




CTCTACGTGTTCGGT




ACTGGTACCCAGCTG




ACCGTCCTG





SEQ ID NO: 744
Full
MALPVTALLLPLALL



amino
LHAARPEVQLQQSGP



acid 
GLVKPSQTLSLTCAI



sequence
SGDSMLSNSDTWNWI




RQSPSRGLEWLGRTY




HRSTWYDDYASSVRG




RVSINVDTSKNQLQD




GNSWSDAFDVWGQGT




MVTVSSGGGGSGGGG




SGGGGSQSALTQPAS




ASGSPGQSVTISCTG




TSSDVGGYNYVSWYQ




QHPGKAPKLMIYDVS




NRPSGVSNRESGSKS




GNTASLTISGLQAED




EADYYCSSYTSSSTL




YVFGTGTQLTVLTTT




PAPRPPTPAPTIASQ




PLSLRPEACRPAAGG




AVHTRGLDFACDIYI




WAPLAGTCGVLLLSL




VITLYCKRGRKKLLY




IFKQPFMRPVQTTQE




EDGCSCRFPEEEEGG




CELRVKFSRSADAPA




YKQGQNQLYNELNLG




RREEYDVLDKRRGRD




PEMGGKPRRKXPQEG




LYNELQKDKMAEAYS




EIGMKGERRRGKGHD




GLYQGLSTATKDTYD




ALHMQALPPRYSLQL




NAVTPEDTGVYYCAR




VR





SEQ ID NO: 745
Full
ATGGCCCTCCCTGTC



nucleic
ACCGCCCTGCTGCTT



acid
CCGCTGGCTCTTCTG



sequence
CTCCACGCCGCTCGG




CCCGAAGTGCAGCTT




CAACAATCAGGACCC




GGACTCGTCAAACCA




TCGCAGACCCTCAGC




CTCACTTGCGCCATC




TCCGGGGACTCCATG




CTGTCCAACTCCGAC




ACTTGGAACTGGATT




CGGCAGAGCCCGTCC




AGAGGATTGGAATGG




CTGGGAAGGACCTAT




CACCGGTCCACTTGG




TACGACGATTACGCC




TCGTCCGTGCGCGGT




CGGGTGTCCATCAAC




GTGGACACCTCCAAG




AACCAGTACTCCCTG




CAACTGAACGCCGTG




ACCCCTGAGGACACT




GGGGTGTACTACTGT




GCGAGAGTGCGGCTG




CAGGATGGGAACTCT




TGGTCCGACGCCTTC




GATGTCTGGGGCCAG




GGCACCATGGTCACT




GTGTCATCCGGCGGT




GGTGGCAGCGGCGGA




GGCGGCAGCGGAGGC




GGAGGAAGCCAGTCG




GCACTGACCCAGCCT




GCCTCAGCCTCCGGG




AGCCCGGGACAGTCC




GTGACCATTTCCTGC




ACCGGGACCTCCTCC




GACGTGGGAGGCTAC




AACTACGTGTCATGG




TACCAGCAGCACCCC




GGAAAGGCACCGAAG




CTGATGATCTACGAC




GTGTCCAACCGCCCG




AGCGGGGTGTCAAAT




CGCTTCTCGGGCTCG




AAGTCGGGAAACACA




GCGAGCCTGACGATC




TCGGGACTGCAAGCC




GAAGATGAGGCTGAC




TACTACTGCTCGTCC




TACACTAGCTCCAGC




ACCCTCTACGTGTTC




GGTACTGGTACCCAG




CTGACCGTCCTGACC




ACFACCCCAGCACCG




AGGCCACCCACCCCG




GCTCCTACCATCGCC




TCCCAGCCTCTGTCC




CTGCGTCCGGAGGCA




TGTAGACCCGCAGCT




GGTGGGGCCGTGCAT




ACCCGGGGTCTTGAC




TTCGCCTGCGATATC




TACATTTGGGCCCCT




CTGGCTGGTACTTGC




GGGGTCCTGCTGCTT




TCACTCGTGATCACT




CTTTACTGTAAGCGC




GGTCGGAAGAAGCTG




CTGTACATCTTTAAG




CAACCCTTCATGAGG




CCTGTGCAGACTACT




CAAGAGGAGGACGGC




TGITCATGCCGGITC




CCAGAGGAGGAGGAA




GGCGGCTGCGAACTG




CGCGTGAAATTCAGC




CGCAGCGCAGATGCT




CCAGCCTACAAGCAG




GGGCAGAACCAGCTC




TACAACGAACTCAAT




CTTGGTCGGAGAGAG




GAGTACGACGTGCTG




GACAAGCGGAGAGGA




CGGGACCCAGAAATG




GGCGGGAAGCCGCGC




AGAAAGAATCCCCAA




GAGGGCCTGTACAAC




GAGCTCCAAAAGGAT




AAGATGGCAGAAGCC




TATAGCGAGATTGGT




ATGAAAGGGGAACGC




AGAAGAGGCAAAGGC




CACGACGGACTGTAC




CAGGGACTCAGCACC




GCCACCAAGGACACC




TATGACGCTCTTCAC




ATGCAGGCCCTGCCG




CCTCGG





CD22-53




SEQ ID NO: 746
HCDR1
SNSAAWN


(Kabat)







SEQ ID NO: 747
HCDR2
RTYYRSKWYSDYAVS


(Kabat)

VKS





SEQ ID NO: 748
HCDR3
DPYDFWSGYPDAFDI


(Kabat)







SEQ ID NO: 749
HCDR1
GDSVSSNSA


(Chothia)







SEQ ID NO: 750
HCDR2
YYRSKWY


(Chothia)







SEQ ID NO: 751
HCDR3
DPYDFWSGYPDAFDI


(Chothia)







SEQ ID NO: 752
VH
EVQLQQSGPGLVKPS




QTLSLTCAISGDSVS




SNSAAWNWIRQSPSR




GLEWLGRTYYRSKWY




SDYAVSVKSRITINP




DTSKNQFSLQLNSVT




PEDTAVYYCARDPYD




FWSGYPDAFDIWGQG




TMVTVSS





SEQ ID NO: 753
DNA VH
GAGGTACAGCTGCAG




CAGTCAGGTCCAGGA




CTGGTGAAGCCCTCG




CAGACCCTCTCACTC




ACCTGTGCCATCTCC




GGGGACAGTGTCTCT




AGCAACAGTGCTGCT




TGGAACTGGATCAGG




CAGTCCCCATCGAGA




GGCCTTGAGTGGCTG




GGAAGGACATACTAC




AGGTCCAAGTGGTAT




AGTGATTATGCAGTA




TCTGTGAAAAGTCGA




ATAACCATCAACCCA




GACACATCCAAGAAC




CAGTTCTCCCTGCAG




CTGAACTCTGTGACT




CCCGAGGACACGGCT




GTGTATTACTGTGCA




AGAGATCCTTACGAT




TTTTGGAGTGGTTAT




CCTGATGCTTTTGAT




ATCTGGGGCCAAGGG




ACAAT




GGTCACCGTCTCTTCA





SEQ ID NO: 754
LCDR1
TGTSSDVGGYNYVS


(Kabat)







SEQ ID NO: 755
LCDR2
EVNNRPS


(Kabat)







SEQ ID NO: 756
LCDR3
SSYTSGRTLYV


(Kabat)







SEQ ID NO: 757
LCDR1
TSSDVGGYNY


(Chothia)







SEQ ID NO: 758
LCDR2
EVN


(Chothia)







SEQ ID NO: 759
LCDR3
YTSGRTLY


(Chothia)







SEQ ID NO: 760
VL
QSALTQPASVSGSPG




QSITISCTGTSSDVG




GYNYVSWYQQHPGKA




PKVIISEVNNRPSGV




SHRFSGSKSGNTASL




TISGLQAEDEADYFC




SSYTSGRTLYVFGTG




SKVTVLG





SEQ ID NO: 761
DNA VL
CAGTCTGCCCTGACT




CAGCCTGCCTCCGTG




TCTGGGTCTCCTGGA




CAGTCGATCACCATC




TCCTGCACTGGAACC




AGCAGTGACGTTGGT




GGTTACAACTATGTC




TCCTGGTACCAACAG




CACCCAGGCAAAGCC




CCCAAGGTCATAATT




TCTGAGGTCAATAAT




CGGCCCTCAGGGGTT




TCTCATCGCTTCTCT




GGGTCCAAGTCTGGC




AACACGGCCTCCCTG




ACCATCTCTGGGCTC




CAGGCTGAGGACGAG




GCTGATTATTTCTGC




AGCTCATATACAAGT




GGCAGGACTCTTTAT




GTCTTCGGAACTGGG




AGCAAGGTCACCGTC




CTAGGT





SEQ ID NO: 762
Linker
GGGGSGGGGSGGGGS





SEQ ID NO: 763
scFv (VH-
EVQLQQSGPGLVKPS



linker-VL)
QTLSLTCAISGDSVS




SNSAAWNWIRQSPSR




GLEWLGRTYYRSKWY




SDYAVSVKSRITINP




DTSKNQFSLQLNSVT




PEDTAVYYCARDPYD




FWSGYPDAFDIWGQG




TMVTVSSGGGGSGGG




GSGGGGSQSALTQPA




SVSGSPGQSITISCT




GTSSDVGGYNYVSWY




QQHPGKAPKVIISEV




NNRPSGVSHRFSGSK




SGNTASLTISGLQAE




DEADYFCSSYTSGRT




LYVFGTGSKVTVLG





CAR22 m971 (murine)




SEQ ID NO: 1332
VH
QVQLQQSGPGLVKPS




QTLSLTCAISGDSVS




SNSAAWNWIRQSPSR




GLEWLGRTYYRSKWY




NDYAVSVKSRITINP




DTSKNQFSLQLNSVT




PEDTAVYYCAREVTG




DLEDAFDIWGQGTMV




TV





SEQ ID NO: 1333
VL
DIQMTQSPSSLSASV




GDRVTITCRASQTIW




SYLNWYQQRPGKAPN




LLIYAASSLQSGVPS




RFSGRGSGTDFTLTI




SSLQAEDFATYYCQQ




SYSIPQTFGQGTKLE




IK





SEQ ID NO: 1334
HC CDR1
SNSAAWN


Kabat







SEQ ID NO: 1335
HC CDR2
RTYYRSKWYNDYAVS


Kabat

VKS





SEQ ID NO: 1336
HC CDR3
EVTGDLEDAFDI


Kabat







SEQ ID NO: 1337
LC CDR1
RASQTIWSYLN


Kabat







SEQ ID NO: 1338
LC CDR2
AASSLQS


Kabat







SEQ ID NO: 1339
LC CDR3
QQSYSIPQT


Kabat







SEQ ID NO: 1340
NT
gtgcacgagtgggtta




catcgaactggatct




caacagcggtaagat




ccttgagagttttcg




ccccgaagaacgttt




tccaatgatgagcac




ttttaaagttctgct




atgtggcgcggtatt




atcccgtattgacgc




cgggcaagagcaact




cggtcgccgcataca




ctattctcagaatga




cttggttgagtactc




accagtcacagaaaa




gcatcttacggatgg




catgacagtaagaga




attatgcagtgctgc




cataaccatgagtga




taacactgcggccaa




cttacttctgacaac




gatcggaggaccgaa




ggagctaaccgcttt




tttgcacaacatggg




ggatcatgtaactcg




ccttgatcgttggga




accggagctgaatga




agccataccaaacga




cgagcgtgacaccac




gatgcctgtagcaat




ggcaacaacgttgcg




caaactattaactgg




cgaactacttactct




agcttcccggcaaca




attaatagactggat




ggaggcggataaagt




tgcaggaccacttct




gcgctcggcccttcc




ggctggctggtttat




tgctgataaatctgg




agccggtgagcgtgg




gtctcgcggtatcat




tgcagcactggggcc




agatggtaagccctc




ccgtatcgtagttat




ctacacgacggggag




tcaggcaactatgga




tgaacgaaatagaca




gatcgctgagatagg




tgcctcactgattaa




gcattggtaactgtc




agaccaagtttactc




atatatactttagat




tgatttaaaacttca




tttttaatttaaaag




gatctaggtgaagat




cctttttgataatct




catgaccaaaatccc




ttaacgtgagttttc




gttccactgagcgtc




agaccccgtagaaaa




gatcaaaggatcttc




ttgagatcctttttt




tctgcgcgtaatctg




ctgcttgcaaacaaa




aaaaccaccgctacc




agcggtggtttgttt




gccggatcaagagct




accaactctttttcc




gaaggtaactggctt




cagcagagcgcagat




accaaatactgttct




tctagtgtagccgta




gttaggccaccactt




caagaactctgtagc




accgcctacatacct




cgctctgctaatcct




gttaccagtggctgc




tgccagtggcgataa




gtcgtgtcttaccgg




gttggactcaagacg




atagttaccggataa




ggcgcagcggtcggg




ctgaacggggggttc




gtgcacacagcccag




cttggagcgaacgac




ctacaccgaactgag




atacctacagcgtga




gctatgagaaagcgc




cacgcttcccgaagg




gagaaaggcggacag




gtatccggtaagcgg




cagggtcggaacagg




agagcgcacgaggga




gcttccagggggaaa




cgcctggtatcttta




tagtcctgtcgggtt




tcgccacctctgact




tgagcgtcgattttt




gtgatgctcgtcagg




ggggcggagcctatg




gaaaaacgccagcaa




cgcggcctttttacg




gttcctggccttttg




ctggccttttgctca




catgttctttcctgc




gttatcccctgattc




tgtggataaccgtat




taccgcctttgagtg




agctgataccgctcg




ccgcagccgaacgac




cgagcgcagcgagtc




agtgagcgaggaagc




ggaagagcgcccaat




acgcaaaccgcctct




ccccgcgcgttggcc




gattcattaatgcag




ctggcacgacaggtt




tcccgactggaaagc




gggcagtgagcgcaa




cgcaattaatgtgag




ttagctcactcatta




ggcaccccaggcttt




acactttatgcttcc




ggctcgtatgttgtg




tggaattgtgagcgg




ataacaatttcacac




aggaaacagctatga




ccatgattacgccaa




gcgcgcaattaaccc




tcactaaagggaaca




aaagctggagctgca




agcttaatgtagtct




tatgcaatactcttg




tagtcttgcaacatg




gtaacgatgagttag




caacatgccttacaa




ggagagaaaaagcac




cgtgcatgccgattg




gtggaagtaaggtgg




tacgatcgtgcctta




ttaggaaggcaacag




acgggtctgacatgg




attggacgaaccact




gaattgccgcattgc




agagatattgtattt




aagtgcctagctcga




tacataaacgggtct




ctctggttagaccag




atctgagcctgggag




ctctctggctaacta




gggaacccactgctt




aagcctcaataaagc




ttgccttgagtgctt




caagtagtgtgtgcc




cgtctgttgtgtgac




tctggtaactagaga




tccctcagacccttt




tagtcagtgtggaaa




atctctagcagtggc




gcccgaacagggact




tgaaagcgaaaggga




aaccagaggagctct




ctcgacgcaggactc




ggcttgctgaagcgc




gcacggcaagaggcg




aggggcggcgactgg




tgagtacgccaaaaa




ttttgactagcggag




gctagaaggagagag




atgggtgcgagagcg




tcagtattaagcggg




ggagaattagatcgc




gatgggaaaaaattc




ggttaaggccagggg




gaaagaaaaaatata




aattaaaacatatag




tatgggcaagcaggg




agctagaacgattcg




cagttaatcctggcc




tgttagaaacatcag




aaggctgtagacaaa




tactgggacagctac




aaccatcccttcaga




caggatcagaagaac




ttagatcattatata




atacagtagcaaccc




tctattgtgtgcatc




aaaggatagagataa




aagacaccaaggaag




ctttagacaagatag




aggaagagcaaaaca




aaagtaagaccaccg




cacagcaagcggccg




ctgatcttcagacct




ggaggaggagatatg




agggacaattggaga




agtgaattatataaa




tataaagtagtaaaa




attgaaccattagga




gtagcacccaccaag




gcaaagagaagagtg




gtgcagagagaaaaa




agagcagtgggaata




ggagctttgttcctt




gggttcttgggagca




gcaggaagcactatg




ggcgcagcgtcaatg




acgctgacggtacag




gccagacaattattg




tctggtatagtgcag




cagcagaacaatttg




ctgagggctattgag




gcgcaacagcatctg




ttgcaactcacagtc




tggggcatcaagcag




ctccaggcaagaatc




ctggctgtggaaaga




tacctaaaggatcaa




cagctcctggggatt




tggggttgctctgga




aaactcatttgcacc




actgctgtgccttgg




aatgctagttggagt




aataaatctctggaa




cagatttggaatcac




acgacctggatggag




tgggacagagaaatt




aacaattacacaagc




ttaatacactcctta




attgaagaatcgcaa




aaccagcaagaaaag




aatgaacaagaatta




ttggaattagataaa




tgggcaagtttgtgg




aattggtttaacata




acaaattggctgtgg




tatataaaattattc




ataatgatagtagga




ggcttggtaggttta




agaatagtttttgct




gtactttctatagtg




aatagagttaggcag




ggatattcaccatta




tcgtttcagacccac




ctcccaaccccgagg




ggacccgacaggccc




gaaggaatagaagaa




gaaggtggagagaga




gacagagacagatcc




attcgattagtgaac




ggatctcgacggtat




cgattagactgtagc




ccaggaatatggcag




ctagattgtacacat




ttagaaggaaaagtt




atcttggtagcagtt




catgtagccagtgga




tatatagaagcagaa




gtaattccagcagag




acagggcaagaaaca




gcatacttcctctta




aaattagcaggaaga




tggccagtaaaaaca




gtacatacagacaat




ggcagcaatttcacc




agtactacagttaag




gccgcctgttggtgg




gcggggatcaagcag




gaatttggcattccc




tacaatccccaaagt




caaggagtaatagaa




tctatgaataaagaa




ttaaagaaaattata




ggacaggtaagagat




caggctgaacatctt




aagacagcagtacaa




atggcagtattcatc




cacaattttaaaaga




aaaggggggattggg




ggggtacagtgcagg




ggaaagaatagtaga




cataatagcaacaga




catacaaactaaaga




attacaaaaacaaat




tacaaaaattcaaaa




ttttcgggtttatta




cagggacagcagaga




tccagtttggctgca




tacgcgtcgtgaggc




tccggtgcccgtcag




tgggcagagcgcaca




tcgcccacagtcccc




gagaagttgggggga




ggggtcggcaattga




accggtgcctagaga




aggtggcgcggggta




aactgggaaagtgat




gtcgtgtactggctc




cgcctttttcccgag




ggtgggggagaaccg




tatataagtgcagta




gtcgccgtgaacgtt




ctttttcgcaacggg




tttgccgccagaaca




caggtaagtgccgtg




tgtggttcccgcggg




cctggcctctttacg




ggttatggcccttgc




gtgccttgaattact




tccacctggctgcag




tacgtgattcttgat




cccgagcttcgggtt




ggaagtgggtgggag




agttcgaggccttgc




gcttaaggagcccct




tcgcctcgtgcttga




gttgaggcctggcct




gggcgctggggccgc




cgcgtgcgaatctgg




tggcaccttcgcgcc




tgtctcgctgctttc




gataagtctctagcc




atttaaaatttttga




tgacctgctgcgacg




ctttttttctggcaa




gatagtcttgtaaat




gcgggccaagatctg




cacactggtatttcg




gtttttggggccgcg




ggcggcgacggggcc




cgtgcgtcccagcgc




acatgttcggcgagg




cggggcctgcgagcg




cggccaccgagaatc




ggacgggggtagtct




caagctggccggcct




gctctggtgcctggc




ctcgcgccgccgtgt




atcgccccgccctgg




gcggcaaggctggcc




cggtcggcaccagtt




gcgtgagcggaaaga




tggccgcttcccggc




cctgctgcagggagc




tcaaaatggaggacg




cggcgctcgggagag




cgggcgggtgagtca




cccacacaaaggaaa




agggcctttccgtcc




tcagccgtcgcttca




tgtgactccactgag




taccgggcgccgtcc




aggcacctcgattag




ttctcgtgcttttgg




agtacgtcgtcttta




ggttggggggagggg




ttttatgcgatggag




tttccccacactgag




tgggtggagactgaa




gttaggccagcttgg




cacttgatgtaattc




tccttggaatttgcc




ctttttgagtttgga




tcttggttcattctc




aagcctcagacagtg




gttcaaagttttttt




cttccatttcaggtg




tcgtgagctagctct




agagccaccatggcc




ctgcctgtgacagcc




ctgctgctgcctctg




gctctgctgctgcat




gccgctagacccgga




tcccaggtgcagctg




cagcagtctggaccc




ggcctcgtgaagcct




agccagaccctgtct




ctgacctgcgccatc




agcggcgatagcgtg




tccagcaatagcgcc




gcctggaactggatc




agacagagccctagc




agaggcctggaatgg




ctgggccggacctac




taccggtccaagtgg




tacaacgactacgcc




gtgtccgtgaagtcc




cggatcaccatcaac




cccgacaccagcaag




aaccagttctccctg




cagctgaacagcgtg




acccccgaggatacc




gccgtgtactactgc




gccagagaagtgacc




ggcgacctggaagat




gccttcgacatctgg




ggccagggcacaatg




gtcaccgtgtctagc




ggaggcggaggatct




ggcggcggaggaagt




ggcggagggggatct




gggggaggcggaagc




gatatccagatgacc




cagagccccagctcc




ctgtctgccagcgtg




ggcgacagagtgacc




atcacctgtagggcc




agccagaccatctgg




tcctacctgaactgg




tatcagcagcggcct




ggcaaggcccccaac




ctgctgatctatgcc




gccagctctctgcag




tccggcgtgcccagc




agattttccggcaga




ggctccggcaccgac




ttcaccctgacaatc




agttccctgcaggcc




gaggacttcgccacc




tactactgccagcag




agctacagcatcccc




cagaccttcggccag




gggaccaagctggaa




atcaagtccggaacc




acgacgccagcgccg




cgaccaccaacaccg




gcgcccaccatcgcg




tcgcagcccctgtcc




ctgcgcccagaggcg




tgccggccagcggcg




gggggcgcagtgcac




acgagggggctggac




ttcgcctgtgatatc




tacatctgggcgccc




ttggccgggacttgt




ggggtccttctcctg




tcactggttatcacc




ctttactgcaaacgg




ggcagaaagaaactc




ctgtatatattcaaa




caaccatttatgaga




ccagtacaaactact




caagaggaagatggc




tgtagctgccgattt




ccagaagaagaagaa




ggaggatgtgaactg




agagtgaagttcagc




aggagcgcagacgcc




cccgcgtacaagcag




ggccagaaccagctc




tataacgagctcaat




ctaggacgaagagag




gagtacgatgttttg




gacaagagacgtggc




cgggaccctgagatg




gggggaaagccgaga




aggaagaaccctcag




gaaggcctgtacaat




gaactgcagaaagat




aagatggcggaggcc




tacagtgagattggg




atgaaaggcgagcgc




cggaggggcaagggg




cacgatggcctttac




cagggtctcagtaca




gccaccaaggacacc




tacgacgcccttcac




atgcaggccctgccc




cctcgctaagtcgac




aatcaacctctggat




tacaaaatttgtgaa




agattgactggtatt




cttaactatgttgct




ccttttacgctatgt




ggatacgctgcttta




atgcctttgtatcat




gctattgcttcccgt




atggctttcattttc




tcotccttgtataaa




tcctggttgctgtct




ctttatgaggagttg




tggcccgttgtcagg




caacgtggcgtggtg




tgcactgtgtttgct




gacgcaacccccact




ggttggggcattgcc




accacctgtcagctc




ctttccgggactttc




gctttccccctccct




attgccacggcggaa




ctcatcgccgcctgc




cttgcccgctgctgg




acaggggctcggctg




ttgggcactgacaat




tccgtggtgttgtcg




gggaagctgacgtcc




tttccatggctgctc




gcctgtgttgccacc




tggattctgcgcggg




acgtccttctgctac




gtcccttcggccctc




aatccagcggacctt




ccttcccgcggcctg




ctgccggctctgcgg




cctcttccgcgtctt




cgccttcgccctcag




acgagtcggatctcc




ctttgggccgcctcc




ccgcctggaattcga




gctcggtacctttaa




gaccaatgacttaca




aggcagctgtagatc




ttagccactttttaa




aagaaaaggggggac




tggaagggctaattc




actcccaacgaagac




aagatctgctttttg




cttgtactgggtctc




tctggttagaccaga




tctgagcctgggagc




tctctggctaactag




ggaacccactgctta




agcctcaataaagct




tgccttgagtgcttc




aagtagtgtgtgccc




gtctgttgtgtgact




ctggtaactagagat




ccctcagaccctttt




agtcagtgtggaaaa




tctctagcagtagta




gttcatgtcatctta




ttattcagtatttat




aacttgcaaagaaat




gaatatcagagagtg




agaggaacttgttta




ttgcagcttataatg




gttacaaataaagca




atagcatcacaaatt




tcacaaataaagcat




ttttttcactgcatt




ctagttgtggtttgt




ccaaactcatcaatg




tatcttatcatgtct




ggctctagctatccc




gcccctaactccgcc




cagttccgcccattc




tccgccccatggctg




actaattttttttat




ttatgcagaggccga




ggccgcctcggcctc




tgagctattccagaa




gtagtgaggaggctt




ttttggaggcctagg




cttttgcgtcgagac




gtacccaattcgccc




tatagtgagtcgtat




tacgcgcgctcactg




gccgtcgttttacaa




cgtcgtgactgggaa




aaccctggcgttacc




caacttaatcgcctt




gcagcacatccccct




ttcgccagctggcgt




aatagcgaagaggcc




cgcaccgatcgccct




tcccaacagttgcgc




agcctgaatggcgaa




tgggacgcgccctgt




agcggcgcattaagc




gcggcgggtgtggtg




gttacgcgcagcgtg




accgctacacttgcc




agcgccctagcgccc




gctcctttcgctttc




ttcccttcctttctc




gccacgttcgccggc




tttccccgtcaagct




ctaaatcgggggctc




cctttagggttccga




tttagtgctttacgg




cacctcgaccccaaa




aaacttgattagggt




gatggttcacgtagt




gggccatcgccctga




tagacggtttttcgc




cctttgacgttggag




tccacgttctttaat




agtggactcttgttc




caaactggaacaaca




ctcaaccctatctcg




gtctattcttttgat




ttataagggattttg




ccgatttcggcctat




tggttaaaaaatgag




ctgatttaacaaaaa




tttaacgcgaatttt




aacaaaatattaacg




cttacaatttaggtg




gcacttttcggggaa




atgtgcgcggaaccc




ctatttgtttatttt




tctaaatacattcaa




atatgtatccgctca




tgagacaataaccct




gataaatgcttcaat




aatattgaaaaagga




agagtatgagtattc




aacatttccgtgtcg




cccttattccctttt




ttgcggcattttgcc




ttcctgtttttgctc




acccagaaacgctgg




tgaaagtaaaagatg




ctgaagatcagttgg









In some embodiments, the antigen binding domain comprises a HC CDR1, a HC CDR2, and a HC CDR3 of any heavy chain binding domain amino acid sequences listed in Table 6, 7 or 9. In embodiments, the antigen binding domain further comprises a LC CDR1, a LC CDR2, and a LC CDR3. In embodiments, the antigen binding domain comprises a LC CDR1, a LC CDR2, and a LC CDR3 of any light chain binding domain amino acid sequences listed in Table 6, 8 or 10.


In some embodiments, the antigen binding domain comprises one, two or all of LC CDR1, LC CDR2, and LC CDR3 of any light chain binding domain amino acid sequences listed in Table 6, 8 or 10, and one, two or all of HC CDR1, HC CDR2, and HC CDR3 of any heavy chain binding domain amino acid sequences listed in Table 6, 7 or 9.


In some embodiments, the CDRs are defined according to the Kabat numbering scheme, the Chothia numbering scheme, or a combination thereof. An overview of the sequences identifications of CDR (Kabat) sequences of the CD22 scFv domains are shown in Table for the heavy chain variable domains and in Table for the light chain variable domains. The SEQ ID NO's refer to those found in Table 6.









TABLE 7







Heavy Chain Variable Domain CDR (Kabat) SEQ ID NO's


of CD22 antibody molecules










Candidate
HCDR1
HCDR2
HCDR3





CD22-57
519
520
521


CD22-58
544
545
546


CD22-59
569
570
571


CD22-60
594
595
596


CD22-61
619
620
621


CD22-62
644
645
646


CD22-63
669
670
671


CD22-64
694
695
696


CD22-65
719
720
721


CAR22m971
1334 
1335 
1336 


(murine)



















TABLE 8







Light Chain Variable Domain CDR (Kabat) SEQ ID NO's


of CD22 Antibody Molecules










Candidate
LCDR1
LCDR2
LCDR3





CD22-57
530
531
532


CD22-58
555
556
557


CD22-59
580
581
582


CD22-60
605
606
607


CD22-61
630
631
632


CD22-62
655
656
657


CD22-63
680
681
682


CD22-64
705
706
707


CD22-65
730
731
732


CAR22m971
1337 
1338 
1339 


(murine)
















TABLE 9







Heavy Chain Variable Region SEQ ID NO's of


CD22 antibody molecules











Heavy Chain



Candidate
Variable region







CD22-57
528



CD22-58
553



CD22-59
578



CD22-60
603



CD22-61
628



CD22-62
653



CD22-63
678



CD22-64
703



CD22-65
728



CAR22m971
1333 



(murine)

















TABLE 10







Light Chain Variable Region SEQ ID NO's of


CD22 antibody molecules











Light Chain



Candidate
Variable region







CD22-57
539



CD22-58
564



CD22-59
589



CD22-60
614



CD22-61
639



CD22-62
664



CD22-63
689



CD22-64
714



CD22-65
739



CAR22m971
1334 



(murine)










In some embodiments, the CD22 CAR comprises a short Gly-Ser linker (e.g., GGGGS linker (SEQ ID NO: 18)) between the VH and VL sequences in the scFv as depicted in Construct CD22-65s, e.g., in Table 6.


In some embodiments, the CD22 CAR does not have a linker sequence between the VH and VL sequences in the scFv as depicted in Construct CD22-65ss, e.g., in Table 6.


In yet another embodiment, the CD22 CAR comprises one or more mutations relative to the amino acid sequence of CD22-65s, e.g., one or more mutations in the FR region of the VH and/or VL. In one embodiment, the CD22 CAR comprises a mutation at amino acid 41 of the VH region CD22-65s (e.g., a substitution of Q at position 41 of the VH of CD22-65s, e.g., for K); and/or a mutation of amino acid 40 of the VL of CD22-65s (e.g., a substitution of Q at position 40 of the VL of CD22-65s, e.g., for D). In one embodiment, the CD22CAR comprises the amino acid sequence of CD22-65sKD depicted below. An alignment of the the CD22-65s (SEQ ID NO: 835) and CD22-65sKD (SEQ ID NO: 837) is depicted below.












CD22-65sKD
  1 EVQWQQSGPGLVKPSQTLSLTCAISGDSMLSNSDTWNWIRKSPSRGLEWL
50




    ||||||||||||||||||||||||||||||||||||||||||||||||||



CD22-65s
  1 EVQLQQSGPGLVKPSQTLSLTCAISGDSMLSNSDTWNWIRQSPSRGLEWL
50





CD22-65sKD
 51 GRTYHRSTWYDDYASSVRGRVSINVDTSKNQYSLQLNAVTPEDTGVYYCA
100



    ||||||||||||||||||||||||||||||||||||||||||||||||||



CD22-65s
 51 GRTYHRSTWYDDYASSVRGRVSIKVDTSKNQYSLQLNAVTPEDTGVYYCA
100





CD22-65sKD
101 RVRLQDGNSWSDAFDVWGQGTMVTVSSGGGGSQSALTQPASASGSPGQSV
150



    ||||||||||||||||||||||||||||||||||||||||||||||||||



CD22-65s
101 RVRLQDGNSWSDAFDVWGQGTMVTVSSGGGGSQSALTQPASASGSPGQSV
150





CD22-65sKD
151 TISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSNRPSGVSNRFSGSK
200



    ||||||||||||||||||||||||||||||||||||||||||||||||||



CD22-65s
151 TISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSNRPSGVSNRFSGSK
200





CD22-65sKD
201 SGNTASLTISGLQAEDEADYYCSSYTSSSTLYVFGTGTQLTVL
243



    |||||||||||||||||||||||||||||||||||||||||||



CD22-65s
201 SGNTASLTISGLQAEDEADYYCSSYTSSSTLYVFGTGTQLTVL
243






In some embodiments, the antigen binding domain comprises a HC CDR1, a HC CDR2, and a HC CDR3 of any heavy chain binding domain amino acid sequences listed in Table 7 or 9. In embodiments, the antigen binding domain further comprises a LC CDR1, a LC CDR2, and a LC CDR3. In embodiments, the antigen binding domain comprises a LC CDR1, a LC CDR2, and a LC CDR3 of any light chain binding domain amino acid sequences listed in Table 8 or 10.


In some embodiments, the antigen binding domain comprises one, two or all of LC CDR1, LC CDR2, and LC CDR3 of any light chain binding domain amino acid sequences listed in Table 8 or 10, and one, two or all of HC CDR1, HC CDR2, and HC CDR3 of any heavy chain binding domain amino acid sequences listed in Table 7 or 9.


In some embodiments, the CDRs are defined according to the Kabat numbering scheme, the Chothia numbering scheme, or a combination thereof.


The order in which the VL and VH domains appear in the scFv can be varied (i.e., VL-VH, or VH-VL orientation), and where either three or four copies of the “G4S” (SEQ ID NO:18) subunit, in which each subunit comprises the sequence GGGGS (SEQ ID NO:18) (e.g., (G4S)3 (SEQ ID NO:107) or (G4S)4(SEQ ID NO:106)), can connect the variable domains to create the entirety of the scFv domain. Alternatively, the CAR construct can include, for example, a linker including the sequence GSTSGSGKPGSGEGSTKG (SEQ ID NO: 1322).


These clones all contained a Q/K residue change in the signal domain of the co-stimulatory domain derived from CD3zeta chain.


CD19 CARs and Binding Domains

Provided herein are compositions of matter and methods of use for the treatment of a disease such as cancer using CD19 chimeric antigen receptors (CAR). The methods include, inter alia, administering a CD19 CAR described herein in combination with another agent such as a CD22 CAR. The methods also include, e.g., administering a CD19 CAR described herein to treat a leukemia, e.g., ALL, e.g., relapsed and/or refractory ALL, or a lymphoma such as Hodgkin lymphoma.


In one aspect, the invention provides a number of chimeric antigen receptors (CAR) comprising an antibody or antibody fragment engineered for specific binding to a CD19 protein. In one aspect, the invention provides a cell (e.g., T cell) engineered to express a CAR, wherein the CAR T cell (“CART”) exhibits an anticancer property. In one aspect a cell is transformed with the CAR and the CAR is expressed on the cell surface. In some embodiments, the cell (e.g., T cell) is transduced with a viral vector encoding a CAR. In some embodiments, the viral vector is a retroviral vector. In some embodiments, the viral vector is a lentiviral vector. In some such embodiments, the cell may stably express the CAR. In another embodiment, the cell (e.g., T cell) is transfected with a nucleic acid, e.g., mRNA, cDNA, DNA, encoding a CAR. In some such embodiments, the cell may transiently express the CAR.


In one aspect, the anti-CD19 protein binding portion of the CAR is a scFv antibody fragment. In one aspect such antibody fragments are functional in that they retain the equivalent binding affinity, e.g., they bind the same antigen with comparable affinity, as the IgG antibody from which it is derived. In one aspect such antibody fragments are functional in that they provide a biological response that can include, but is not limited to, activation of an immune response, inhibition of signal-transduction origination from its target antigen, inhibition of kinase activity, and the like, as will be understood by a skilled artisan. In one aspect, the anti-CD19 antigen binding domain of the CAR is a scFv antibody fragment that is humanized compared to the murine sequence of the scFv from which it is derived. In an embodiment, the humanized anti-CD19 binding domain comprises the amino acid sequence of SEQ ID NO:2, or an amino acid sequence at least 95%, 96%, 97%, 09%, or 99% identical thereto. In one aspect, the parental murine scFv sequence is the CAR19 construct provided in PCT publication WO2012/079000 and provided herein as SEQ ID NO:59. In one embodiment, the anti-CD19 binding domain is a scFv described in WO2012/079000 and provided in SEQ ID NO:59, or a sequence at least 95%, e.g., 95-99%, identical thereto. In an embodiment, the anti-CD19 binding domain is part of a CAR construct provided in PCT publication WO2012/079000 and provided herein as SEQ ID NO:58, or a sequence at least 95%, e.g., 95%-99%, identical thereto. In an embodiment, the anti-CD19 binding domain comprises at least one (e.g., 2, 3, 4, 5, or 6) CDRs selected from Table 4 and/or Table 5.


In some aspects, the antibodies of the invention are incorporated into a chimeric antigen receptor (CAR). In one aspect, the CAR comprises the polypeptide sequence provided as SEQ ID NO: 12 in PCT publication WO2012/079000, and provided herein as SEQ ID NO: 58, wherein the scFv domain is substituted by one or more sequences selected from SEQ ID NOS: 1-12. In one aspect, the scFv domains of SEQ ID NOS:1-12 are humanized variants of the scFv domain of SEQ ID NO:59, which is an scFv fragment of murine origin that specifically binds to human CD19. Humanization of this mouse scFv may be desired for the clinical setting, where the mouse-specific residues may induce a human-anti-mouse antigen (HAMA) response in patients who receive CART19 treatment, e.g., treatment with T cells transduced with the CAR19 construct.


In one aspect, the anti-CD19 binding domain, e.g., humanized scFv, portion of a CAR of the invention is encoded by a transgene whose sequence has been codon optimized for expression in a mammalian cell. In one aspect, entire CAR construct of the invention is encoded by a transgene whose entire sequence has been codon optimized for expression in a mammalian cell. Codon optimization refers to the discovery that the frequency of occurrence of synonymous codons (i.e., codons that code for the same amino acid) in coding DNA is biased in different species. Such codon degeneracy allows an identical polypeptide to be encoded by a variety of nucleotide sequences. A variety of codon optimization methods is known in the art, and include, e.g., methods disclosed in at least U.S. Pat. Nos. 5,786,464 and 6,114,148.


In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:1. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:2. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:3. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:4. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:5. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:6. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:7. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:8. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:9. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:10. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:11. In one aspect, the humanized CAR19 comprises the scFv portion provided in SEQ ID NO:12.


In one aspect, the CARs of the invention combine an antigen binding domain of a specific antibody with an intracellular signaling molecule. For example, in some aspects, the intracellular signaling molecule includes, but is not limited to, CD3-zeta chain, 4-1BB and CD28 signaling modules and combinations thereof. In one aspect, the CD19 CAR comprises a CAR selected from the sequence provided in one or more of SEQ ID NOS: 31-42. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:31. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:32. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:33. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:34. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:35. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:36. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:37. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:38. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:39. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:40. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:41. In one aspect, the CD19 CAR comprises the sequence provided in SEQ ID NO:42.


Thus, in one aspect, the antigen binding domain comprises a humanized antibody or an antibody fragment. In one embodiment, the humanized anti-CD19 binding domain comprises one or more (e.g., all three) light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of a murine or humanized anti-CD19 binding domain described herein, and/or one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of a murine or humanized anti-CD19 binding domain described herein, e.g., a humanized anti-CD19 binding domain comprising one or more, e.g., all three, LC CDRs and one or more, e.g., all three, HC CDRs. In one embodiment, the humanized anti-CD19 binding domain comprises one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of a murine or humanized anti-CD19 binding domain described herein, e.g., the humanized anti-CD19 binding domain has two variable heavy chain regions, each comprising a HC CDR1, a HC CDR2 and a HC CDR3 described herein. In one embodiment, the humanized anti-CD19 binding domain comprises a humanized light chain variable region described herein (e.g., in Table 2) and/or a humanized heavy chain variable region described herein (e.g., in Table 2). In one embodiment, the humanized anti-CD19 binding domain comprises a humanized heavy chain variable region described herein (e.g., in Table 2), e.g., at least two humanized heavy chain variable regions described herein (e.g., in Table 2). In one embodiment, the anti-CD19 binding domain is a scFv comprising a light chain and a heavy chain of an amino acid sequence of Table 2. In an embodiment, the anti-CD19 binding domain (e.g., an scFv) comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 2, or a sequence with 95-99% identity with an amino acid sequence of Table 2; and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 2, or a sequence with 95-99% identity to an amino acid sequence of Table 2. In one embodiment, the humanized anti-CD19 binding domain comprises a sequence selected from a group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, and SEQ ID NO:12, or a sequence with 95-99% identity thereof. In one embodiment, the nucleic acid sequence encoding the humanized anti-CD19 binding domain comprises a sequence selected from a group consisting of SEQ ID NO:61, SEQ ID NO:62, SEQ ID NO:63, SEQ ID NO:64, SEQ ID NO:65, SEQ ID NO:66, SEQ ID NO:67, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:71 and SEQ ID NO:72, or a sequence with 95-99% identity thereof. In one embodiment, the humanized anti-CD19 binding domain is a scFv, and a light chain variable region comprising an amino acid sequence described herein, e.g., in Table 2, is attached to a heavy chain variable region comprising an amino acid sequence described herein, e.g., in Table 2, via a linker, e.g., a linker described herein. In one embodiment, the humanized anti-CD19 binding domain includes a (Gly4-Ser)n linker, wherein n is 1, 2, 3, 4, 5, or 6, e.g., 3 or 4 (SEQ ID NO:53). The light chain variable region and heavy chain variable region of a scFv can be, e.g., in any of the following orientations: light chain variable region-linker-heavy chain variable region or heavy chain variable region-linker-light chain variable region.


In one aspect, the antigen binding domain portion comprises one or more sequence selected from SEQ ID NOS:1-12. In one aspect the humanized CAR is selected from one or more sequence selected from SEQ ID NOS: 31-42. In some aspects, a non-human antibody is humanized, where specific sequences or regions of the antibody are modified to increase similarity to an antibody naturally produced in a human or fragment thereof.


In one embodiment, the CAR molecule comprises an anti-CD19 binding domain comprising one or more (e.g., all three) light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of an anti-CD19 binding domain described herein, and one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of an anti-CD19 binding domain described herein, e.g., an anti-CD19 binding domain comprising one or more, e.g., all three, LC CDRs and one or more, e.g., all three, HC CDRs. In one embodiment, the anti-CD19 binding domain comprises one or more (e.g., all three) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of an anti-CD19 binding domain described herein, e.g., the anti-CD19 binding domain has two variable heavy chain regions, each comprising a HC CDR1, a HC CDR2 and a HC CDR3 described herein.


In one aspect, the anti-CD19 binding domain is characterized by particular functional features or properties of an antibody or antibody fragment. For example, in one aspect, the portion of a CAR composition of the invention that comprises an antigen binding domain specifically binds human CD19. In one aspect, the invention relates to an antigen binding domain comprising an antibody or antibody fragment, wherein the antibody binding domain specifically binds to a CD19 protein or fragment thereof, wherein the antibody or antibody fragment comprises a variable light chain and/or a variable heavy chain that includes an amino acid sequence of SEQ ID NO: 1-12 or SEQ ID NO:59. In one aspect, the antigen binding domain comprises an amino acid sequence of an scFv selected from SEQ ID NOs: 1-12 or SEQ ID NO:59. In certain aspects, the scFv is contiguous with and in the same reading frame as a leader sequence. In one aspect the leader sequence is the polypeptide sequence provided as SEQ ID NO:13.


In one aspect, the portion of the CAR comprising the antigen binding domain comprises an antigen binding domain that targets CD19. In one aspect, the antigen binding domain targets human CD19. In one aspect, the antigen binding domain of the CAR has the same or a similar binding specificity as, or includes, the FMC63 scFv fragment described in Nicholson et al. Mol. Immun. 34 (16-17): 1157-1165 (1997). In one aspect, the portion of the CAR comprising the antigen binding domain comprises an antigen binding domain that targets a B-cell antigen, e.g., a human B-cell antigen. A CD19 antibody molecule can be, e.g., an antibody molecule (e.g., a humanized anti-CD19 antibody molecule) described in WO2014/153270, which is incorporated herein by reference in its entirety. WO2014/153270 also describes methods of assaying the binding and efficacy of various CART constructs.


In one embodiment, the anti-CD19 binding domain comprises a murine light chain variable region described herein (e.g., in Table 3) and/or a murine heavy chain variable region described herein (e.g., in Table 3). In one embodiment, the anti-CD19 binding domain is a scFv comprising a murine light chain and a murine heavy chain of an amino acid sequence of Table 3. In an embodiment, the anti-CD19 binding domain (e.g., an scFv) comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 3, or a sequence with 95-99% identity with an amino acid sequence of Table 3; and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 3, or a sequence with 95-99% identity to an amino acid sequence of Table 3. In one embodiment, the anti-CD19 binding domain comprises a sequence of SEQ ID NO:59, or a sequence with 95-99% identity thereof. In one embodiment, the anti-CD19 binding domain is a scFv, and a light chain variable region comprising an amino acid sequence described herein, e.g., in Table 3, is attached to a heavy chain variable region comprising an amino acid sequence described herein, e.g., in Table 3, via a linker, e.g., a linker described herein. In one embodiment, the antigen binding domain includes a (Gly4-Ser)n linker, wherein n is 1, 2, 3, 4, 5, or 6, e.g., 3 or 4 (SEQ ID NO: 53). The light chain variable region and heavy chain variable region of a scFv can be, e.g., in any of the following orientations: light chain variable region-linker-heavy chain variable region or heavy chain variable region-linker-light chain variable region.


Furthermore, the present invention provides (among other things) CD19 CAR compositions, optionally in combination with a CD22 CAR, and their use in medicaments or methods for treating, among other diseases, cancer or any malignancy or autoimmune diseases involving cells or tissues which express CD19.


In one aspect, the CAR of the invention can be used to eradicate CD19-expressing normal cells, thereby applicable for use as a cellular conditioning therapy prior to cell transplantation. In one aspect, the CD19-expressing normal cell is a CD19-expressing normal stem cell and the cell transplantation is a stem cell transplantation.


In one aspect, the invention provides a cell (e.g., T cell) engineered to express a chimeric antigen receptor (CAR), wherein the CAR-expressing cell, e.g., CAR T cell (“CART”) exhibits an anticancer property. A suitable antigen is CD19. In one aspect, the antigen binding domain of the CAR comprises a partially humanized anti-CD19 antibody fragment. In one aspect, the antigen binding domain of the CAR comprises a partially humanized anti-CD19 antibody fragment comprising an scFv. Accordingly, the invention provides (among other things) a CD19-CAR that comprises a humanized anti-CD19 binding domain and is engineered into an immune effector cell, e.g., a T cell or an NK cell, and methods of their use for adoptive therapy.


In one aspect, the CAR, e.g., CD19-CAR comprises at least one intracellular domain selected from the group of a CD137 (4-1BB) signaling domain, a CD28 signaling domain, a CD3zeta signal domain, and any combination thereof. In one aspect, the CAR, e.g., CD19-CAR comprises at least one intracellular signaling domain is from one or more co-stimulatory molecule(s) other than a CD137 (4-1BB) or CD28.


The present invention encompasses, but is not limited to, a recombinant DNA construct comprising sequences encoding a CAR, wherein the CAR comprises an antibody or antibody fragment that binds specifically to CD19, or CD22, wherein the sequence of the antibody fragment is contiguous with and in the same reading frame as a nucleic acid sequence encoding an intracellular signaling domain. The intracellular signaling domain can comprise a costimulatory signaling domain and/or a primary signaling domain, e.g., a zeta chain. The costimulatory signaling domain refers to a portion of the CAR comprising at least a portion of the intracellular domain of a costimulatory molecule. In one embodiment, the antigen binding domain is a murine antibody or antibody fragment described herein. In one embodiment, the antigen binding domain is a humanized antibody or antibody fragment.


In specific aspects, a CAR construct of the invention comprises a scFv domain selected from the group consisting of SEQ ID NOS:1-12 or an scFV domain of SEQ ID NO:59, wherein the scFv may be preceded by an optional leader sequence such as provided in SEQ ID NO: 13, and followed by an optional hinge sequence such as provided in SEQ ID NO:14 or SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49, a transmembrane region such as provided in SEQ ID NO:15, an intracellular signalling domain that includes SEQ ID NO:16 or SEQ ID NO:51 and a CD3 zeta sequence that includes SEQ ID NO:17 or SEQ ID NO:43, wherein the domains are contiguous with and in the same reading frame to form a single fusion protein.


Also included in the invention (among other things) is a nucleotide sequence that encodes the polypeptide of each of the scFv fragments selected from the group consisting of SEQ IS NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ IS NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12 and SEQ ID NO:59. Also included in the invention (among other things) is a nucleotide sequence that encodes the polypeptide of each of the scFv fragments selected from the group consisting of SEQ IS NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ IS NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12 and SEQ ID NO:59, and each of the domains of SEQ ID NOS: 13-17, plus an encoded CD19 CAR fusion protein of the invention.


In one aspect an exemplary CD19 CAR constructs comprise an optional leader sequence, an extracellular antigen binding domain, a hinge, a transmembrane domain, and an intracellular stimulatory domain.


In one aspect an exemplary CD19 CAR construct comprises an optional leader sequence, an extracellular antigen binding domain, a hinge, a transmembrane domain, an intracellular costimulatory domain and an intracellular stimulatory domain. In some embodiments, specific CD19 CAR constructs containing humanized scFv domains of the invention are provided as SEQ ID NOS: 31-42, or a murine scFv domain as provided as SEQ ID NO:59.


Full-length CAR sequences are also provided herein as SEQ ID NOS: 31-42 and 58, as shown in Table 2 and Table 3.


An exemplary leader sequence is provided as SEQ ID NO: 13. An exemplary hinge/spacer sequence is provided as SEQ ID NO: 14 or SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49. An exemplary transmembrane domain sequence is provided as SEQ ID NO:15. An exemplary sequence of the intracellular signaling domain of the 4-1BB protein is provided as SEQ ID NO: 16. An exemplary sequence of the intracellular signaling domain of CD27 is provided as SEQ ID NO:51. An exemplary CD3zeta domain sequence is provided as SEQ ID NO: 17 or SEQ ID NO:43. These sequences may be used, e.g., in combination with an scFv that recognizes CD22.


Exemplary sequences of various scFv fragments and other CAR components are provided herein. It is noted that these CAR components (e.g., of SEQ ID NO: 121, or a sequence of Table 2, 3, 6, 11A, 11B, 16, or 25) without a leader sequence (e.g., without the amino acid sequence of SEQ ID NO: 13 or a nucleotide sequence of SEQ ID NO: 54), are also provided herein.


In embodiments, the CAR sequences described herein contain a Q/K residue change in the signal domain of the co-stimulatory domain derived from CD3zeta chain.


In one aspect, the present invention encompasses a recombinant nucleic acid construct comprising a nucleic acid molecule encoding a CAR, wherein the nucleic acid molecule comprises the nucleic acid sequence encoding an anti-CD19 binding domain, e.g., described herein, that is contiguous with and in the same reading frame as a nucleic acid sequence encoding an intracellular signaling domain. In one aspect, the anti-CD19 binding domain is selected from one or more of SEQ ID NOS:1-12 and 58. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of the sequence provided in one or more of SEQ ID NOS:61-72 and 97. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:61. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:62. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:63. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:64. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:65. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:66. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:67. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:68. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:69. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:70. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:71. In one aspect, the anti-CD19 binding domain is encoded by a nucleotide residues 64 to 813 of SEQ ID NO:72.


Provided herein are CD19 inhibitors and combination therapies. In some embodiments, the CD19 inhibitor (e.g., a cell therapy or an antibody) is administered in combination with a B cell inhibitor, e.g., one or more inhibitors of CD10, CD19, CD20, CD22, CD34, CD123, FLT-3, or ROR1. A CD19 inhibitor includes but is not limited to a CD19 CAR-expressing cell, e.g., a CD19 CART cell, or an anti-CD19 antibody (e.g., an anti-CD19 mono- or bispecific antibody) or a fragment or conjugate thereof. In an embodiment, the CD19 inhibitor is administered in combination with a B-cell inhibitor, e.g., a CAR-expressing cell described herein.


Numerous CD19 CAR-expressing cells are described in this disclosure. For instance, in some embodiments, a CD19 inhibitor includes an anti-CD19 CAR-expressing cell, e.g., CART, e.g., a cell expressing an anti-CD19 CAR construct described in Table 2 or encoded by a CD19 binding CAR comprising a scFv, CDRs, or VH and VL chains described in Tables 2, 4, or 5. For example, an anti-CD19 CAR-expressing cell, e.g., CART, is a generated by engineering a CD19-CAR (that comprises a CD19 binding domain) into a cell (e.g., a T cell or NK cell), e.g., for administration in combination with a CAR-expressing cell described herein. Also provided herein are methods of use of the CAR-expressing cells described herein for adoptive therapy.


In one embodiment, an antigen binding domain comprises one, two three (e.g., all three) heavy chain CDRs, HC CDR1, HC CDR2 and HC CDR3, from an antibody listed herein, e.g., in Table 2, 4, or 5 and/or one, two, three (e.g., all three) light chain CDRs, LC CDR1, LC CDR2 and LC CDR3, from an antibody listed herein, e.g., in Table 2, 4, or 5. In one embodiment, the antigen binding domain comprises a heavy chain variable region and/or a variable light chain region of an antibody listed or described above.


In an embodiment, the CD19 binding domain (e.g., an scFv) comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 2, or a sequence with 95-99% identity with an amino acid sequence of Table 2; and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 2, or a sequence with 95-99% identity to an amino acid sequence of Table 2. In embodiments, the CD19 binding domain comprises one or more CDRs (e.g., one each of a HC CDR1, HC CDR2, HC CDR3, LC CDR1, LC CDR2, and LC CDR3) of Table 4 or Table 5, or CDRs having one, two, three, four, five, or six modifications (e.g., substitutions) of one or more of the CDRs.


Exemplary anti-CD19 antibodies or fragments or conjugates thereof include but are not limited to blinatumomab, SAR3419 (Sanofi), MEDI-551 (MedImmune LLC), Combotox, DT2219ARL (Masonic Cancer Center), MOR-208 (also called XmAb-5574; MorphoSys), XmAb-5871 (Xencor), MDX-1342 (Bristol-Myers Squibb), SGN-CD19A (Seattle Genetics), and AFM11 (Affimed Therapeutics). See, e.g., Hammer. MAbs. 4.5(2012): 571-77. Blinatomomab is a bispecific antibody comprised of two scFvs—one that binds to CD19 and one that binds to CD3. Blinatomomab directs T cells to attack cancer cells. See, e.g., Hammer et al.; Clinical Trial Identifier No. NCT00274742 and NCT01209286. MEDI-551 is a humanized anti-CD19 antibody with a Fc engineered to have enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). See, e.g., Hammer et al.; and Clinical Trial Identifier No. NCT01957579. Combotox is a mixture of immunotoxins that bind to CD19 and CD22. The immunotoxins are made up of scFv antibody fragments fused to a deglycosylated ricin A chain. See, e.g., Hammer et al.; and Herrera et al. J. Pediatr. Hematol. Oncol. 31.12(2009):936-41; Schindler et al. Br. J. Haematol. 154.4(2011):471-6. DT2219ARL is a bispecific immunotoxin targeting CD19 and CD22, comprising two scFvs and a truncated diphtheria toxin. See, e.g., Hammer et al.; and Clinical Trial Identifier No. NCT00889408. SGN-CD19A is an antibody-drug conjugate (ADC) comprised of an anti-CD19 humanized monoclonal antibody linked to a synthetic cytotoxic cell-killing agent, monomethyl auristatin F (MMAF). See, e.g., Hammer et al.; and Clinical Trial Identifier Nos. NCT01786096 and NCT01786135. SAR3419 is an anti-CD19 antibody-drug conjugate (ADC) comprising an anti-CD19 humanized monoclonal antibody conjugated to a maytansine derivative via a cleavable linker. See, e.g., Younes et al. J. Clin. Oncol. 30.2(2012): 2776-82; Hammer et al.; Clinical Trial Identifier No. NCT00549185; and Blanc et al. Clin Cancer Res. 2011; 17:6448-58. XmAb-5871 is an Fc-engineered, humanized anti-CD19 antibody. See, e.g., Hammer et al. MDX-1342 is a human Fc-engineered anti-CD19 antibody with enhanced ADCC. See, e.g., Hammer et al. In embodiments, the antibody molecule is a bispecific anti-CD19 and anti-CD3 molecule. For instance, AFM11 is a bispecific antibody that targets CD19 and CD3. See, e.g., Hammer et al.; and Clinical Trial Identifier No. NCT02106091. In some embodiments, an anti-CD19 antibody described herein is conjugated or otherwise bound to a therapeutic agent, e.g., a chemotherapeutic agent, peptide vaccine (such as that described in Izumoto et al. 2008 J Neurosurg 108:963-971), immunosuppressive agent, or immunoablative agent, e.g., cyclosporin, azathioprine, methotrexate, mycophenolate, FK506, CAMPATH, anti-CD3 antibody, cytoxin, fludarabine, rapamycin, mycophenolic acid, steroid, FR901228, or cytokine.


Exemplary anti-CD19 antibody molecules (including antibodies or fragments or conjugates thereof) can include a scFv, CDRs, or VH and VL chains described in Tables 2, 4, or 5. In an embodiment, the CD19-binding antibody molecule comprises: a light chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a light chain variable region provided in Table 2, or a sequence with 95-99% identity with an amino acid sequence of Table 2; and/or a heavy chain variable region comprising an amino acid sequence having at least one, two or three modifications (e.g., substitutions) but not more than 30, 20 or 10 modifications (e.g., substitutions) of an amino acid sequence of a heavy chain variable region provided in Table 2, or a sequence with 95-99% identity to an amino acid sequence of Table 2. In embodiments, the CD19-binding antibody molecule comprises one or more CDRs (e.g., one each of a HC CDR1, HC CDR2, HC CDR3, LC CDR1, LC CDR2, and LC CDR3) of Table 4 or Table 5, or CDRs having one, two, three, four, five, or six modifications (e.g., substitutions) of one or more of the CDRs. The antibody molecule may be, e.g., an isolated antibody molecule.


In one embodiment, an antigen binding domain against CD19 is an antigen binding portion, e.g., CDRs, of an antigen binding domain described in a Table herein. In one embodiment, a CD19 antigen binding domain can be from any CD19 CAR, e.g., LG-740; U.S. Pat. Nos. 8,399,645; 7,446,190; Xu et al., Leuk Lymphoma. 2013 54(2):255-260(2012); Cruz et al., Blood 122(17):2965-2973 (2013); Brentjens et al., Blood, 118(18):4817-4828 (2011); Kochenderfer et al., Blood 116(20):4099-102 (2010); Kochenderfer et al., Blood 122 (25):4129-39(2013); and 16th Annu Meet Am Soc Gen Cell Ther (ASGCT) (May 15-18, Salt Lake City) 2013, Abst 10, each of which is herein incorporated by reference in its entirety.


In one embodiment, the CAR T cell that specifically binds to CD19 has the USAN designation TISAGENLECLEUCEL-T. CTL019 is made by a gene modification of T cells is mediated by stable insertion via transduction with a self-inactivating, replication deficient Lentiviral (LV) vector containing the CTL019 transgene under the control of the EF-1 alpha promoter. CTL019 can be a mixture of transgene positive and negative T cells that are delivered to the subject on the basis of percent transgene positive T cells.


In one aspect the nucleic acid sequence of a CAR construct of the invention is selected from one or more of SEQ ID NOS:85-96. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:85. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:86. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:87. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:88. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:89. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:90. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:91. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:92. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:93. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:94. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:95. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:96. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:97. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:98. In one aspect the nucleic acid sequence of a CAR construct is SEQ ID NO:99.


Humanization of Murine Anti-CD19 Antibody

Humanization of murine CD19 antibody is desired for the clinical setting, where the mouse-specific residues may induce a human-anti-mouse antigen (HAMA) response in patients who receive CART19 treatment, i.e., treatment with T cells transduced with the CAR19 construct. The production, characterization, and efficacy of humanized CD19 CAR sequences is described in International Application WO2014/153270 which is herein incorporated by reference in its entirety, including Examples 1-5 (p. 115-159), for instance Tables 3, 4, and 5 (p. 125-147).


CAR Constructs, e.g., CD19 CAR Constructs

Of the CD19 CAR constructs described in International Application WO2014/153270, certain sequences are reproduced herein. It is understood that the sequences in this section can also be used in the context of other CARs, e.g., CD22 CARs.


The sequences of the humanized scFv fragments (SEQ ID NOS: 1-12) are provided below in Table 2. Full CAR constructs were generated using SEQ ID NOs: 1-12 with additional sequences, SEQ ID NOs: 13-17, shown below, to generate full CAR constructs with SEQ ID NOs: 31-42.










leader (amino acid sequence) 



(SEQ ID NO: 13)



MALPVTALLLPLALLLHAARP






leader (nucleic acid sequence) 


(SEQ ID NO: 54)



ATGGCCCTGCCTGTGACAGCCCTGCTGCTGCCTCTGGCTCTGCTGCTGCATGCCGCTAGACC






C





CD8 hinge (amino acid sequence) 


(SEQ ID NO: 14)



TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD






CD8 hinge (nucleic acid sequence) 


(SEQ ID NO: 55)



ACCACGACGCCAGCGCCGCGACCACCAACACCGGCGCCCACCATCGCGTCGCAGCCCCTGT






CCCTGCGCCCAGAGGCGTGCCGGCCAGCGGCGGGGGGCGCAGTGCACACGAGGGGGCTGG





ACTTCGCCTGTGAT





CD8 transmembrane (amino acid sequence) 


(SEQ ID NO: 15)



IYIWAPLAGTCGVLLLSLVITLYC






transmembrane (nucleic acid sequence) 


(SEQ ID NO: 56)



ATCTACATCTGGGCGCCCTTGGCCGGGACTTGTGGGGTCCTTCTCCTGTCACTGGTTATCACC






CTTTACTGC





4-1BB Intracellular domain (amino acid sequence) 


(SEQ ID NO: 16)



KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL






4-1BB Intracellular domain (nucleic acid sequence) 


(SEQ ID NO: 60)



AAACGGGGCAGAAAGAAACTCCTGTATATATTCAAACAACCATTTATGAGACCAGTACAAA






CTACTCAAGAGGAAGATGGCTGTAGCTGCCGATTTCCAGAAGAAGAAGAAGGAGGATGTGA





ACTG





CD3 zeta domain (amino acid sequence) 


(SEQ ID NO: 17)



RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNEL






QKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR





CD3 zeta (nucleic acid sequence) 


(SEQ ID NO: 101)



AGAGTGAAGTTCAGCAGGAGCGCAGACGCCCCCGCGTACAAGCAGGGCCAGAACCAGCTCT






ATAACGAGCTCAATCTAGGACGAAGAGAGGAGTACGATGTTTTGGACAAGAGACGTGGCCG





GGACCCTGAGATGGGGGGAAAGCCGAGAAGGAAGAACCCTCAGGAAGGCCTGTACAATGA





ACTGCAGAAAGATAAGATGGCGGAGGCCTACAGTGAGATTGGGATGAAAGGCGAGCGCCG





GAGGGGCAAGGGGCACGATGGCCTTTACCAGGGTCTCAGTACAGCCACCAAGGACACCTAC





GACGCCCTTCACATGCAGGCCCTGCCCCCTCGC





CD3 zeta domain (amino acid sequence; NOM Reference Sequence NM_000734.3) 


(SEQ ID NO: 43)



RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNEL






QKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR





CD3 zeta (nucleic acid sequence; NCBI Reference Sequence NM_000734.3); 


(SEQ ID NO: 44)



AGAGTGAAGTTCAGCAGGAGCGCAGACGCCCCCGCGTACCAGCAGGGCCAG






AACCAGCTCTATAACGAGCTCAATCTAGGACGAAGAGAGGAGTACGATGTTT





TGGACAAGAGACGTGGCCGGGACCCTGAGATGGGGGGAAAGCCGAGAAGGA





AGAACCCTCAGGAAGGCCTGTACAATGAACTGCAGAAAGATAAGATGGCGG





AGGCCTACAGTGAGATTGGGATGAAAGGCGAGCGCCGGAGGGGCAAGGGGC





ACGATGGCCTTTACCAGGGTCTCAGTACAGCCACCAAGGACACCTACGACGC





CCTTCACATGCAGGCCCTGCCCCCTCGC





CD28 domain 


(amino acid sequence, SEQ ID NO: 1317)



RSKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRS






CD28 domain 


(nucleotide sequence, SEQ ID NO: 1318)



AGGAGTAAGAGGAGCAGGCTCCTGCACAGTGACTACATGAACATGACTCCCCGCCG






CCCCGGGCCCACCCGCAAGCATTACCAGCCCTATGCCCCACCACGCGACTTCGCAG





CCTATCGCTCC





Wild-type ICOS domain 


(amino acid sequence, SEQ ID NO: 1319)



TKKKYSSSVHDPNGEYMFMRAVNTAKKSRLTDVTL






Wild-type ICOS domain 


(nucleotide sequence, SEQ ID NO: 1320)



ACAAAAAAGAAGTATTCATCCAGTGTGCACGACCCTAACGGTGAATACATGTTCAT






GAGAGCAGTGAACACAGCCAAAAAATCCAGACTCACAGATGTGACCCTA





Y to F mutant ICOS domain


(amino acid sequence, SEQ ID NO: 1321)



TKKKYSSSVHDPNGEFMFMRAVNTAKKSRLTDVTL






IgG4 Hinge (amino acid sequence) 


(SEQ ID NO: 102)



ESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVE






VHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQ





VYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTV





DKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGKM





IgG4 Hinge (nucleotide sequence) 


(SEQ ID NO: 103)



GAGAGCAAGTACGGCCCTCCCTGCCCCCCTTGCCCTGCCCCCGAGTTCCTGGGCGGACCCAG






CGTGTTCCTGTTCCCCCCCAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCCGAGGTGA





CCTGTGTGGTGGTGGACGTGTCCCAGGAGGACCCCGAGGTCCAGTTCAACTGGTACGTGGA





CGGCGTGGAGGTGCACAACGCCAAGACCAAGCCCCGGGAGGAGCAGTTCAATAGCACCTAC





CGGGTGGTGTCCGTGCTGACCGTGCTGCACCAGGACTGGCTGAACGGCAAGGAATACAAGT





GTAAGGTGTCCAACAAGGGCCTGCCCAGCAGCATCGAGAAAACCATCAGCAAGGCCAAGG





GCCAGCCTCGGGAGCCCCAGGTGTACACCCTGCCCCCTAGCCAAGAGGAGATGACCAAGAA





CCAGGTGTCCCTGACCTGCCTGGTGAAGGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGG





AGAGCAACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCTGTGCTGGACAGCGACGG





CAGCTTCTTCCTGTACAGCCGGCTGACCGTGGACAAGAGCCGGTGGCAGGAGGGCAACGTC





TTTAGCTGCTCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGAGCCTGAGCCT





GTCCCTGGGCAAGATG






These clones all contained a Q/K residue change in the signal domain of the co-stimulatory domain derived from 4-1BB.









TABLE 2 







Humanized CD19 CAR Constructs









Name
SEQ ID
Sequence










CAR 1









CAR1 scF17
1
EIVMTQSPATLSLSPGERATLSCRASQDISKYLNWYQQKPGQAPRLLIYHT


domain

SRLHSGIPARFSGSGSGTDYTLTISSLQPEDFAVYFCQQGNTLPYTFGQGT




KLEIKGGGGSGGGGSGGGGSQVQLQESGPGLVKPSETLSLTCTVSGVSLPD




YGVSWIRQPPGKGLEWIGVIWGSETTYYSSSLKSRVTISKDNSKNQVSLKL




SSVTAADTAVYYCAKHYYYGGSYAMDYWGQGTLVTVSS


103101
61
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR1

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Soluble

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg


scF17-nt

tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaa




gcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagc




ggagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaaggg




tctggaatggattggagtgatttggggctctgagactacttactactcttcatccc




tcaagtcacgcgtcaccatctcaaaggacaactctaagaatcaggtgtcactgaaa




ctgtcatctgtgaccgcagccgacaccgccgtgtactattgcgctaagcattacta




ttatggcgggagctacgcaatggattactggggacagggtactctggtcaccgtgt




ccagccaccaccatcatcaccatcaccat


103101
73


MALPVTALLLPLALLLHAARP
eivmtqspatlslspgeratlscrasqdiskylnw



CAR1

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Soluble

ntlpytfgqgtkleikggggsggggsggggsqvqlqesgpglvkpsetlsltctvs


scF17-aa

gvslpdygvswirqppgkglewigviwgsettyyssslksrvtiskdnsknqvslk




lssvtaadtavyycakhyyyggsyamdywgqgtlvtvsshhhhhhhh


104875
85
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR 

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


1-Full-nt

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg




tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaa




gcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagc




ggagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaaggg




tctggaatggattggagtgatttggggctctgagactacttactactcttcatccc




tcaagtcacgcgtcaccatctcaaaggacaactctaagaatcaggtgtcactgaaa




ctgtcatctgtgaccgcagccgacaccgccgtgtactattgcgctaagcattacta




ttatggcgggagctacgcaatggattactggggacagggtactctggtcaccgtgt




ccagcaccactaccccagcaccgaggccacccaccccggctcctaccatcgcctcc




cagcctctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgca




tacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggta




cttgcggggtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcgg




aagaagctgctgtacatctttaagcaacccttcatgaggcctgtgcagactactca




agaggaggacggctgttcatgccggttcccagaggaggaggaaggcggctgcgaac




tgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcaggggcagaac




cagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaa




gcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaag




agggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagatt




ggtatgaaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggact




cagcaccgccaccaaggacacctatgacgctcttcacatgcaggccctgccgcctc




gg


104875
31
MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlscrasqdiskylnw


CAR 1-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Full-aa



ntlpyt
fgqgtkleikggggsggggsggggsqvqlqesgpglvkpsetlsltctvs





gvslpdygvswirqppgkglewigviwgsettyyssslksrvtiskdnsknqvslk




lssvtaadtavyycakhyyyggsyamdywgqgtlvtvsstttpaprpptpaptias




qplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitlyckrgr




kkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapaykqgqn




qlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeaysei




gmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR 2









CAR2 scFv
2
eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhs


domain

giparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggs




ggggsggggsqvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkgle




wigviwgsettyyqsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyg




gsyamdywgqgtlvtvss


103102
62
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR2-

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Soluble

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg


scFv-nt

tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaa




gcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagc




ggagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaaggg




tctggaatggattggagtgatttggggctctgagactacttactaccaatcatccc




tcaagtcacgcgtcaccatctcaaaggacaactctaagaatcaggtgtcactgaaa




ctgtcatctgtgaccgcagccgacaccgccgtgtactattgcgctaagcattacta




ttatggcgggagctacgcaatggattactggggacagggtactctggtcaccgtgt




ccagccaccaccatcatcaccatcaccat


103102
74


MALPVTALLLPLALLLHAARP
eivmtqspatlslspgeratlscrasqdiskylnw



CAR2-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Soluble

ntlpytfgqgtkleikggggsggggsggggsqvqlqesgpglvkpsetlsltctvs


scFv-aa

gvslpdygvswirqppgkglewigviwgsettyyqsslksrvtiskdnsknqvslk




lssvtaadtavyycakhyyyggsyamdywgqgtlvtvsshhhhhhhh


104876
86
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR 2-

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Full-nt

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg




tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaa




gcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagc




ggagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaaggg




tctggaatggattggagtgatttggggctctgagactacttactaccaatcatccc




tcaagtcacgcgtcaccatctcaaaggacaactctaagaatcaggtgtcactgaaa




ctgtcatctgtgaccgcagccgacaccgccgtgtactattgcgctaagcattacta




ttatggcgggagctacgcaatggattactggggacagggtactctggtcaccgtgt




ccagcaccactaccccagcaccgaggccacccaccccggctcctaccatcgcctcc




cagcctctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgca




tacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggta




cttgcggggtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcgg




aagaagctgctgtacatctttaagcaacccttcatgaggcctgtgcagactactca




agaggaggacggctgttcatgccggttcccagaggaggaggaaggcggctgcgaac




tgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcaggggcagaac




cagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaa




gcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaag




agggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagatt




ggtatgaaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggact




cagcaccgccaccaaggacacctatgacgctcttcacatgcaggccctgccgcctc




gg


104876
32
MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlscrasqdiskylnw


CAR 2-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Full-aa



ntlpyt
fgqgtkleikggggsggggsggggsqvqlqesgpglvkpsetlsltctvs





gvslpdygvswirqppgkglewigviwgsettyyqsslksrvtiskdnsknqvslk




lssvtaadtavyycakhyyyggsyamdywgqgtlvtvsstttpaprpptpaptias




qplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitlyckrgr




kkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapaykqgqn




qlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeaysei




gmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR 3









CAR3 scFv
3
qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgset


domain

tyyssslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgq




gtlvtvssggggsggggsggggseivmtqspatlslspgeratlscrasqdiskyl




nwyqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcq




qgntlpytfgqgtkleik


103104
63
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


3-Soluble

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc


scFv-nt

tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactattcatcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccgaaatcgtgatgacccagagccctgcaaccctgtcc




ctttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaa




atacctcaattggtatcaacagaagccgggacaggcccctaggcttcttatctacc




acacctctcgcctgcatagcgggattcccgcacgctttagcgggtctggaagcggg




accgactacactctgaccatctcatctctccagcccgaggacttcgccgtctactt




ctgccagcagggtaacaccctgccgtacaccttcggccagggcaccaagcttgaga




tcaaacatcaccaccatcatcaccatcac


103104
75


MALPVTALLLPLALLLHAARP
qvqlqesgpglvkpsetlsltctvsgvslpdygvs



CAR 3-

wirqppgkglewigviwgsettyyssslksrvtiskdnsknqvslklssvtaadta


Soluble

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggseivmtqspatls


scFv-aa

lspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsg




tdytltisslqpedfavyfcqqgntlpytfgqgtkleikhhhhhhhh


104877
87
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR 3-

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


Full-nt

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc




tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactattcatcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccgaaatcgtgatgacccagagccctgcaaccctgtcc




ctttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaa




atacctcaattggtatcaacagaagccgggacaggcccctaggcttcttatctacc




acacctctcgcctgcatagcgggattcccgcacgctttagcgggtctggaagcggg




accgactacactctgaccatctcatctctccagcccgaggacttcgccgtctactt




ctgccagcagggtaacaccctgccgtacaccttcggccagggcaccaagcttgaga




tcaaaaccactactcccgctccaaggccacccacccctgccccgaccatcgcctct




cagccgctttccctgcgtccggaggcatgtagacccgcagctggtggggccgtgca




tacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggta




cttgcggggtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcgg




aagaagctgctgtacatctttaagcaacccttcatgaggcctgtgcagactactca




agaggaggacggctgttcatgccggttcccagaggaggaggaaggcggctgcgaac




tgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcaggggcagaac




cagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaa




gcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaag




agggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagatt




ggtatgaaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggact




cagcaccgccaccaaggacacctatgacgctcttcacatgcaggccctgccgcctc




gg


104877
33
MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslpdygvs


CAR 3-

wirqppgkglewigviwgsettyyssslksrvtiskdnsknqvslklssvtaadta


Full-aa

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggseivmtqspatls




lspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsg




tdytltisslqpedfavyfcqqgntlpytfgqgtkleiktttpaprpptpaptias




qplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitlyckrgr




kkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapaykqgqn




qlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeaysei




gmkgerrrgkghdglycolstatkdtydalhmqalppr










CAR4









CAR4 scFv
4
qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgset


domain

tyyqsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgq




gtlvtvssggggsggggsggggseivmtqspatlslspgeratlscrasqdiskyl




nwyqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcq




qgntlpytfgqgtkleik


103106
64
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR4-

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


Soluble

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc


scFv-nt

tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactatcaatcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccgaaatcgtgatgacccagagccctgcaaccctgtcc




ctttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaa




atacctcaattggtatcaacagaagccgggacaggcccctaggcttcttatctacc




acacctctcgcctgcatagcgggattcccgcacgctttagcgggtctggaagcggg




accgactacactctgaccatctcatctctccagcccgaggacttcgccgtctactt




ctgccagcagggtaacaccctgccgtacaccttcggccagggcaccaagcttgaga




tcaaacatcaccaccatcatcaccatcac


103106
76


MALPVTALLLPLALLLHAARP
qvqlqesgpglvkpsetlsltctvsgvslpdygvs



CAR4-

wirqppgkglewigviwgsettyyqsslksrvtiskdnsknqvslklssvtaadta


Soluble

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggseivmtqspatls


scFv-aa

lspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsg




tdytltisslqpedfavyfcqqgntlpytfgqgtkleikhhhhhhhh


104878
88
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR 4-

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


Full-nt

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc




tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactatcaatcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccgaaatcgtgatgacccagagccctgcaaccctgtcc




ctttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaa




atacctcaattggtatcaacagaagccgggacaggcccctaggcttcttatctacc




acacctctcgcctgcatagcgggattcccgcacgctttagcgggtctggaagcggg




accgactacactctgaccatctcatctctccagcccgaggacttcgccgtctactt




ctgccagcagggtaacaccctgccgtacaccttcggccagggcaccaagcttgaga




tcaaaaccactactcccgctccaaggccacccacccctgccccgaccatcgcctct




cagccgctttccctgcgtccggaggcatgtagacccgcagctggtggggccgtgca




tacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggta




cttgcggggtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcgg




aagaagctgctgtacatctttaagcaacccttcatgaggcctgtgcagactactca




agaggaggacggctgttcatgccggttcccagaggaggaggaaggcggctgcgaac




tgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcaggggcagaac




cagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaa




gcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaag




agggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagatt




ggtatgaaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggact




cagcaccgccaccaaggacacctatgacgctcttcacatgcaggccctgccgcctc




gg


104878
34
MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslpdygvs


CAR 4-

wirqppgkglewigviwgsettyygsslksrvtiskdnsknqvslklssvtaadta


Full-aa

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggseivmtqspatls




lspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsg




tdytltisslqpedfavyfcqqgntlpytfgqgtkleiktttpaprpptpaptias




qplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitlyckrgr




kkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapaykqgqn




qlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeaysei




gmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR 5









CAR5 scFv
5
eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhs


domain

giparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggs




ggggsggggsggggsqvqlqesgpglvkpsetlsltctvsgvslpdygvswirqpp




gkglewigviwgsettyyssslksrvtiskdnsknqvslklssvtaadtavyycak




hyyyggsyamdywgqgtlvtvss


99789
65
atggccctcccagtgaccgctctgctgctgcctctcgcacttcttctccatgccgc


CAR5-

tcggcctgagatcgtcatgacccaaagccccgctaccctgtccctgtcacccggcg


Soluble

agagggcaaccctttcatgcagggccagccaggacatttctaagtacctcaactgg


scFv-nt

tatcagcagaagccagggcaggctcctcgcctgctgatctaccacaccagccgcct




ccacagcggtatccccgccagattttccgggagcgggtctggaaccgactacaccc




tcaccatctcttctctgcagcccgaggatttcgccgtctatttctgccagcagggg




aatactctgccgtacaccttcggtcaaggtaccaagctggaaatcaagggaggcgg




aggatcaggcggtggcggaagcggaggaggtggctccggaggaggaggttcccaag




tgcagcttcaagaatcaggacccggacttgtgaagccatcagaaaccctctccctg




acttgtaccgtgtccggtgtgagcctccccgactacggagtctcttggattcgcca




gcctccggggaagggtcttgaatggattggggtgatttggggatcagagactactt




actactcttcatcacttaagtcacgggtcaccatcagcaaagataatagcaagaac







caagtgtcacttaagctgtcatctgtgaccgccgctgacaccgccgtgtactattg




tgccaaacattactattacggagggtcttatgctatggactactggggacagggga




ccctggtgactgtctctagccatcaccatcaccaccatcatcac


99789
77


MALPVTALLLPLALLLHAARP
eivmtqspatlslspgeratlscrasqdiskylnw



CAR5-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Soluble

ntlpytfgqgtkleikggggsggggsggggsggggsqvqlqesgpglvkpsetlsl


scFv-aa

tctvsgvslpdygvswirqppgkglewigviwgsettyyssslksrvtiskdnskn




qvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtvsshhhhhhhh


104879
89
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR 5-

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Full-nt

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg




tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagcggcggaggcgggagccagg




tccaactccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactg




acttgtactgtgagcggagtgtctctccccgattacggggtgtcttggatcagaca




gccaccggggaagggtctggaatggattggagtgatttggggctctgagactactt




actactcttcatccctcaagtcacgcgtcaccatctcaaaggacaactctaagaat




caggtgtcactgaaactgtcatctgtgaccgcagccgacaccgccgtgtactattg




cgctaagcattactattatggcgggagctacgcaatggattactggggacagggta




ctctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggct




cctaccatcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagc




tggtggggccgtgcatacccggggtcttgacttcgcctgcgatatctacatttggg




cccctctggctggtacttgcggggtcctgctgctttcactcgtgatcactctttac




tgtaagcgcggtcggaagaagctgctgtacatctttaagcaacccttcatgaggcc




tgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg




aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctac




aagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagta




cgacgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgca




gaaagaatccccaagagggcctgtacaacgagctccaaaaggataagatggcagaa




gcctatagcgagattggtatgaaaggggaacgcagaagaggcaaaggccacgacgg




actgtaccagggactcagcaccgccaccaaggacacctatgacgctcttcacatgc




aggccctgccgcctcgg


104879
35
MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlscrasqdiskylnw


CAR 5-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Full-aa



ntlpyt
fgqgtkleikggggsggggsggggsggggsqvqlqesgpglvkpsetlsl





tctvsgvslpdygvswirqppgkglewigviwgsettyyssslksrvtiskdnskn




qvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtvsstttpaprpptpa




ptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitly




ckrgrkkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapay




kqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmae




ayseigmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR 6









CAR6-
6
eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhs


scF17

giparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggs


domain

ggggsggggsggggsqvqlqesgpglvkpsetlsltctvsgvslpdygvswirqpp




gkglewigviwgsettyyqsslksrvtiskdnsknqvslklssvtaadtavyycak




hyyyggsyamdywgqgtlvtvss


99790
66
atggccctcccagtgaccgctctgctgctgcctctcgcacttcttctccatgccgc


CAR6-

tcggcctgagatcgtcatgacccaaagccccgctaccctgtccctgtcacccggcg


Soluble

agagggcaaccctttcatgcagggccagccaggacatttctaagtacctcaactgg


scFv-nt

tatcagcagaagccagggcaggctcctcgcctgctgatctaccacaccagccgcct




ccacagcggtatccccgccagattttccgggagcgggtctggaaccgactacaccc




tcaccatctcttctctgcagcccgaggatttcgccgtctatttctgccagcagggg




aatactctgccgtacaccttcggtcaaggtaccaagctggaaatcaagggaggcgg




aggatcaggcggtggcggaagcggaggaggtggctccggaggaggaggttcccaag




tgcagcttcaagaatcaggacccggacttgtgaagccatcagaaaccctctccctg




acttgtaccgtgtccggtgtgagcctccccgactacggagtctcttggattcgcca




gcctccggggaagggtcttgaatggattggggtgatttggggatcagagactactt




actaccagtcatcacttaagtcacgggtcaccatcagcaaagataatagcaagaac




caagtgtcacttaagctgtcatctgtgaccgccgctgacaccgccgtgtactattg




tgccaaacattactattacggagggtcttatgctatggactactggggacagggga




ccctggtgactgtctctagccatcaccatcaccaccatcatcac


99790
78


MALPVTALLLPLALLLHAARP
eivmtqspatlslspgeratlscrasqdiskylnw



CAR6-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Soluble

ntlpytfgqgtkleikggggsggggsggggsggggsqvqlqesgpglvkpsetlsl


scFv-aa

tctvsgvslpdygvswirqppgkglewigviwgsettyyqsslksrvtiskdnskn




qvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtvsshhhhhhhh


104880
90
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR6-

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Full-nt

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg




tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagcggaggcggagggagccagg




tccaactccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactg




acttgtactgtgagcggagtgtctctccccgattacggggtgtcttggatcagaca




gccaccggggaagggtctggaatggattggagtgatttggggctctgagactactt




actaccaatcatccctcaagtcacgcgtcaccatctcaaaggacaactctaagaat




caggtgtcactgaaactgtcatctgtgaccgcagccgacaccgccgtgtactattg




cgctaagcattactattatggcgggagctacgcaatggattactggggacagggta




ctctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggct




cctaccatcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagc




tggtggggccgtgcatacccggggtcttgacttcgcctgcgatatctacatttggg




cccctctggctggtacttgcggggtcctgctgctttcactcgtgatcactctttac




tgtaagcgcggtcggaagaagctgctgtacatctttaagcaacccttcatgaggcc




tgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg




aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctac




aagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagta




cgacgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgca




gaaagaatccccaagagggcctgtacaacgagctccaaaaggataagatggcagaa




gcctatagcgagattggtatgaaaggggaacgcagaagaggcaaaggccacgacgg




actgtaccagggactcagcaccgccaccaaggacacctatgacgctcttcacatgc




aggccctgccgcctcgg


104880
36
MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlscrasqdiskylnw


CAR6-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Full-aa



ntlpyt
fgqgtkleikggggsggggsggggsggggsqvqlqesgpglvkpsetlsl





tctvsgvslpdygvswirqppgkglewigviwgsettyygsslksrvtiskdnskn




qvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtvsstttpaprpptpa




ptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitly




ckrgrkkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapay




kqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmae




ayseigmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR7









CAR7 scFv
7
qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgset


domain

tyyssslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgq




gtlvtvssggggsggggsggggsggggseivmtqspatlslspgeratlscrasqd




iskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfa




vyfcqqgntlpytfgqgtkleik


100796
67
atggcactgcctgtcactgccctcctgctgcctctggccctccttctgcatgccgc


CAR7-

caggccccaagtccagctgcaagagtcaggacccggactggtgaagccgtctgaga


Soluble

ctctctcactgacttgtaccgtcagcggcgtgtccctccccgactacggagtgtca


scFv-nt

tggatccgccaacctcccgggaaagggcttgaatggattggtgtcatctggggttc




tgaaaccacctactactcatcttccctgaagtccagggtgaccatcagcaaggata




attccaagaaccaggtcagccttaagctgtcatctgtgaccgctgctgacaccgcc




gtgtattactgcgccaagcactactattacggaggaagctacgctatggactattg




gggacagggcactctcgtgactgtgagcagcggcggtggagggtctggaggtggag




gatccggtggtggtgggtcaggcggaggagggagcgagattgtgatgactcagtca




ccagccaccctttctctttcacccggcgagagagcaaccctgagctgtagagccag




ccaggacatttctaagtacctcaactggtatcagcaaaaaccggggcaggcccctc




gcctcctgatctaccatacctcacgccttcactctggtatccccgctcggtttagc




ggatcaggatctggtaccgactacactctgaccatttccagcctgcagccagaaga




tttcgcagtgtatttctgccagcagggcaatacccttccttacaccttcggtcagg




gaaccaagctcgaaatcaagcaccatcaccatcatcaccaccat


100796
79


MALPVTALLLPLALLLHAARP
qvqlqesgpglvkpsetlsltctvsgvslpdygvs



CAR7-

wirqppgkglewigviwgsettyyssslksrvtiskdnsknqvslklssvtaadta


Soluble

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggsggggseivmtqs


scFv-aa

patlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfs




gsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikhhhhhhhh


104881
91
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR 7

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


Full-nt

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc




tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactattcatcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccggaggtggcggaagcgaaatcgtgatgacccagagc




cctgcaaccctgtccctttctcccggggaacgggctaccctttcttgtcgggcatc




acaagatatctcaaaatacctcaattggtatcaacagaagccgggacaggccccta




ggcttcttatctaccacacctctcgcctgcatagcgggattcccgcacgctttagc




gggtctggaagcgggaccgactacactctgaccatctcatctctccagcccgagga




cttcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggccagg




gcaccaagcttgagatcaaaaccactactcccgctccaaggccacccacccctgcc




ccgaccatcgcctctcagccgctttccctgcgtccggaggcatgtagacccgcagc




tggtggggccgtgcatacccggggtcttgacttcgcctgcgatatctacatttggg




cccctctggctggtacttgcggggtcctgctgctttcactcgtgatcactctttac




tgtaagcgcggtcggaagaagctgctgtacatctttaagcaacccttcatgaggcc




tgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg




aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctac




aagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagta




cgacgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgca




gaaagaatccccaagagggcctgtacaacgagctccaaaaggataagatggcagaa




gcctatagcgagattggtatgaaaggggaacgcagaagaggcaaaggccacgacgg




actgtaccagggactcagcaccgccaccaaggacacctatgacgctcttcacatgc




aggccctgccgcctcgg


104881
37
MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslpdygvs


CAR 7

wirqppgkglewigviwgsettyyssslksrvtiskdnsknqvslklssvtaadta


Full-aa

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggsggggseivmtqs




patlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfs




gsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleiktttpaprpptpa




ptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitly




ckrgrkkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapay




kqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmae




ayseigmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR 8









CAR8 scFv
8
qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgset


domain

tyyqsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgq




gtlvtvssggggsggggsggggsggggseivmtqspatlslspgeratlscrasqd




iskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfa




vyfcqqgntlpytfgqgtkleik


100798
68
atggcactgcctgtcactgccctcctgctgcctctggccctccttctgcatgccgc


CAR8-

caggccccaagtccagctgcaagagtcaggacccggactggtgaagccgtctgaga


Soluble

ctctctcactgacttgtaccgtcagcggcgtgtccctccccgactacggagtgtca


scFv-nt

tggatccgccaacctcccgggaaagggcttgaatggattggtgtcatctggggttc




tgaaaccacctactaccagtcttccctgaagtccagggtgaccatcagcaaggata




attccaagaaccaggtcagccttaagctgtcatctgtgaccgctgctgacaccgcc




gtgtattactgcgccaagcactactattacggaggaagctacgctatggactattg




gggacagggcactctcgtgactgtgagcagcggcggtggagggtctggaggtggag




gatccggtggtggtgggtcaggcggaggagggagcgagattgtgatgactcagtca




ccagccaccctttctctttcacccggcgagagagcaaccctgagctgtagagccag




ccaggacatttctaagtacctcaactggtatcagcaaaaaccggggcaggcccctc




gcctcctgatctaccatacctcacgccttcactctggtatccccgctcggtttagc




ggatcaggatctggtaccgactacactctgaccatttccagcctgcagccagaaga




tttcgcagtgtatttctgccagcagggcaatacccttccttacaccttcggtcagg




gaaccaagctcgaaatcaagcaccatcaccatcatcatcaccac


100798
80


MALPVTALLLPLALLLHAARP
qvqlqesgpglvkpsetlsltctvsgvslpdygvs



CAR8-

wirqppgkglewigviwgsettyyqsslksrvtiskdnsknqvslklssvtaadta


Soluble

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggsggggseivmtqs


scFv-aa

patlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfs




gsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikhhhhhhhh


104882
92
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR 8-

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


Full-nt

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc




tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactatcaatcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccggaggcggtgggtcagaaatcgtgatgacccagagc




cctgcaaccctgtccctttctcccggggaacgggctaccctttcttgtcgggcatc




acaagatatctcaaaatacctcaattggtatcaacagaagccgggacaggccccta




ggcttcttatctaccacacctctcgcctgcatagcgggattcccgcacgctttagc




gggtctggaagcgggaccgactacactctgaccatctcatctctccagcccgagga




cttcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggccagg




gcaccaagcttgagatcaaaaccactactcccgctccaaggccacccacccctgcc




ccgaccatcgcctctcagccgctttccctgcgtccggaggcatgtagacccgcagc




tggtggggccgtgcatacccggggtcttgacttcgcctgcgatatctacatttggg




cccctctggctggtacttgcggggtcctgctgctttcactcgtgatcactctttac




tgtaagcgcggtcggaagaagctgctgtacatctttaagcaacccttcatgaggcc




tgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg




aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctac




aagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagta




cgacgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgca




gaaagaatccccaagagggcctgtacaacgagctccaaaaggataagatggcagaa




gcctatagcgagattggtatgaaaggggaacgcagaagaggcaaaggccacgacgg




actgtaccagggactcagcaccgccaccaaggacacctatgacgctcttcacatgc




aggccctgccgcctcgg


104882
38
MALPVTALLLPLALLLHAARPqvqlqesgpglvkpsetlsltctvsgvslpdygvs


CAR 8-

wirqppgkglewigviwgsettyyqsslksrvtiskdnsknqvslklssvtaadta


Full-aa

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggsggggseivmtqs




patlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfs




gsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleiktttpaprpptpa




ptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitly




ckrgrkkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapay




kqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmae




ayseigmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR 9









CAR9 scFv
9
eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhs


domain

giparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggs




ggggsggggsggggsqvqlqesgpglvkpsetlsltctvsgvslpdygvswirqpp




gkglewigviwgsettyynsslksrvtiskdnsknqvslklssvtaadtavyycak




hyyyggsyamdywgqgtlvtvss


99789
69
atggccctcccagtgaccgctctgctgctgcctctcgcacttcttctccatgccgc


CAR9-

tcggcctgagatcgtcatgacccaaagccccgctaccctgtccctgtcacccggcg


Soluble

agagggcaaccctttcatgcagggccagccaggacatttctaagtacctcaactgg


scFv-nt

tatcagcagaagccagggcaggctcctcgcctgctgatctaccacaccagccgcct




ccacagcggtatccccgccagattttccgggagcgggtctggaaccgactacaccc




tcaccatctcttctctgcagcccgaggatttcgccgtctatttctgccagcagggg




aatactctgccgtacaccttcggtcaaggtaccaagctggaaatcaagggaggcgg




aggatcaggcggtggcggaagcggaggaggtggctccggaggaggaggttcccaag




tgcagcttcaagaatcaggacccggacttgtgaagccatcagaaaccctctccctg




acttgtaccgtgtccggtgtgagcctccccgactacggagtctcttggattcgcca




gcctccggggaagggtcttgaatggattggggtgatttggggatcagagactactt




actacaattcatcacttaagtcacgggtcaccatcagcaaagataatagcaagaac




caagtgtcacttaagctgtcatctgtgaccgccgctgacaccgccgtgtactattg




tgccaaacattactattacggagggtcttatgctatggactactggggacagggga




ccctggtgactgtctctagccatcaccatcaccaccatcatcac


99789
81


MALPVTALLLPLALLLHAARP
eivmtqspatlslspgeratlscrasqdiskylnw



CAR9-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Soluble

ntlpytfgqgtkleikggggsggggsggggsggggsqvqlqesgpglvkpsetlsl


scFv-aa

tctvsgvslpdygvswirqppgkglewigviwgsettyynsslksrvtiskdnskn




qvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtvsshhhhhhhh


105974
93
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR 9-

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Full-nt

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg




tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagcggaggcggtgggagccagg




tccaactccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactg




acttgtactgtgagcggagtgtctctccccgattacggggtgtcttggatcagaca




gccaccggggaagggtctggaatggattggagtgatttggggctctgagactactt




actacaactcatccctcaagtcacgcgtcaccatctcaaaggacaactctaagaat




caggtgtcactgaaactgtcatctgtgaccgcagccgacaccgccgtgtactattg




cgctaagcattactattatggcgggagctacgcaatggattactggggacagggta




ctctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggct




cctaccatcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagc




tggtggggccgtgcatacccggggtcttgacttcgcctgcgatatctacatttggg




cccctctggctggtacttgcggggtcctgctgctttcactcgtgatcactctttac




tgtaagcgcggtcggaagaagctgctgtacatctttaagcaacccttcatgaggcc




tgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg




aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctac




aagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagta




cgacgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgca




gaaagaatccccaagagggcctgtacaacgagctccaaaaggataagatggcagaa




gcctatagcgagattggtatgaaaggggaacgcagaagaggcaaaggccacgacgg




actgtaccagggactcagcaccgccaccaaggacacctatgacgctcttcacatgc




aggccctgccgcctcgg


105974
39
MALPVTALLLPLALLLHAARPeivmtqspatlslspgeratlscrasqdiskylnw


CAR 9-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Full-aa



ntlpyt
fgqgtkleikggggsggggsggggsggggsqvqlqesgpglvkpsetlsl





tctvsgvslpdygvswirqppgkglewigviwgsettyynsslksrvtiskdnskn




qvslklssvtaadtavyycakhyyyggsyamdywgqgtlvtvsstttpaprpptpa




ptiasqplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitly




ckrgrkkllyifkqpfmrpvqttgeedgcscrfpeeeeggcelrvkfsrsadapay




kqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmae




ayseigmkgerrrgkghdglyqglstatkdtydalhmqalppr










CAR10









CAR10
10
qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgset


scFv

tyynsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgq


domain

gtlvtvssggggsggggsggggsggggseivmtqspatlslspgeratlscrasqd




iskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfa




vyfcqqgntlpytfgqgtkleik


100796
70
atggcactgcctgtcactgccctcctgctgcctctggccctccttctgcatgccgc


CAR10-

caggccccaagtccagctgcaagagtcaggacccggactggtgaagccgtctgaga


Soluble

ctctctcactgacttgtaccgtcagcggcgtgtccctccccgactacggagtgtca


scFv-nt

tggatccgccaacctcccgggaaagggcttgaatggattggtgtcatctggggttc




tgaaaccacctactacaactcttccctgaagtccagggtgaccatcagcaaggata




attccaagaaccaggtcagccttaagctgtcatctgtgaccgctgctgacaccgcc




gtgtattactgcgccaagcactactattacggaggaagctacgctatggactattg




gggacagggcactctcgtgactgtgagcagcggcggtggagggtctggaggtggag




gatccggtggtggtgggtcaggcggaggagggagcgagattgtgatgactcagtca




ccagccaccctttctctttcacccggcgagagagcaaccctgagctgtagagccag




ccaggacatttctaagtacctcaactggtatcagcaaaaaccggggcaggcccctc




gcctcctgatctaccatacctcacgccttcactctggtatccccgctcggtttagc




ggatcaggatctggtaccgactacactctgaccatttccagcctgcagccagaaga




tttcgcagtgtatttctgccagcagggcaatacccttccttacaccttcggtcagg




gaaccaagctcgaaatcaagcaccatcaccatcatcaccaccat


100796
82


MALPVTALLLPLALLLHAARP
qvqlqesgpglvkpsetlsltctvsgvslpdygvs



CAR10-

wirqppgkglewigviwgsettyynsslksrvtiskdnsknqvslklssvtaadta


Soluble

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggsggggseivmtqs


scFv-aa

patlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfs




gsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikhhhhhhhh


105975
94
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR 10

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Full-nt

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg




tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagcggaggcggtgggagccagg




tccaactccaagaaagcggaccgggtcttgtgaagccatcagaaactctttcactg




acttgtactgtgagcggagtgtctctccccgattacggggtgtcttggatcagaca




gccaccggggaagggtctggaatggattggagtgatttggggctctgagactactt




actacaactcatccctcaagtcacgcgtcaccatctcaaaggacaactctaagaat




caggtgtcactgaaactgtcatctgtgaccgcagccgacaccgccgtgtactattg




cgctaagcattactattatggcgggagctacgcaatggattactggggacagggta




ctctggtcaccgtgtccagcaccactaccccagcaccgaggccacccaccccggct




cctaccatcgcctcccagcctctgtccctgcgtccggaggcatgtagacccgcagc




tggtggggccgtgcatacccggggtcttgacttcgcctgcgatatctacatttggg




cccctctggctggtacttgcggggtcctgctgctttcactcgtgatcactctttac




tgtaagcgcggtcggaagaagctgctgtacatctttaagcaacccttcatgaggcc




tgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg




aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctac




aagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagta




cgacgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgca




gaaagaatccccaagagggcctgtacaacgagctccaaaaggataagatggcagaa




gcctatagcgagattggtatgaaaggggaacgcagaagaggcaaaggccacgacgg




actgtaccagggactcagcaccgccaccaaggacacctatgacgctcttcacatgc




aggccctgccgcctcgg


105975
40
MALPVTALLLPLALLLHAARPEIVMTQSPATLSLSPGERATLSCRASQDISKYLNW


CAR 10

YQQKPGQAPRLLIYHTSRLHSGIPARFSGSGSGTDYTLTISSLQPEDFAVYFCQQG


Full-aa



NTLPYT
FGQGTKLEIKGGGGSGGGGSGGGGSGGGGSQVQLQESGPGLVKPSETLSL





TCTVSGVSLPDYGVSWIRQPPGKGLEWIGVIWGSETTYYNSSLKSRVTISKDNSKN




QVSLKLSSVTAADTAVYYCAKHYYYGGSYAMDYWGQGTLVTVSSTTTPAPRPPTPA




PTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLY




CKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAY




KQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAE




AYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR










CAR11









CAR11
11
eivmtqspatlslspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhs


scFv

giparfsgsgsgtdytltisslqpedfavyfcqqgntlpytfgqgtkleikggggs


domain

ggggsggggsqvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkgle




wigviwgsettyynsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyg




gsyamdywgqgtlvtvss


103101
71
Atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR11-

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Soluble

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg


scFv-nt

tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaa




gcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagc




ggagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaaggg




tctggaatggattggagtgatttggggctctgagactacttactacaattcatccc




tcaagtcacgcgtcaccatctcaaaggacaactctaagaatcaggtgtcactgaaa




ctgtcatctgtgaccgcagccgacaccgccgtgtactattgcgctaagcattacta




ttatggcgggagctacgcaatggattactggggacagggtactctggtcaccgtgt




ccagccaccaccatcatcaccatcaccat


103101
83


MALPVTALLLPLALLLHAARP
eivmtqspatlslspgeratlscrasqdiskylnw



CAR11-

yqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcqqg


Soluble

ntlpytfgqgtkleikggggsggggsggggsqvqlqesgpglvkpsetlsltctvs


scFv-aa

gvslpdygvswirqppgkglewigviwgsettyynsslksrvtiskdnsknqvslk




lssvtaadtavyycakhyyyggsyamdywgqgtlvtvsshhhhhhhh


105976
95
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR 11

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


Full-nt

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc




tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactataactcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccggaggtggcggaagcgaaatcgtgatgacccagagc




cctgcaaccctgtccctttctcccggggaacgggctaccctttcttgtcgggcatc




acaagatatctcaaaatacctcaattggtatcaacagaagccgggacaggccccta




ggcttcttatctaccacacctctcgcctgcatagcgggattcccgcacgctttagc




gggtctggaagcgggaccgactacactctgaccatctcatctctccagcccgagga




cttcgccgtctacttctgccagcagggtaacaccctgccgtacaccttcggccagg




gcaccaagcttgagatcaaaaccactactcccgctccaaggccacccacccctgcc




ccgaccatcgcctctcagccgctttccctgcgtccggaggcatgtagacccgcagc




tggtggggccgtgcatacccggggtcttgacttcgcctgcgatatctacatttggg




cccctctggctggtacttgcggggtcctgctgctttcactcgtgatcactctttac




tgtaagcgcggtcggaagaagctgctgtacatctttaagcaacccttcatgaggcc




tgtgcagactactcaagaggaggacggctgttcatgccggttcccagaggaggagg




aaggcggctgcgaactgcgcgtgaaattcagccgcagcgcagatgctccagcctac




aagcaggggcagaaccagctctacaacgaactcaatcttggtcggagagaggagta




cgacgtgctggacaagcggagaggacgggacccagaaatgggcgggaagccgcgca




gaaagaatccccaagagggcctgtacaacgagctccaaaaggataagatggcagaa




gcctatagcgagattggtatgaaaggggaacgcagaagaggcaaaggccacgacgg




actgtaccagggactcagcaccgccaccaaggacacctatgacgctcttcacatgc




aggccctgccgcctcgg


105976
41
MALPVTALLLPLALLLHAARPQVQLQESGPGLVKPSETLSLTCTVSGVSLPDYGVS


CAR 11

WIRQPPGKGLEWIGVIWGSETTYYNSSLKSRVTISKDNSKNQVSLKLSSVTAADTA


Full-aa

VYYCAKHYYYGGSYAMDYWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVMTQS




PATLSLSPGERATLSCRASQDISKYLNWYQQKPGQAPRLLIYHTSRLHSGIPARFS




GSGSGTDYTLTISSLQPEDFAVYFCQQGNTLPYTFGQGTKLEIKTTTPAPRPPTPA




PTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLY




CKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAY




KQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAE




AYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR










CAR12









CAR12
12
qvqlqesgpglvkpsetlsltctvsgvslpdygvswirqppgkglewigviwgset


scFv

tyynsslksrvtiskdnsknqvslklssvtaadtavyycakhyyyggsyamdywgq


domain

gtlvtvssggggsggggsggggseivmtqspatlslspgeratlscrasqdiskyl




nwyqqkpgqaprlliyhtsrlhsgiparfsgsgsgtdytltisslqpedfavyfcq




qgntlpytfgqgtkleik


103104
72
atggctctgcccgtgaccgcactcctcctgccactggctctgctgcttcacgccgc


CAR12-

tcgcccacaagtccagcttcaagaatcagggcctggtctggtgaagccatctgaga


Soluble

ctctgtccctcacttgcaccgtgagcggagtgtccctcccagactacggagtgagc


scFv-nt

tggattagacagcctcccggaaagggactggagtggatcggagtgatttggggtag




cgaaaccacttactataactcttccctgaagtcacgggtcaccatttcaaaggata




actcaaagaatcaagtgagcctcaagctctcatcagtcaccgccgctgacaccgcc




gtgtattactgtgccaagcattactactatggagggtcctacgccatggactactg




gggccagggaactctggtcactgtgtcatctggtggaggaggtagcggaggaggcg




ggagcggtggaggtggctccgaaatcgtgatgacccagagccctgcaaccctgtcc




ctttctcccggggaacgggctaccctttcttgtcgggcatcacaagatatctcaaa




atacctcaattggtatcaacagaagccgggacaggcccctaggcttcttatctacc




acacctctcgcctgcatagcgggattcccgcacgctttagcgggtctggaagcggg




accgactacactctgaccatctcatctctccagcccgaggacttcgccgtctactt




ctgccagcagggtaacaccctgccgtacaccttcggccagggcaccaagcttgaga




tcaaacatcaccaccatcatcaccatcac


103104
84


MALPVTALLLPLALLLHAARP
qvqlqesgpglvkpsetlsltctvsgvslpdygvs



CAR12-

wirqppgkglewigviwgsettyynsslksrvtiskdnsknqvslklssvtaadta


Soluble

vyycakhyyyggsyamdywgqgtlvtvssggggsggggsggggseivmtqspatls


scFv-aa

lspgeratlscrasqdiskylnwyqqkpgqaprlliyhtsrlhsgiparfsgsgsg




tdytltisslqpedfavyfcqqgntlpytfgqgtkleikhhhhhhhh


105977
96
atggccctccctgtcaccgccctgctgcttccgctggctcttctgctccacgccgc


CAR 12 

tcggcccgaaattgtgatgacccagtcacccgccactcttagcctttcacccggtg


Full-nt

agcgcgcaaccctgtcttgcagagcctcccaagacatctcaaaataccttaattgg




tatcaacagaagcccggacaggctcctcgccttctgatctaccacaccagccggct




ccattctggaatccctgccaggttcagcggtagcggatctgggaccgactacaccc




tcactatcagctcactgcagccagaggacttcgctgtctatttctgtcagcaaggg




aacaccctgccctacacctttggacagggcaccaagctcgagattaaaggtggagg




tggcagcggaggaggtgggtccggcggtggaggaagccaggtccaactccaagaaa




gcggaccgggtcttgtgaagccatcagaaactctttcactgacttgtactgtgagc




ggagtgtctctccccgattacggggtgtcttggatcagacagccaccggggaaggg




tctggaatggattggagtgatttggggctctgagactacttactacaactcatccc




tcaagtcacgcgtcaccatctcaaaggacaactctaagaatcaggtgtcactgaaa




ctgtcatctgtgaccgcagccgacaccgccgtgtactattgcgctaagcattacta




ttatggcgggagctacgcaatggattactggggacagggtactctggtcaccgtgt




ccagcaccactaccccagcaccgaggccacccaccccggctcctaccatcgcctcc




cagcctctgtccctgcgtccggaggcatgtagacccgcagctggtggggccgtgca




tacccggggtcttgacttcgcctgcgatatctacatttgggcccctctggctggta




cttgcggggtcctgctgctttcactcgtgatcactctttactgtaagcgcggtcgg




aagaagctgctgtacatctttaagcaacccttcatgaggcctgtgcagactactca




agaggaggacggctgttcatgccggttcccagaggaggaggaaggcggctgcgaac




tgcgcgtgaaattcagccgcagcgcagatgctccagcctacaagcaggggcagaac




cagctctacaacgaactcaatcttggtcggagagaggagtacgacgtgctggacaa




gcggagaggacgggacccagaaatgggcgggaagccgcgcagaaagaatccccaag




agggcctgtacaacgagctccaaaaggataagatggcagaagcctatagcgagatt




ggtatgaaaggggaacgcagaagaggcaaaggccacgacggactgtaccagggact




cagcaccgccaccaaggacacctatgacgctcttcacatgcaggccctgccgcctc




gg


105977
42
MALPVTALLLPLALLLHAARPEIVMTQSPATLSLSPGERATLSCRASQDISKYLNW


CAR 12-

YQQKPGQAPRLLIYHTSRLHSGIPARFSGSGSGTDYTLTISSLQPEDFAVYFCQQG


Full-aa



NTLPYT
FGQGTKLEIKGGGGSGGGGSGGGGSQVQLQESGPGLVKPSETLSLTCTVS





GVSLPDYGVSWIRQPPGKGLEWIGVIWGSETTYYNSSLKSRVTISKDNSKNQVSLK




LSSVTAADTAVYYCAKHYYYGGSYAMDYWGQGTLVTVSSTTTPAPRPPTPAPTIAS




QPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGR




KKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQN




QLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEI




GMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR
















TABLE 3





Murine CD19 CAR Constructs


CTL019

















CTL019-
 97
Atggccctgcccgtcaccgctctgctgctgccccttgctctgcttcttcatgcagc


Soluble

aaggccggacatccagatgacccaaaccacctcatccctctctgcctctcttggag


scFv-

acagggtgaccatttcttgtcgcgccagccaggacatcagcaagtatctgaactgg


Histag-

tatcagcagaagccggacggaaccgtgaagctcctgatctaccatacctctcgcct


nt

gcatagcggcgtgccctcacgcttctctggaagcggatcaggaaccgattattctc




tcactatttcaaatcttgagcaggaagatattgccacctatttctgccagcagggt




aataccctgccctacaccttcggaggagggaccaagctcgaaatcaccggtggagg




aggcagcggcggtggagggtctggtggaggtggttctgaggtgaagctgcaagaat




caggccctggacttgtggccccttcacagtccctgagcgtgacttgcaccgtgtcc




ggagtctccctgcccgactacggagtgtcatggatcagacaacctccacggaaagg




actggaatggctcggtgtcatctggggtagcgaaactacttactacaattcagccc




tcaaaagcaggctgactattatcaaggacaacagcaagtcccaagtctttcttaag




atgaactcactccagactgacgacaccgcaatctactattgtgctaagcactacta




ctacggaggatcctacgctatggattactggggacaaggtacttccgtcactgtct




cttcacaccatcatcaccatcaccatcac





CTL019-
 98


MALPVTALLLPLALLLHAARP
diqmtqttsslsaslgdrvtiscrasqdiskylnw



Soluble

yqqkpdgtvklliyhtsrlhsgvpsrfsgsgsgtdysltisnleqediatyfcqqg


scFv-

ntlpytfgggtkleitggggsggggsggggsevklqesgpglvapsqslsvtctvs


Histag-

gvslpdygvswirqpprkglewlgviwgsettyynsalksrltiikdnsksqvflk


aa

mnslqtddtaiyycakhyyyggsyamdywgqgtsvtvsshhhhhhhh





CTL019
 99
atggccttaccagtgaccgccttgctcctgccgctggccttgctgctccacgccgc


Full-nt

caggccggacatccagatgacacagactacatcctccctgtctgcctctctgggag




acagagtcaccatcagttgcagggcaagtcaggacattagtaaatatttaaattgg




tatcagcagaaaccagatggaactgttaaactcctgatctaccatacatcaagatt




acactcaggagtcccatcaaggttcagtggcagtgggtctggaacagattattctc




tcaccattagcaacctggagcaagaagatattgccacttacttttgccaacagggt




aatacgcttccgtacacgttcggaggggggaccaagctggagatcacaggtggcgg




tggctcgggcggtggtgggtcgggtggcggcggatctgaggtgaaactgcaggagt




caggacctggcctggtggcgccctcacagagcctgtccgtcacatgcactgtctca




ggggtctcattacccgactatggtgtaagctggattcgccagcctccacgaaaggg




tctggagtggctgggagtaatatggggtagtgaaaccacatactataattcagctc




tcaaatccagactgaccatcatcaaggacaactccaagagccaagttttcttaaaa




atgaacagtctgcaaactgatgacacagccatttactactgtgccaaacattatta




ctacggtggtagctatgctatggactactggggccaaggaacctcagtcaccgtct




cctcaaccacgacgccagcgccgcgaccaccaacaccggcgcccaccatcgcgtcg




cagcccctgtccctgcgcccagaggcgtgccggccagcggcggggggcgcagtgca




cacgagggggctggacttcgcctgtgatatctacatctgggcgcccttggccggga




cttgtggggtccttctcctgtcactggttatcaccctttactgcaaacggggcaga




aagaaactcctgtatatattcaaacaaccatttatgagaccagtacaaactactca




agaggaagatggctgtagctgccgatttccagaagaagaagaaggaggatgtgaac




tgagagtgaagttcagcaggagcgcagacgcccccgcgtacaagcagggccagaac




cagctctataacgagctcaatctaggacgaagagaggagtacgatgttttggacaa




gagacgtggccgggaccctgagatggggggaaagccgagaaggaagaaccctcagg




aaggcctgtacaatgaactgcagaaagataagatggcggaggcctacagtgagatt




gggatgaaaggcgagcgccggaggggcaaggggcacgatggcctttaccagggtct




cagtacagccaccaaggacacctacgacgcccttcacatgcaggccctgccccctc




gc





CTL019
 58
MALPVTALLLPLALLLHAARPdiqmtqttsslsaslgdrvtiscrasqdiskylnw


Full-aa

yqqkpdgtvklliyhtsrlhsgvpsrfsgsgsgtdysltisnleqediatyfcqqg




ntlpytfgggtkleitggggsggggsggggsevklqesgpglvapsqslsvtctvs




gvslpdygvswirqpprkglewlgviwgsettyynsalksrltiikdnsksqvflk




mnslqtddtaiyycakhyyyggsyamdywgqgtsvtvsstttpaprpptpaptias




qplslrpeacrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitlyckrgr




kkllyifkqpfmrpvqttqeedgcscrfpeeeeggcelrvkfsrsadapaykqgqn




qlynelnlgrreeydvldkrrgrdpemggkprrknpqeglynelqkdkmaeaysei




gmkgerrrgkghdglyqglstatkdtydalhmqalppr





CTL019
 59
Diqmtqttsslsaslgdrvtiscrasqdiskylnwyqqkpdgtvklliyhtsrlhs


scFv

gvpsrfsgsgsgtdysltisnleqediatyfcqqgntlpytfgggtkleitggggs


domain

ggggsggggsevklqesgpglvapsqslsvtctvsgvslpdygvswirqpprkgle




wlgviwgsettyynsalksrltiikdnsksqvflkmnslqtddtaiyycakhyyyg




gsyamdywgqgtsvtvss





mCAR1
109
QVQLLESGAELVRPGSSVKISCKASGYAFSSYWMNWVKQRPGQGLEWIGQIYPGDG


scFv

DTNYNGKFKGQATLTADKSSSTAYMQLSGLTSEDSAVYSCARKTISSVVDFYFDYW




GQGTTVTGGGSGGGSGGGSGGGSELVLTQSPKFMSTSVGDRVSVTCKASQNVGTNV




AWYQQKPGQSPKPLIYSATYRNSGVPDRFTGSGSGTDFTLTITNVQSKDLADYFCQ




YNRYPYTSFFFTKLEIKRRS





mCAR1
110
QVQLLESGAELVRPGSSVKISCKASGYAFSSYWMNWVKQRPGQGLEWIGQIYPGDG


Full-aa

DTNYNGKFKGQATLTADKSSSTAYMQLSGLTSEDSAVYSCARKTISSVVDFYFDYW




GQGTTVTGGGSGGGSGGGSGGGSELVLTQSPKFMSTSVGDRVSVTCKASQNVGTNV




AWYQQKPGQSPKPLIYSATYRNSGVPDRFTGSGSGTDFTLTITNVQSKDLADYFCQ




YNRYPYTSFFFTKLEIKRRSKIEVMYPPPYLDNEKSNGTIIHVKGKHLCPSPLFPG




PSKPFWVLVVVGGVLACYSLLVTVAFIIFWVRSKRSRLLHSDYMNMTPRRPGPTRK




HYQPYAPPRDFAAYRSRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRG




RDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTA




TKDTYDALHMQALPPR





mCAR2
111
DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS


scFv

GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG




SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK




GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHY




YYGGSYAMDYWGQGTSVTVSSE





mCAR2
112
DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS


CAR-aa

GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG




SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK




GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIY




YCAKHYYYGGSYAMDYWGQGTSVTVSSESKYGPPCPPCPMFWVLVVVGGVLACYSL




LVTVAFIIFWVKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFEEEEGGCELRVKF




SRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYN




ELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRL





mCAR2
113
DIQMTQTT SSLSASLGDR VTISCRASQD ISKYLNWYQQ KPDGTVKLLI


Full-aa

YHTSRLHSGV PSRFSGSGSG TDYSLTISNL EQEDIATYFC QQGNTLPYTF




GGGTKLEITG STSGSGKPGS GEGSTKGEVK LQESGPGLVA PSQSLSVTCT




VSGVSLPDYG VSWIRQPPRK GLEWLGVIWG SETTYYNSAL KSRLTIIKDN




SKSQVFLKMN SLQTDDTAIY YCAKHYYYGG SYAMDYWGQG TSVTVSSESK




YGPPCPPCPM FWVLVVVGGV LACYSLLVTV




AFIIFWVKRG RKKLLYIFKQ PFMRPVQTTQ EEDGCSCRFE EEEGGCELRV




KFSRSADAPA YQQGQNQLYN ELNLGRREEY DVLDKRRGRD PEMGGKPRRK




NPQEGLYNEL QKDKMAEAYS EIGMKGERRR GKGHDGLYQG LSTATKDTYD




ALHMQALPPR LEGGGEGRGS LLTCGDVEEN PGPRMLLLVT SLLLCELPHP




AFLLIPRKVC NGIGIGEFKD SLSINATNIK HFKNCTSISG DLHILPVAFR




GDSFTHTPPL DPQELDILKT VKEITGFLLI QAWPENRTDL HAFENLEIIR




GRTKQHGQFS LAVVSLNITS LGLRSLKEIS DGDVIISGNK NLCYANTINW




KKLFGTSGQK TKIISNRGEN SCKATGQVCH ALCSPEGCWG PEPRDCVSCR




NVSRGRECVD KCNLLEGEPR EFVENSECIQ CHPECLPQAM NITCTGRGPD




NCIQCAHYID GPHCVKTCPA GVMGENNTLV WKYADAGHVC HLCHPNCTYG




CTGPGLEGCP TNGPKIPSIA TGMVGALLLL LVVALGIGLF M





mCAR3
114
DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS


scFv

GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG




SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK




GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHY




YYGGSYAMDYWGQGTSVTVSS





mCAR3
115
DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHS


Full-aa

GVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSG




SGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRK




GLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHY




YYGGSYAMDYWGQGTSVTVSSAAAIEVMYPPPYLDNEKSNGTIIHVKGKHLCPSPL




FPGPSKPFWVLVVVGGVLACYSLLVTVAFIIFWVRSKRSRLLHSDYMNMTPRRPGP




TRKHYQPYAPPRDFAAYRSRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDK




RRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGL




STATKDTYDALHMQALPPR









In some embodiments, the CDRs are defined according to the Kabat numbering scheme, the Chothia numbering scheme, or a combination thereof.


The sequences of humanized CDR sequences of the scFv domains are shown in Table 4 for the heavy chain variable domains and in Table 5 for the light chain variable domains. “ID” stands for the respective SEQ ID NO for each CDR.









TABLE 4







Heavy Chain Variable Domain CDRs (Kabat)














Candidate
FW
HCDR1
ID
HCDR2
ID
HCDR3
ID





murine_CART19

GVSLPDYGVS
19
VIWGSETTYYNSALKS
20
HYYYGGSYAMDY
24





humanized_CART19 a
VH4
GVSLPDYGVS
19
VIWGSETTYYcustom-character Scustom-character LKS
21
HYYYGGSYAMDY
24





humanized_CART19 b
VH4
GVSLPDYGVS
19
VIWGSETTYYcustom-character Scustom-character LKS
22
HYYYGGSYAMDY
24





humanized_CART19 c
VH4
GVSLPDYGVS
19
VIWGSETTYYNScustom-character LKS
23
HYYYGGSYAMDY
24
















TABLE 5







Light Chain Variable Domain CDRs














Candidate
FW
LCDR1
ID
LCDR2
ID
LCDR3
ID





murine_

RASQDI
25
HTSRL
26
QQGNT
27


CART19

SKYLN

HS

LPYT






humanized_
VK3
RASQDI
25
HTSRL
26
QQGNT
27


CART19 a

SKYLN

HS

LPYT






humanized_
VK3
RASQDI
25
HTSRL
26
QQGNT
27


CART19 b

SKYLN

HS

LPYT






humanized_
VK3
RASQDI
25
HTSRL
26
QQGNT
27


CART19 c

SKYLN

HS

LPYT









The CAR scFv fragments were then cloned into lentiviral vectors to create a full length CAR construct in a single coding frame, and using the EF1 alpha promoter for expression (SEQ ID NO: 100).


EF1 Alpha Promoter









(SEQ ID NO: 100)


CGTGAGGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGT





CCCCGAGAAGTTGGGGGGAGGGGTCGGCAATTGAACCGGTGCCTAGAGA





AGGTGGCGCGGGGTAAACTGGGAAAGTGATGTCGTGTACTGGCTCCGCC





TTTTTCCCGAGGGTGGGGGAGAACCGTATATAAGTGCAGTAGTCGCCGT





GAACGTTCTTTTTCGCAACGGGTTTGCCGCCAGAACACAGGTAAGTGCC





GTGTGTGGTTCCCGCGGGCCTGGCCTCTTTACGGGTTATGGCCCTTGCG





TGCCTTGAATTACTTCCACCTGGCTGCAGTACGTGATTCTTGATCCCGA





GCTTCGGGTTGGAAGTGGGTGGGAGAGTTCGAGGCCTTGCGCTTAAGGA





GCCCCTTCGCCTCGTGCTTGAGTTGAGGCCTGGCCTGGGCGCTGGGGCC





GCCGCGTGCGAATCTGGTGGCACCTTCGCGCCTGTCTCGCTGCTTTCGA





TAAGTCTCTAGCCATTTAAAATTTTTGATGACCTGCTGCGACGCTTTTT





TTCTGGCAAGATAGTCTTGTAAATGCGGGCCAAGATCTGCACACTGGTA





TTTCGGTTTTTGGGGCCGCGGGCGGCGACGGGGCCCGTGCGTCCCAGCG





CACATGTTCGGCGAGGCGGGGCCTGCGAGCGCGGCCACCGAGAATCGGA





CGGGGGTAGTCTCAAGCTGGCCGGCCTGCTCTGGTGCCTGGCCTCGCGC





CGCCGTGTATCGCCCCGCCCTGGGCGGCAAGGCTGGCCCGGTCGGCACC





AGTTGCGTGAGCGGAAAGATGGCCGCTTCCCGGCCCTGCTGCAGGGAGC





TCAAAATGGAGGACGCGGCGCTCGGGAGAGCGGGCGGGTGAGTCACCCA





CACAAAGGAAAAGGGCCTTTCCGTCCTCAGCCGTCGCTTCATGTGACTC





CACGGAGTACCGGGCGCCGTCCAGGCACCTCGATTAGTTCTCGAGCTTT





TGGAGTACGTCGTCTTTAGGTTGGGGGGAGGGGTTTTATGCGATGGAGT





TTCCCCACACTGAGTGGGTGGAGACTGAAGTTAGGCCAGCTTGGCACTT





GATGTAATTCTCCTTGGAATTTGCCCTTTTTGAGTTTGGATCTTGGTTC





ATTCTCAAGCCTCAGACAGTGGTTCAAAGTTTTTTTCTTCCATTTCAGG





TGTCGTGA.






RNA Transfection

Disclosed herein are methods for producing an in vitro transcribed RNA CAR. The present invention also includes (among other things) a CAR encoding RNA construct that can be directly transfected into a cell. A method for generating mRNA for use in transfection can involve in vitro transcription (IVT) of a template with specially designed primers, followed by polyA addition, to produce a construct containing 3′ and 5′ untranslated sequence (“UTR”), a 5′ cap and/or Internal Ribosome Entry Site (IRES), the nucleic acid to be expressed, and a polyA tail, typically 50-2000 bases in length (SEQ ID NO:118). RNA so produced can efficiently transfect different kinds of cells. In one aspect, the template includes sequences for the CAR.


In one aspect the CAR is encoded by a messenger RNA (mRNA). In one aspect the mRNA encoding the CAR is introduced into an immune effector cell, e.g., a T cell or a NK cell, for production of a CAR-expressing cell, e.g., a CART cell or a CAR NK cell.


Methods of producing an in vitro transcribed RNA CAR are described on pages 192-196 of International Application WO 2016/164731 filed on 8 Apr. 2016, hereby incorporated by reference.


Non-Viral Delivery Methods

In some aspects, non-viral methods can be used to deliver a nucleic acid encoding a CAR described herein into a cell or tissue or a subject.


In some embodiments, the non-viral method includes the use of a transposon (also called a transposable element). In some embodiments, a transposon is a piece of DNA that can insert itself at a location in a genome, for example, a piece of DNA that is capable of self-replicating and inserting its copy into a genome, or a piece of DNA that can be spliced out of a longer nucleic acid and inserted into another place in a genome. For example, a transposon comprises a DNA sequence made up of inverted repeats flanking genes for transposition.


Exemplary methods of nucleic acid delivery systems and methods of using thereof are described on pages 196-198 of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


Nucleic Acid Constructs Encoding a CAR, e.g., a CD19 CAR, or CD22 CAR

The present invention also provides nucleic acid molecules encoding one or more CAR constructs described herein, e.g., CD19 CAR, or CD22 CAR. In one aspect, the nucleic acid molecule is provided as a messenger RNA transcript. In one aspect, the nucleic acid molecule is provided as a DNA construct.


Accordingly, in one aspect, the invention pertains to an isolated nucleic acid molecule encoding a chimeric antigen receptor (CAR), wherein the CAR comprises a binding domain (e.g., that binds CD19, or CD22) a transmembrane domain, and an intracellular signaling domain comprising a stimulatory domain, e.g., a costimulatory signaling domain and/or a primary signaling domain, e.g., zeta chain.


In one embodiment, the binding domain is an anti-CD19 binding domain described herein, e.g., an anti-CD19 binding domain which comprises a sequence selected from a group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12 and SEQ ID NO:59, or a sequence with 95-99% identity thereof.


In one embodiment, the nucleic acid comprises CD22-encoding a nucleic acid set out in Table 6 or a sequence with 95-99% identity thereof. In one embodiment, the nucleic acid is a nucleic acid encoding an amino acid sequence set out in any of Tables 6-10 or a sequence with 95-99% identity thereof.


In one embodiment, the transmembrane domain is transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154. In one embodiment, the transmembrane domain comprises a sequence of SEQ ID NO: 15, or a sequence with 95-99% identity thereof. In one embodiment, the anti-CD19 binding domain is connected to the transmembrane domain by a hinge region, e.g., a hinge described herein. In one embodiment, the hinge region comprises SEQ ID NO:14 or SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49, or a sequence with 95-99% identity thereof. In one embodiment, the isolated nucleic acid molecule further comprises a sequence encoding a costimulatory domain. In one embodiment, the costimulatory domain is a functional signaling domain of a protein selected from the group consisting of OX40, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). In one embodiment, the costimulatory domain is a functional signaling domain of a protein selected from the group consisting of MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, 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, NKG2D, NKG2C, 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, CD19a, and a ligand that specifically binds with CD83. In one embodiment, the costimulatory domain comprises a sequence of SEQ ID NO:16, or a sequence with 95-99% identity thereof. In one embodiment, the intracellular signaling domain comprises a functional signaling domain of 4-1BB and a functional signaling domain of CD3 zeta. In one embodiment, the intracellular signaling domain comprises the sequence of SEQ ID NO: 16 or SEQ ID NO:51, or a sequence with 95-99% identity thereof, and the sequence of SEQ ID NO: 17 or SEQ ID NO:43, or a sequence with 95-99% identity thereof, wherein the sequences comprising the intracellular signaling domain are expressed in the same frame and as a single polypeptide chain.


In another aspect, the invention pertains to an isolated nucleic acid molecule encoding a CAR construct comprising a leader sequence of SEQ ID NO: 13, a scFv domain having a sequence selected from the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, and SEQ ID NO:59, (or a sequence with 95-99% identity thereof), a hinge region of SEQ ID NO:14 or SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49 (or a sequence with 95-99% identity thereof), a transmembrane domain having a sequence of SEQ ID NO: 15 (or a sequence with 95-99% identity thereof), a 4-1BB costimulatory domain having a sequence of SEQ ID NO:16 or a CD27 costimulatory domain having a sequence of SEQ ID NO:51 (or a sequence with 95-99% identity thereof), and a CD3 zeta stimulatory domain having a sequence of SEQ ID NO:17 or SEQ ID NO:43 (or a sequence with 95-99% identity thereof).


In another aspect, the invention pertains to an isolated polypeptide molecule encoded by the nucleic acid molecule. In one embodiment, the isolated polypeptide molecule comprises a sequence selected from the group consisting of SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:59 or a sequence with 95-99% identity thereof.


In another aspect, the invention pertains to a nucleic acid molecule encoding a chimeric antigen receptor (CAR) molecule that comprises an anti-CD19 binding domain, a transmembrane domain, and an intracellular signaling domain comprising a stimulatory domain, and wherein said anti-CD19 binding domain comprises a sequence selected from the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12 and SEQ ID NO:59, or a sequence with 95-99% identity thereof.


In one embodiment, the encoded CAR molecule (e.g., CD19 CAR, CD20 CAR, or CD22 CAR) further comprises a sequence encoding a costimulatory domain. In one embodiment, the costimulatory domain is a functional signaling domain of a protein selected from the group consisting of OX40, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18) and 4-1BB (CD137). In one embodiment, the costimulatory domain comprises a sequence of SEQ ID NO:16. In one embodiment, the transmembrane domain is a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154. In one embodiment, the transmembrane domain comprises a sequence of SEQ ID NO:15. In one embodiment, the intracellular signaling domain comprises a functional signaling domain of 4-1BB and a functional signaling domain of zeta. In one embodiment, the intracellular signaling domain comprises the sequence of SEQ ID NO: 16 and the sequence of SEQ ID NO: 17, wherein the sequences comprising the intracellular signaling domain are expressed in the same frame and as a single polypeptide chain. In one embodiment, the anti-CD19 binding domain is connected to the transmembrane domain by a hinge region. In one embodiment, the hinge region comprises SEQ ID NO:14. In one embodiment, the hinge region comprises SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49.


In another aspect, the invention pertains to an encoded CAR molecule comprising a leader sequence of SEQ ID NO: 13, a scFv domain having a sequence selected from the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, and SEQ ID NO:59, or a sequence with 95-99% identity thereof, a hinge region of SEQ ID NO:14 or SEQ ID NO:45 or SEQ ID NO:47 or SEQ ID NO:49, a transmembrane domain having a sequence of SEQ ID NO: 15, a 4-1BB costimulatory domain having a sequence of SEQ ID NO:16 or a CD27 costimulatory domain having a sequence of SEQ ID NO:51, and a CD3 zeta stimulatory domain having a sequence of SEQ ID NO:17 or SEQ ID NO:43. In one embodiment, the encoded CAR molecule comprises a sequence selected from a group consisting of SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, SEQ ID NO:39, SEQ ID NO:40, SEQ ID NO:41, SEQ ID NO:42, and SEQ ID NO:59, or a sequence with 95-99% identity thereof.


The nucleic acid sequences coding for the desired molecules can be obtained using recombinant methods known in the art, such as, for example by screening libraries from cells expressing the gene, by deriving the gene from a vector known to include the same, or by isolating directly from cells and tissues containing the same, using standard techniques. Alternatively, the gene of interest can be produced synthetically, rather than cloned.


The present invention also provides vectors in which a DNA of the present invention is inserted. Vectors derived from retroviruses such as the lentivirus are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene and its propagation in daughter cells. Lentiviral vectors have the added advantage over vectors derived from onco-retroviruses such as murine leukemia viruses in that they can transduce non-proliferating cells, such as hepatocytes. They also have the added advantage of low immunogenicity. A retroviral vector may also be, e.g., a gammaretroviral vector. A gammaretroviral vector may include, e.g., a promoter, a packaging signal (w), a primer binding site (PBS), one or more (e.g., two) long terminal repeats (LTR), and a transgene of interest, e.g., a gene encoding a CAR. A gammaretroviral vector may lack viral structural gens such as gag, pol, and env. Exemplary gammaretroviral vectors include Murine Leukemia Virus (MLV), Spleen-Focus Forming Virus (SFFV), and Myeloproliferative Sarcoma Virus (MPSV), and vectors derived therefrom. Other gammaretroviral vectors are described, e.g., in Tobias Maetzig et al., “Gammaretroviral Vectors: Biology, Technology and Application” Viruses. 2011 June; 3(6): 677-713.


In another embodiment, the vector comprising the nucleic acid encoding the desired CAR of the invention is an adenoviral vector (A5/35). In another embodiment, the expression of nucleic acids encoding CARs can be accomplished using of transposons such as sleeping beauty, crispr, CAS9, and zinc finger nucleases. See below June et al. 2009 Nature Reviews Immunology 9.10: 704-716, is incorporated herein by reference.


A vector may also include, e.g., a signal sequence to facilitate secretion, a polyadenylation signal and transcription terminator (e.g., from Bovine Growth Hormone (BGH) gene), an element allowing episomal replication and replication in prokaryotes (e.g. SV40 origin and ColE1 or others known in the art) and/or elements to allow selection (e.g., ampicillin resistance gene and/or zeocin marker).


In brief summary, the expression of natural or synthetic nucleic acids encoding CARs is typically achieved by operably linking a nucleic acid encoding the CAR polypeptide or portions thereof to a promoter, and incorporating the construct into an expression vector. The vectors can be suitable for replication and integration eukaryotes. Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence.


In some aspects, the expression constructs of the present invention may also be used for nucleic acid immunization and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art. See, e.g., U.S. Pat. Nos. 5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties. In another embodiment, the invention provides a gene therapy vector.


The nucleic acid can be cloned into a number of types of vectors. For example, the nucleic acid can be cloned into a vector including, but not limited to a plasmid, a phagemid, a phage derivative, an animal virus, and a cosmid. Vectors of particular interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.


Further, the expression vector may be provided to a cell in the form of a viral vector. Viral vector technology is well known in the art and is described, for example, in Sambrook et al., 2012, MOLECULAR CLONING: A LABORATORY MANUAL, volumes 1-4, Cold Spring Harbor Press, NY), and in other virology and molecular biology manuals. Viruses, which are useful as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses. In general, a suitable vector contains an origin of replication functional in at least one organism, a promoter sequence, convenient restriction endonuclease sites, and one or more selectable markers, (e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).


A number of viral based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. A selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo. A number of retroviral systems are known in the art. In some embodiments, adenovirus vectors are used. A number of adenovirus vectors are known in the art. In one embodiment, lentivirus vectors are used.


Additional promoter elements, e.g., enhancers, regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription. Exemplary promoters include the CMV IE gene, EF-1α, ubiquitin C, or phosphoglycerokinase (PGK) promoters. In an embodiment, the promoter is a PGK promoter, e.g., a truncated PGK promoter as described herein.


An example of a promoter that is capable of expressing a CAR transgene in a mammalian T cell is the EF1a promoter. The native EF1a promoter drives expression of the alpha subunit of the elongation factor-1 complex, which is responsible for the enzymatic delivery of aminoacyl tRNAs to the ribosome. The EF1a promoter has been extensively used in mammalian expression plasmids and has been shown to be effective in driving CAR expression from transgenes cloned into a lentiviral vector. See, e.g., Milone et al., Mol. Ther. 17(8): 1453-1464 (2009). In one aspect, the EF1a promoter comprises the sequence provided as SEQ ID NO:100.


Another example of a promoter is the immediate early cytomegalovirus (CMV) promoter sequence. This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto. However, other constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the elongation factor-1α promoter, the hemoglobin promoter, and the creatine kinase promoter. Further, the invention should not be limited to the use of constitutive promoters. Inducible promoters are also contemplated as part of the invention. The use of an inducible promoter provides a molecular switch capable of turning on expression of the polynucleotide sequence which it is operatively linked when such expression is desired, or turning off the expression when expression is not desired. Examples of inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline promoter.


Another example of a promoter is the phosphoglycerate kinase (PGK) promoter. In embodiments, a truncated PGK promoter (e.g., a PGK promoter with one or more, e.g., 1, 2, 5, 10, 100, 200, 300, or 400, nucleotide deletions when compared to the wild-type PGK promoter sequence) may be desired. The nucleotide sequences of exemplary PGK promoters are provided below.









WT PGK Promoter:


(SEQ ID NO: 1323)


ACCCCTCTCTCCAGCCACTAAGCCAGTTGCTCCCTCGGCTGACGGCTGC





ACGCGAGGCCTCCGAACGTCTTACGCCTTGTGGCGCGCCCGTCCTTGTC





CCGGGTGTGATGGCGGGGTGTGGGGCGGAGGGCGTGGCGGGGAAGGGCC





GGCGACGAGAGCCGCGCGGGACGACTCGTCGGCGATAACCGGTGTCGGG





TAGCGCCAGCCGCGCGACGGTAACGAGGGACCGCGACAGGCAGACGCTC





CCATGATCACTCTGCACGCCGAAGGCAAATAGTGCAGGCCGTGCGGCGC





TTGGCGTTCCTTGGAAGGGCTGAATCCCCGCCTCGTCCTTCGCAGCGGC





CCCCCGGGTGTTCCCATCGCCGCTTCTAGGCCCACTGCGACGCTTGCCT





GCACTTCTTACACGCTCTGGGTCCCAGCCGCGGCGACGCAAAGGGCCTT





GGTGCGGGTCTCGTCGGCGCAGGGACGCGTTTGGGTCCCGACGGAACCT 





TTTCCGCGTTGGGGTTGGGGCACCATAAGCT





Exemplary truncated PGK Promoters:


PGK100:


(SEQ ID NO: 1324)


ACCCCTCTCTCCAGCCACTAAGCCAGTTGCTCCCTCGGCTGACGGCTGC





ACGCGAGGCCTCCGAACGTCTTACGCCTTGTGGCGCGCCCGTCCTTGTC





CCGGGTGTGATGGCGGGGTG 





PGK200:


(SEQ ID NO: 1325)


ACCCCTCTCTCCAGCCACTAAGCCAGTTGCTCCCTCGGCTGACGGCTGC





ACGCGAGGCCTCCGAACGTCTTACGCCTTGTGGCGCGCCCGTCCTTGTC





CCGGGTGTGATGGCGGGGTGTGGGGCGGAGGGCGTGGCGGGGAAGGGCC





GGCGACGAGAGCCGCGCGGGACGACTCGTCGGCGATAACCGGTGTCGGG





TAGCGCCAGCCGCGCGACGGTAACG





PGK300:


(SEQ ID NO: 1326)


ACCCCTCTCTCCAGCCACTAAGCCAGTTGCTCCCTCGGCTGACGGCTGC





ACGCGAGGCCTCCGAACGTCTTACGCCTTGTGGCGCGCCCGTCCTTGTC





CCGGGTGTGATGGCGGGGTGTGGGGCGGAGGGCGTGGCGGGGAAGGGCC





GGCGACGAGAGCCGCGCGGGACGACTCGTCGGCGATAACCGGTGTCGGG





TAGCGCCAGCCGCGCGACGGTAACGAGGGACCGCGACAGGCAGACGCTC





CCATGATCACTCTGCACGCCGAAGGCAAATAGTGCAGGCCGTGCGGCGC





TTGGCGTTCCTTGGAAGGGCTGAATCCCCG 





PGK400:


(SEQ ID NO: 1327)


ACCCCTCTCTCCAGCCACTAAGCCAGTTGCTCCCTCGGCTGACGGCTGC





ACGCGAGGCCTCCGAACGTCTTACGCCTTGTGGCGCGCCCGTCCTTGTC





CCGGGTGTGATGGCGGGGTGTGGGGCGGAGGGCGTGGCGGGGAAGGGCC





GGCGACGAGAGCCGCGCGGGACGACTCGTCGGCGATAACCGGTGTCGGG





TAGCGCCAGCCGCGCGACGGTAACGAGGGACCGCGACAGGCAGACGCTC





CCATGATCACTCTGCACGCCGAAGGCAAATAGTGCAGGCCGTGCGGCGC





TTGGCGTTCCTTGGAAGGGCTGAATCCCCGCCTCGTCCTTCGCAGCGGC





CCCCCGGGTGTTCCCATCGCCGCTTCTAGGCCCACTGCGACGCTTGCCT





GCACTTCTTACACGCTCTGGGTCCCAGCCG






A vector may also include, e.g., a signal sequence to facilitate secretion, a polyadenylation signal and transcription terminator (e.g., from Bovine Growth Hormone (BGH) gene), an element allowing episomal replication and replication in prokaryotes (e.g. SV40 origin and ColE1 or others known in the art) and/or elements to allow selection (e.g., ampicillin resistance gene and/or zeocin marker).


In order to assess the expression of a CAR polypeptide or portions thereof, the expression vector to be introduced into a cell can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or infected through viral vectors. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both selectable markers and reporter genes may be flanked with appropriate regulatory sequences to enable expression in the host cells. Useful selectable markers include, for example, antibiotic-resistance genes, such as neo and the like.


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


In embodiments, the vector may comprise two or more nucleic acid sequences encoding a CAR, e.g., a first CAR that binds to CD19 and a second CAR, e.g., an inhibitory CAR or a CAR that specifically binds to a second antigen, e.g., CD10, CD20, CD22, CD34, CD123, FLT-3, ROR1, CD79b, CD179b, or CD79a. In such embodiments, the two or more nucleic acid sequences encoding the CAR are encoded by a single nucleic molecule in the same frame and as a single polypeptide chain. In this aspect, the two or more CARs, can, e.g., be separated by one or more peptide cleavage sites. (e.g., an auto-cleavage site or a substrate for an intracellular protease). Examples of peptide cleavage sites include the following, wherein the GSG residues are optional:











T2A:



(SEQ ID NO: 1328)



(GSG)EGRGSLLTCGDVEENPGP







P2A:



(SEQ ID NO: 1329)



(GSG)ATNFSLLKQAGDVEENPGP







E2A:



(SEQ ID NO: 1330)



(GSG)QCTNYALLKLAGDVESNPGP







F2A:



(SEQ ID NO: 1331)



(GSG)VKQTLNFDLLKLAGDVESNPGP






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


Physical methods for introducing a polynucleotide into a host cell include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well-known in the art. See, for example, Sambrook et al., 2012, MOLECULAR CLONING: A LABORATORY MANUAL, volumes 1-4, Cold Spring Harbor Press, NY). A suitable method for the introduction of a polynucleotide into a host cell is calcium phosphate transfection


Biological methods for introducing a polynucleotide of interest into a host cell include the use of DNA and RNA vectors. Viral vectors, and especially retroviral vectors, have become the most widely used method for inserting genes into mammalian, e.g., human cells. Other viral vectors can be derived from lentivirus, poxviruses, herpes simplex virus I, adenoviruses and adeno-associated viruses, and the like. See, for example, U.S. Pat. Nos. 5,350,674 and 5,585,362.


Chemical means for introducing a polynucleotide into a host cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (e.g., an artificial membrane vesicle). Other methods of state-of-the-art targeted delivery of nucleic acids are available, such as delivery of polynucleotides with targeted nanoparticles or other suitable sub-micron sized delivery system.


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


Lipids suitable for use are described on page 209 of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


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


The present invention further provides a vector comprising a CAR encoding nucleic acid molecule. In one aspect, a CAR vector can be directly transduced into a cell, e.g., a T cell. In one aspect, the vector is a cloning or expression vector, e.g., a vector including, but not limited to, one or more plasmids (e.g., expression plasmids, cloning vectors, minicircles, minivectors, double minute chromosomes), retroviral and lentiviral vector constructs. In one aspect, the vector is capable of expressing the CAR construct in mammalian T cells. In one aspect, the mammalian T cell is a human T cell.


Immune Effector Cells Expressing a CAR

In another aspect, the present invention provides a population of CAR-expressing cells. In some embodiments, the population of CAR-expressing cells comprises a cell that expresses one or more CARs described herein. In some embodiments, the population of CAR-expressing cells comprises a mixture of cells expressing different CARs.


For example, in one embodiment, the population of CART cells can include a first cell expressing a CAR having an antigen binding domain to a tumor antigen described herein, e.g., CD19, and a second cell expressing a CAR having a different antigen binding domain, e.g., an antigen binding domain to a different tumor antigen described herein, e.g., an antigen binding domain to a tumor antigen described herein that differs from the tumor antigen bound by the antigen binding domain of the CAR expressed by the first cell, e.g., CD22.


As another example, the population of CAR-expressing cells can include a first cell expressing a CAR that includes an antigen binding domain to a tumor antigen described herein, and a second cell expressing a CAR that includes an antigen binding domain to a target other than a tumor antigen as described herein. In one embodiment, the population of CAR-expressing cells includes, e.g., a first cell expressing a CAR that includes a primary intracellular signaling domain, and a second cell expressing a CAR that includes a secondary signaling domain. Either one or both of the CAR expressing cells can have a truncated PGK promoter, e.g., as described herein, operably linked to the nucleic acid encoding the CAR.


In another aspect, the present invention provides a population of cells wherein at least one cell in the population expresses a CAR having an antigen binding domain to a tumor antigen described herein, and a second cell expressing another agent, e.g., an agent which enhances the activity of a CAR-expressing cell. The CAR expressing cells of the population can have a truncated PGK promoter, e.g., as described herein, operably linked to the nucleic acid encoding the CAR. In one embodiment, the agent can be an agent which inhibits an inhibitory molecule. Inhibitory molecules, e.g., PD-1, can, in some embodiments, decrease the ability of a CAR-expressing cell to mount an immune effector response. Examples of inhibitory molecules include PD-1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (CEACAM-1, CEACAM-3, and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GALS, adenosine, and TGFR (e.g., TGFRbeta). In one embodiment, the agent which inhibits an inhibitory molecule comprises a first polypeptide, e.g., an inhibitory molecule, associated with a second polypeptide that provides a positive signal to the cell, e.g., an intracellular signaling domain described herein. In one embodiment, the agent comprises a first polypeptide, e.g., of an inhibitory molecule such as PD1, PD-L1, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3, and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 or TGFR beta, or a fragment of any of these, and a second polypeptide which is an intracellular signaling domain described herein (e.g., comprising a costimulatory domain (e.g., 41BB, CD27, OX40 or CD28, e.g., as described herein) and/or a primary signaling domain (e.g., a CD3 zeta signaling domain described herein). In one embodiment, the agent comprises a first polypeptide of PD-1 or a fragment thereof, and a second polypeptide of an intracellular signaling domain described herein (e.g., a CD28 signaling domain described herein and/or a CD3 zeta signaling domain described herein).


Co-Expression of CAR with Other Molecules or Agents


Co-Expression of a Second CAR

In one aspect, the CAR-expressing cell described herein can further comprise a second CAR, e.g., a second CAR that includes a different antigen binding domain, e.g., to the same target as the first CAR (e.g., CD19) or a different target (e.g., CD22). In one embodiment, the second CAR includes an antigen binding domain to a target expressed on ALL cells, such as, CD22. In one embodiment, the CAR-expressing cell comprises a first CAR that targets a first antigen and includes an intracellular signaling domain having a costimulatory signaling domain but not a primary signaling domain, and a second CAR that targets a second, different, antigen and includes an intracellular signaling domain having a primary signaling domain but not a costimulatory signaling domain. While not wishing to be bound by theory, placement of a costimulatory signaling domain, e.g., 4-1BB, CD28, CD27 or OX-40, onto the first CAR, and the primary signaling domain, e.g., CD3 zeta, on the second CAR can limit the CAR activity to cells where both targets are expressed. In one embodiment, the CAR expressing cell comprises a first CD19 CAR that includes a CD19 binding domain, a transmembrane domain and a costimulatory domain and a second CAR that targets an antigen other than CD19 (e.g., an antigen expressed on ALL cells, e.g., CD22) and includes an antigen binding domain, a transmembrane domain and a primary signaling domain. In another embodiment, the CAR expressing cell comprises a first CD19 CAR that includes a CD19 binding domain, a transmembrane domain and a primary signaling domain and a second CAR that targets an antigen other than CD19 (e.g., an antigen expressed on ALL cells, e.g., CD22) and includes an antigen binding domain to the antigen, a transmembrane domain and a costimulatory signaling domain.


In one aspect, the CAR-expressing cell described herein can further comprise a second CAR, e.g., a second CAR that includes a different antigen binding domain, e.g., to the same target (e.g., CD19) or a different target (e.g., a target other than CD19, e.g., CD22). In one embodiment, the CAR-expressing cell comprises a first CAR that targets a first antigen and includes an intracellular signaling domain having a costimulatory signaling domain but not a primary signaling domain, and a second CAR that targets a second, different, antigen and includes an intracellular signaling domain having a primary signaling domain but not a costimulatory signaling domain. Placement of a costimulatory signaling domain, e.g., 4-1BB, CD28, CD27, OX-40 or ICOS, onto the first CAR, and the primary signaling domain, e.g., CD3 zeta, on the second CAR can limit the CAR activity to cells where both targets are expressed. In one embodiment, the CAR expressing cell comprises a first CAR that includes an antigen binding domain, a transmembrane domain and a costimulatory domain and a second CAR that targets another antigen and includes an antigen binding domain, a transmembrane domain and a primary signaling domain. In another embodiment, the CAR expressing cell comprises a first CAR that includes an antigen binding domain, a transmembrane domain and a primary signaling domain and a second CAR that targets another antigen and includes an antigen binding domain to the antigen, a transmembrane domain and a costimulatory signaling domain.


In one embodiment, when the CAR-expressing cell comprises two or more different CARs, the antigen binding domains of the different CARs can be such that the antigen binding domains do not interact with one another. For example, a cell expressing a first and second CAR can have an antigen binding domain of the first CAR, e.g., as a fragment, e.g., an scFv, that does not form an association with the antigen binding domain of the second CAR, e.g., the antigen binding domain of the second CAR is a VHH.


Co-Expression of an Agent that Enhances CAR Activity


In another aspect, the CAR-expressing cell described herein can further express another agent, e.g., an agent that enhances the activity or fitness of a CAR-expressing cell.


For example, in one embodiment, the agent can be an agent which inhibits a molecule that modulates or regulates, e.g., inhibits, T cell function. In some embodiments, the molecule that modulates or regulates T cell function is an inhibitory molecule. Inhibitory molecules, e.g., PD1, can, in some embodiments, decrease the ability of a CAR-expressing cell to mount an immune effector response. Examples of inhibitory molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAGS, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GALS, adenosine, and TGF (e.g., TGF beta).


In one embodiment, an inhibitory nucleic acid, e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used to inhibit expression of a molecule that modulates or regulates, e.g., inhibits, T-cell function in the CAR-expressing cell. In an embodiment the agent is an shRNA, e.g., an shRNA described herein. In an embodiment, the agent that modulates or regulates, e.g., inhibits, T-cell function is inhibited within a CAR-expressing cell. For example, a dsRNA molecule that inhibits expression of a molecule that modulates or regulates, e.g., inhibits, T-cell function is linked to the nucleic acid that encodes a component, e.g., all of the components, of the CAR.


In one embodiment, the agent which inhibits an inhibitory molecule comprises a first polypeptide, e.g., an inhibitory molecule, associated with a second polypeptide that provides a positive signal to the cell, e.g., an intracellular signaling domain described herein. In one embodiment, the agent comprises a first polypeptide, e.g., of an inhibitory molecule such as PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAGS, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GALS, adenosine, or TGF (e.g., TGF beta), or a fragment of any of these (e.g., at least a portion of an extracellular domain of any of these), and a second polypeptide which is an intracellular signaling domain described herein (e.g., comprising a costimulatory domain (e.g., 41BB, CD27 or CD28, e.g., as described herein) and/or a primary signaling domain (e.g., a CD3 zeta signaling domain described herein). In one embodiment, the agent comprises a first polypeptide of PD1 or a fragment thereof (e.g., at least a portion of an extracellular domain of PD1), and a second polypeptide of an intracellular signaling domain described herein (e.g., a CD28 signaling domain described herein and/or a CD3 zeta signaling domain described herein). PD1 is an inhibitory member of the CD28 family of receptors that also includes CD28, CTLA-4, ICOS, and BTLA. PD-1 is expressed on activated B cells, T cells and myeloid cells (Agata et al. 1996 Int. Immunol 8:765-75). Two ligands for PD1, PD-L1 and PD-L2 have been shown to downregulate T cell activation upon binding to PD1 (Freeman et a. 2000 J Exp Med 192:1027-34; Latchman et al. 2001 Nat Immunol 2:261-8; Carter et al. 2002 Eur J Immunol 32:634-43). PD-L1 is abundant in human cancers (Dong et al. 2003 J Mol Med 81:281-7; Blank et al. 2005 Cancer Immunol. Immunother 54:307-314; Konishi et al. 2004 Clin Cancer Res 10:5094). Immune suppression can be reversed by inhibiting the local interaction of PD1 with PD-L1.


In one embodiment, the agent comprises the extracellular domain (ECD) of an inhibitory molecule, e.g., Programmed Death 1 (PD1), can be fused to a transmembrane domain and intracellular signaling domains such as 41BB and CD3 zeta (also referred to herein as a PD1 CAR). In one embodiment, the PD1 CAR, when used in combinations with a CD19 CAR described herein, improves the persistence of the T cell. In one embodiment, the CAR is a PD1 CAR comprising the extracellular domain of PD1 indicated as underlined in SEQ ID NO: 121. In one embodiment, the PD1 CAR comprises the amino acid sequence of SEQ ID NO:121.









(SEQ ID NO: 121)


Malpvtalllplalllhaarppgwfldspdrpwnpptfspallvvtegd






natftcsfsntsesfvlnwyrmspsnqtdklaafpedrsqpgqdcrfrv







tqlpngrdfhmsvvrarrndsgtylcgaislapkaqikeslraelrvte







rraevptahpspsprpagqfqtlvtttpaprpptpaptiasqplslrpe






acrpaaggavhtrgldfacdiyiwaplagtcgvlllslvitlyckrgrk





kllyifkqpfmrpvqttqeedgcscrfpeeeeggcelrvkfsrsadapa





ykqgqnqlynelnlgrreeydvldkrrgrdpemggkprrknpqeglyne





lqkdkmaeayseigmkgerrrgkghdglyqglstatkdtydalhmqalp





pr.






In one embodiment, the PD1 CAR comprises the amino acid sequence provided below (SEQ ID NO:119).









(SEQ ID NO: 119)



pgwfldspdrpwnpptfspallvvtegdnatftcsfsntsesfvlnwyr







mspsnqtdklaafpedrsqpgqdcrfrvtqlpngrdfhmsvvrarrnds







gtylcgaislapkaqikeslraelrvterraevptahpspsprpagqfq







tlvtttpaprpptpaptiasqplslrpeacrpaaggavhtrgldfacdi






yiwaplagtcgvlllslvitlyckrgrkkllyifkqpfmrpvqttqeed





gcscrfpeeeeggcelrvkfsrsadapaykqgqnqlynelnlgrreeyd





vldkrrgrdpemggkprrknpqeglynelqkdkmaeayseigmkgerrr





gkghdglyqglstatkdtydalhmqalppr.






Tin one embodiment, the agent comprises a nucleic acid sequence encoding the PD1 CAR, e.g., the PD1 CAR described herein. In one embodiment, the nucleic acid sequence for the PD1 CAR is shown below, with the PD1 ECD underlined below in SEQ ID NO: 120









(SEQ ID NO: 120)


atggccctccctgtcactgccctgcttctccccctcgcactcctgctcc





acgccgctagaccacccggatggtttctggactctccggatcgcccgtg






gaatcccccaaccttctcaccggcactcttggttgtgactgagggcgat







aatgcgaccttcacgtgctcgttctccaacacctccgaatcattcgtgc







tgaactggtaccgcatgagcccgtcaaaccagaccgacaagctcgccgc







gtttccggaagatcggtcgcaaccgggacaggattgtcggttccgcgtg







actcaactgccgaatggcagagacttccacatgagcgtggtccgcgcta







ggcgaaacgactccgggacctacctgtgcggagccatctcgctggcgcc







taaggcccaaatcaaagagagcttgagggccgaactgagagtgaccgag







cgcagagctgaggtgccaactgcacatccatccccatcgcctcggcctg







cggggcagtttcagaccctggtcacgaccactccggcgccgcgcccacc






gactccggccccaactatcgcgagccagcccctgtcgctgaggccggaa





gcatgccgccctgccgccggaggtgctgtgcatacccggggattggact





tcgcatgcgacatctacatttgggctcctctcgccggaacttgtggcgt





gctccttctgtccctggtcatcaccctgtactgcaagcggggtcggaaa





aagcttctgtacattttcaagcagcccttcatgaggcccgtgcaaacca





cccaggaggaggacggttgctcctgccggttccccgaagaggaagaagg





aggttgcgagctgcgcgtgaagttctcccggagcgccgacgcccccgcc





tataagcagggccagaaccagctgtacaacgaactgaacctgggacggc





gggaagagtacgatgtgctggacaagcggcgcggccgggaccccgaaat





gggcgggaagcctagaagaaagaaccctcaggaaggcctgtataacgag





ctgcagaaggacaagatggccgaggcctactccgaaattgggatgaagg





gagagcggcggaggggaaaggggcacgacggcctgtaccaaggactgtc





caccgccaccaaggacacatacgatgccctgcacatgcaggcccttccc





cctcgc.






In another example, in one embodiment, the agent which enhances the activity of a CAR-expressing cell can be a costimulatory molecule or costimulatory molecule ligand. Examples of costimulatory molecules include MHC class I molecule, BTLA and a Toll ligand receptor, as well as OX40, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). Further examples of such costimulatory molecules include CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, 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, NKG2D, NKG2C, 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, CD19a, and a ligand that specifically binds with CD83., e.g., as described herein. Examples of costimulatory molecule ligands include CD80, CD86, CD40L, ICOSL, CD70, OX40L, 4-1BBL, GITRL, and LIGHT. In embodiments, the costimulatory molecule ligand is a ligand for a costimulatory molecule different from the costimulatory molecule domain of the CAR. In embodiments, the costimulatory molecule ligand is a ligand for a costimulatory molecule that is the same as the costimulatory molecule domain of the CAR. In an embodiment, the costimulatory molecule ligand is 4-1BBL. In an embodiment, the costimulatory ligand is CD80 or CD86. In an embodiment, the costimulatory molecule ligand is CD70. In embodiments, a CAR-expressing immune effector cell described herein can be further engineered to express one or more additional costimulatory molecules or costimulatory molecule ligands.


Co-Expression of CAR with a Chemokine Receptor


In embodiments, the CAR-expressing cell described herein further comprises a chemokine receptor molecule. Exemplary chemokine receptors that can be used are described on pages 217-218 of of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


Conditional Expression of Immune Response-Enhancing Agents

Also provided herein are compositions and methods for conditionally expressing an agent that enhances the immune response or activity of a CAR-expressing cell described herein. Exemplary compositions and methods for conditionally expressing an agent that enhances the immune response or activity of a CAR-expressing cell are described in International Application WO 2016/164731 filed on Apr. 8, 2016, the entire contents of which is hereby expressly incorporated by reference.


Sources of Cells

Prior to expansion and genetic modification or other modification, a source of cells, e.g., T cells or natural killer (NK) cells, can be obtained from a subject. Examples of subjects include humans, monkeys, chimpanzees, dogs, cats, mice, rats, and transgenic species thereof. T cells can be obtained from a number of 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 aspects of the present disclosure, immune effector cells, e.g., T cells, can be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled artisan, such as Ficoll™ separation. In one aspect, cells from the circulating blood of an individual are obtained by apheresis. The apheresis product typically contains lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets. In one aspect, the cells collected by apheresis may be washed to remove the plasma fraction and, optionally, to place the cells in an appropriate buffer or media for subsequent processing steps. In one embodiment, the cells are washed with phosphate buffered saline (PBS). In an alternative embodiment, the wash solution lacks calcium and may lack magnesium or may lack many if not all divalent cations.


Initial activation steps in the absence of calcium can lead to magnified activation. As those of ordinary skill in the art would readily appreciate a washing step may be accomplished by methods known to those in the art, such as by using a semi-automated “flow-through” centrifuge (for example, the Cobe 2991 cell processor, the Baxter CytoMate, or the Haemonetics Cell Saver 5) according to the manufacturer's instructions. After washing, the cells may be resuspended in a variety of biocompatible buffers, such as, for example, Ca-free, Mg-free PBS, PlasmaLyte A, or other saline solution with or without buffer. Alternatively, the undesirable components of the apheresis sample may be removed and the cells directly resuspended in culture media.


It is recognized that the methods of the application can utilize culture media conditions comprising 5% or less, for example 2%, human AB serum, and employ known culture media conditions and compositions, for example those described in Smith et al., “Ex vivo expansion of human T cells for adoptive immunotherapy using the novel Xeno-free CTS Immune Cell Serum Replacement” Clinical & Translational Immunology (2015) 4, e31; doi:10.1038/cti.2014.31.


In one aspect, T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLL™ gradient or by counterflow centrifugal elutriation.


The methods described herein can include, e.g., selection of a specific subpopulation of immune effector cells, e.g., T cells, that are a T regulatory cell-depleted population, CD25+ depleted cells, using, e.g., a negative selection technique, e.g., described herein. In some embodiments, the population of T regulatory depleted cells contains less than 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% of CD25+ cells.


In one embodiment, T regulatory cells, e.g., CD25+ T cells, are removed from the population using an anti-CD25 antibody, or fragment thereof, or a CD25-binding ligand, IL-2. In one embodiment, the anti-CD25 antibody, or fragment thereof, or CD25-binding ligand is conjugated to a substrate, e.g., a bead, or is otherwise coated on a substrate, e.g., a bead. In one embodiment, the anti-CD25 antibody, or fragment thereof, is conjugated to a substrate as described herein.


In one embodiment, the T regulatory cells, e.g., CD25+ T cells, are removed from the population using CD25 depletion reagent from Miltenyi™. In one embodiment, the ratio of cells to CD25 depletion reagent is 1e7 cells to 20 uL, or 1e7 cells to 15 uL, or 1e7 cells to 10 uL, or 1e7 cells to 5 uL, or 1e7 cells to 2.5 uL, or 1e7 cells to 1.25 uL. In one embodiment, e.g., for T regulatory cells, e.g., CD25+ depletion, greater than 500 million cells/ml is used. In a further aspect, a concentration of cells of 600, 700, 800, or 900 million cells/ml is used.


In one embodiment, the population of immune effector cells to be depleted includes about 6×109 CD25+ T cells. In other aspects, the population of immune effector cells to be depleted include about 1×109 to 1×1010 CD25+ T cell, and any integer value in between. In one embodiment, the resulting population T regulatory depleted cells has 2×109 T regulatory cells, e.g., CD25+ cells, or less (e.g., 1×109, 5×108, 1×108, 5×107, 1×107, or less CD25+ cells).


In one embodiment, the T regulatory cells, e.g., CD25+ cells, are removed from the population using the CliniMAC system with a depletion tubing set, such as, e.g., tubing 162-01. In one embodiment, the CliniMAC system is run on a depletion setting such as, e.g., DEPLETION2.1.


Without wishing to be bound by a particular theory, decreasing the level of negative regulators of immune cells (e.g., decreasing the number of unwanted immune cells, e.g., TREG cells), in a subject prior to apheresis or during manufacturing of a CAR-expressing cell product significantly reduces the risk of subject relapse. For example, methods of depleting TREG cells are known in the art. Methods of decreasing TREG cells include, but are not limited to, cyclophosphamide, anti-GITR antibody (an anti-GITR antibody described herein), CD25-depletion, mTOR inhibitor, and combinations thereof.


In some embodiments, the manufacturing methods comprise reducing the number of (e.g., depleting) TREG cells prior to manufacturing of the CAR-expressing cell. For example, manufacturing methods comprise contacting the sample, e.g., the apheresis sample, with an anti-GITR antibody and/or an anti-CD25 antibody (or fragment thereof, or a CD25-binding ligand), e.g., to deplete TREG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product.


Without wishing to be bound by a particular theory, decreasing the level of negative regulators of immune cells (e.g., decreasing the number of unwanted immune cells, e.g., TREG cells), in a subject prior to apheresis or during manufacturing of a CAR-expressing cell product can reduce the risk of a TREG relapse. In an embodiment, a subject is pre-treated with one or more therapies that reduce TREG cells prior to collection of cells for CAR-expressing cell product manufacturing, thereby reducing the risk of subject relapse to CAR-expressing cell treatment. In an embodiment, methods of decreasing TREG cells include, but are not limited to, administration to the subject of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, or a combination thereof. In an embodiment, methods of decreasing TREG cells include, but are not limited to, administration to the subject of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, mTOR inhibitor, or a combination thereof. Administration of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, or a combination thereof, can occur before, during or after an infusion of the CAR-expressing cell product. Administration of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, mTOR inhibitor, or a combination thereof, can occur before, during or after an infusion of the CAR-expressing cell product.


In some embodiments, the manufacturing methods comprise reducing the number of (e.g., depleting) TREG cells prior to manufacturing of the CAR-expressing cell. For example, manufacturing methods comprise contacting the sample, e.g., the apheresis sample, with an anti-GITR antibody and/or an anti-CD25 antibody (or fragment thereof, or a CD25-binding ligand), e.g., to deplete TREG cells prior to manufacturing of the CAR-expressing cell (e.g., T cell, NK cell) product.


In an embodiment, a subject is pre-treated with one or more therapies that reduce TREG cells prior to collection of cells for CAR-expressing cell product manufacturing, thereby reducing the risk of subject relapse to CAR-expressing cell treatment. In an embodiment, methods of decreasing TREG cells include, but are not limited to, administration to the subject of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, or a combination thereof. Administration of one or more of cyclophosphamide, anti-GITR antibody, CD25-depletion, or a combination thereof, can occur before, during or after an infusion of the CAR-expressing cell product.


In an embodiment, a subject is pre-treated with cyclophosphamide prior to collection of cells for CAR-expressing cell product manufacturing, thereby reducing the risk of subject relapse to CAR-expressing cell treatment. In an embodiment, a subject is pre-treated with an anti-GITR antibody prior to collection of cells for CAR-expressing cell product manufacturing, thereby reducing the risk of subject relapse to CAR-expressing cell treatment.


In one embodiment, the population of cells to be removed are neither the regulatory T cells or tumor cells, but cells that otherwise negatively affect the expansion and/or function of CART cells, e.g. cells expressing CD14, CD11b, CD33, CD15, or other markers expressed by potentially immune suppressive cells. In one embodiment, such cells are envisioned to be removed concurrently with regulatory T cells and/or tumor cells, or following said depletion, or in another order.


The methods described herein can include more than one selection step, e.g., more than one depletion step. Enrichment of a T cell population by negative selection can be accomplished, e.g., with a combination of antibodies directed to surface markers unique to the negatively selected cells. One method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected. For example, to enrich for CD4+ cells by negative selection, a monoclonal antibody cocktail can include antibodies to CD14, CD20, CD11b, CD16, HLA-DR, and CD8.


The methods described herein can further include removing cells from the population which express a tumor antigen, e.g., a tumor antigen that does not comprise CD25, e.g., CD19, CD30, CD38, CD123, CD20, CD14 or CD11b, to thereby provide a population of T regulatory depleted, e.g., CD25+ depleted, and tumor antigen depleted cells that are suitable for expression of a CAR, e.g., a CAR described herein. In one embodiment, tumor antigen expressing cells are removed simultaneously with the T regulatory, e.g., CD25+ cells. For example, an anti-CD25 antibody, or fragment thereof, and an anti-tumor antigen antibody, or fragment thereof, can be attached to the same substrate, e.g., bead, which can be used to remove the cells or an anti-CD25 antibody, or fragment thereof, or the anti-tumor antigen antibody, or fragment thereof, can be attached to separate beads, a mixture of which can be used to remove the cells. In other embodiments, the removal of T regulatory cells, e.g., CD25+ cells, and the removal of the tumor antigen expressing cells is sequential, and can occur, e.g., in either order.


Also provided are methods that include removing cells from the population which express a check point inhibitor, e.g., a check point inhibitor described herein, e.g., one or more of PD1+ cells, LAG3+ cells, and TIM3+ cells, to thereby provide a population of T regulatory depleted, e.g., CD25+ depleted cells, and check point inhibitor depleted cells, e.g., PD1+, LAG3+ and/or TIM3+ depleted cells. Exemplary check point inhibitors include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GALS, adenosine, and TGFR (e.g., TGFRbeta), e.g., as described herein. In one embodiment, check point inhibitor expressing cells are removed simultaneously with the T regulatory, e.g., CD25+ cells. For example, an anti-CD25 antibody, or fragment thereof, and an anti-check point inhibitor antibody, or fragment thereof, can be attached to the same bead which can be used to remove the cells, or an anti-CD25 antibody, or fragment thereof, and the anti-check point inhibitor antibody, or fragment thereof, can be attached to separate beads, a mixture of which can be used to remove the cells. In other embodiments, the removal of T regulatory cells, e.g., CD25+ cells, and the removal of the check point inhibitor expressing cells is sequential, and can occur, e.g., in either order.


Methods described herein can include a positive selection step For example, T cells can be isolated by incubation with anti-CD3/anti-CD28 (e.g., 3×28)-conjugated beads, such as DYNABEADS® M-450 CD3/CD28 T, for a time period sufficient for positive selection of the desired T cells. In one aspect, the time period is about 30 minutes. In a further aspect, the time period ranges from 30 minutes to 36 hours or longer and all integer values there between. In a further aspect, the time period is at least 1, 2, 3, 4, 5, or 6 hours. In yet another aspect, the time period is 10 to 24 hours. In one aspect, the incubation time period is 24 hours. Longer incubation times may be used to isolate T cells in any situation where there are few T cells as compared to other cell types, such in isolating tumor infiltrating lymphocytes (TIL) from tumor tissue or from immunocompromised individuals. Further, use of longer incubation times can increase the efficiency of capture of CD8+ T cells. Thus, by simply shortening or lengthening the time T cells are allowed to bind to the CD3/CD28 beads and/or by increasing or decreasing the ratio of beads to T cells (as described further herein), subpopulations of T cells can be preferentially selected for or against at culture initiation or at other time points during the process. Additionally, by increasing or decreasing the ratio of anti-CD3 and/or anti-CD28 antibodies on the beads or other surface, subpopulations of T cells can be preferentially selected for or against at culture initiation or at other desired time points.


In one embodiment, a T cell population can be selected that expresses one or more of IFN-γ, TNFα, IL-17A, IL-2, IL-3, IL-4, GM-CSF, IL-10, IL-13, granzyme B, and perforin, or other appropriate molecules, e.g., other cytokines. Methods for screening for cell expression can be determined, e.g., by the methods described in PCT Publication No.: WO 2013/126712.


For isolation of a desired population of cells by positive or negative selection, the concentration of cells and surface (e.g., particles such as beads) can be varied. In certain aspects, it may be desirable to significantly decrease the volume in which beads and cells are mixed together (e.g., increase the concentration of cells), to ensure maximum contact of cells and beads. For example, in one aspect, a concentration of about 10 billion cells/ml, 9 billion/ml, 8 billion/ml, 7 billion/ml, 6 billion/ml, or 5 billion/ml is used. In one aspect, a concentration of 1 billion cells/ml is used. In one aspect, a concentration of cells from 75, 80, 85, 90, 95, or 100 million cells/ml is used. In further aspects, concentrations of 125 or 150 million cells/ml can be used.


Using high concentrations can result in increased cell yield, cell activation, and cell expansion. Further, use of high cell concentrations allows more efficient capture of cells that may weakly express target antigens of interest, such as CD28-negative T cells, or from samples where there are many tumor cells present (e.g., leukemic blood, tumor tissue, etc.). Such populations of cells may have therapeutic value and would be desirable to obtain. For example, using high concentration of cells allows more efficient selection of CD8+ T cells that normally have weaker CD28 expression.


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


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


T cells for stimulation can also be frozen after a washing step. Wishing not to be bound by theory, the freeze and subsequent thaw step provides a more uniform product by removing granulocytes and to some extent monocytes in the cell population. After the washing step that removes plasma and platelets, the cells may be suspended in a freezing solution. While many freezing solutions and parameters are known in the art and will be useful in this context, one method involves using PBS containing 20% DMSO and 8% human serum albumin, or culture media containing 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin and 7.5% DMSO, or 31.25% Plasmalyte-A, 31.25% Dextrose 5%, 0.45% NaCl, 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin, and 7.5% DMSO or other suitable cell freezing media containing for example, Hespan and PlasmaLyte A, the cells then are frozen to −80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank. Other methods of controlled freezing may be used as well as uncontrolled freezing immediately at −20° C. or in liquid nitrogen.


In certain aspects, cryopreserved cells are thawed and washed as described herein and allowed to rest for one hour at room temperature prior to activation using the methods of the present invention.


Also contemplated in the context of the invention is the collection of blood samples or apheresis product from a subject at a time period prior to when the expanded cells as described herein might be needed. As such, the source of the cells to be expanded can be collected at any time point necessary, and desired cells, such as T cells, isolated and frozen for later use in immune effector cell therapy for any number of diseases or conditions that would benefit from immune effector cell therapy, such as those described herein. In one aspect a blood sample or an apheresis is taken from a generally healthy subject. In certain aspects, a blood sample or an apheresis is taken from a generally healthy subject who is at risk of developing a disease, but who has not yet developed a disease, and the cells of interest are isolated and frozen for later use. In certain aspects, the T cells may be expanded, frozen, and used at a later time. In certain aspects, samples are collected from a patient shortly after diagnosis of a particular disease as described herein but prior to any treatments. In a further aspect, the cells are isolated from a blood sample or an apheresis from a subject prior to any number of relevant treatment modalities, including but not limited to treatment with agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxan, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, and irradiation.


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


In one embodiment, the immune effector cells expressing a CAR molecule, e.g., a CAR molecule described herein, are obtained from a subject that has received a low, immune enhancing dose of an mTOR inhibitor. In an embodiment, the population of immune effector cells, e.g., T cells, to be engineered to express a CAR, are harvested after a sufficient time, or after sufficient dosing of the low, immune enhancing, dose of an mTOR inhibitor, such that the level of PD1 negative immune effector cells, e.g., T cells, or the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells, in the subject or harvested from the subject has been, at least transiently, increased.


In other embodiments, population of immune effector cells, e.g., T cells, which have, or will be engineered to express a CAR, can be treated ex vivo by contact with an amount of an mTOR inhibitor that increases the number of PD1 negative immune effector cells, e.g., T cells or increases the ratio of PD1 negative immune effector cells, e.g., T cells/PD1 positive immune effector cells, e.g., T cells.


In one embodiment, a T cell population is diacylglycerol kinase (DGK)-deficient. DGK-deficient cells include cells that do not express DGK RNA or protein, or have reduced or inhibited DGK activity. DGK-deficient cells can be generated by genetic approaches, e.g., administering RNA-interfering agents, e.g., siRNA, shRNA, miRNA, to reduce or prevent DGK expression. Alternatively, DGK-deficient cells can be generated by treatment with DGK inhibitors described herein.


In one embodiment, a T cell population is Ikaros-deficient. Ikaros-deficient cells include cells that do not express Ikaros RNA or protein, or have reduced or inhibited Ikaros activity, Ikaros-deficient cells can be generated by genetic approaches, e.g., administering RNA-interfering agents, e.g., siRNA, shRNA, miRNA, to reduce or prevent Ikaros expression. Alternatively, Ikaros-deficient cells can be generated by treatment with Ikaros inhibitors, e.g., lenalidomide.


In embodiments, a T cell population is DGK-deficient and Ikaros-deficient, e.g., does not express DGK and Ikaros, or has reduced or inhibited DGK and Ikaros activity. Such DGK and Ikaros-deficient cells can be generated by any of the methods described herein.


In an embodiment, the NK cells are obtained from the subject. In another embodiment, the NK cells are an NK cell line, e.g., NK-92 cell line (Conkwest).


Allogeneic CAR

In embodiments described herein, the immune effector cell can be an allogeneic immune effector cell, e.g., T cell or NK cell. For example, the cell can be an allogeneic T cell, e.g., an allogeneic T cell lacking expression of a functional T cell receptor (TCR) and/or human leukocyte antigen (HLA), e.g., HLA class I and/or HLA class II.


A T cell lacking a functional TCR can be, e.g., engineered such that it does not express any functional TCR on its surface, engineered such that it does not express one or more subunits that comprise a functional TCR (e.g., engineered such that it does not express (or exhibits reduced expression) of TCR alpha, TCR beta, TCR gamma, TCR delta, TCR epsilon, and/or TCR zeta) or engineered such that it produces very little functional TCR on its surface (e.g., engineered such that it does not express (or exhibits reduced expression) of TCR alpha, TCR beta, TCR gamma, TCR delta, TCR epsilon, and/or TCR zeta). Alternatively, the T cell can express a substantially impaired TCR, e.g., by expression of mutated or truncated forms of one or more of the subunits of the TCR. The term “substantially impaired TCR” means that this TCR will not elicit an adverse immune reaction in a host.


A T cell described herein can be, e.g., engineered such that it does not express a functional HLA on its surface. For example, a T cell described herein, can be engineered such that cell surface expression HLA, e.g., HLA class 1 and/or HLA class II, is downregulated. In some embodiments, downregulation of HLA may be accomplished by reducing or eliminating expression of beta-2 microglobulin (B2M).


In some embodiments, the T cell can lack a functional TCR and a functional HLA, e.g., HLA class I and/or HLA class II.


Modified T cells that lack expression of a functional TCR and/or HLA can be obtained by any suitable means, including a knock out or knock down of one or more subunit of TCR or HLA. For example, the T cell can include a knock down of TCR and/or HLA using siRNA, shRNA, clustered regularly interspaced short palindromic repeats (CRISPR) transcription-activator like effector nuclease (TALEN), or zinc finger endonuclease (ZFN).


In some embodiments, the allogeneic cell can be a cell which does not express or expresses at low levels an inhibitory molecule, e.g. a cell engineered by any method described herein. For example, the cell can be a cell that does not express or expresses at low levels an inhibitory molecule, e.g., that can decrease the ability of a CAR-expressing cell to mount an immune effector response. Examples of inhibitory molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAGS, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GALS, adenosine, and TGFR (e.g., TGFR beta). Inhibition of an inhibitory molecule, e.g., by inhibition at the DNA, RNA or protein level, can optimize a CAR-expressing cell performance. In embodiments, an inhibitory nucleic acid, e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used.


siRNA and shRNA to Inhibit TCR or HLA


In some embodiments, TCR expression and/or HLA expression can be inhibited using siRNA or shRNA that targets a nucleic acid encoding a TCR and/or HLA, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR beta), in a T cell.


Expression systems for siRNA and shRNAs, and exemplary shRNAs, are described, e.g., in paragraphs 649 and 650 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.


CRISPR to Inhibit TCR or HLA

“CRISPR” or “CRISPR to TCR and/or HLA” or “CRISPR to inhibit TCR and/or HLA” as used herein refers to a set of clustered regularly interspaced short palindromic repeats, or a system comprising such a set of repeats. “Cas”, as used herein, refers to a CRISPR-associated protein. A “CRISPR/Cas” system refers to a system derived from CRISPR and Cas which can be used to silence or mutate a TCR and/or HLA gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR beta).


The CRISPR/Cas system, and uses thereof, are described, e.g., in paragraphs 651-658 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.


TALEN to Inhibit TCR and/or HLA


“TALEN” or “TALEN to HLA and/or TCR” or “TALEN to inhibit HLA and/or TCR” refers to a transcription activator-like effector nuclease, an artificial nuclease which can be used to edit the HLA and/or TCR gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR beta).


TALENs, TALEs, and uses thereof, are described, e.g., in paragraphs 659-665 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.


Zinc Finger Nuclease to Inhibit HLA and/or TCR


“ZFN” or “Zinc Finger Nuclease” or “ZFN to HLA and/or TCR” or “ZFN to inhibit HLA and/or TCR” refer to a zinc finger nuclease, an artificial nuclease which can be used to edit the HLA and/or TCR gene, and/or an inhibitory molecule described herein (e.g., PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GAL9, adenosine, and TGFR beta).


ZFNs, and uses thereof, are described, e.g., in paragraphs 666-671 of International Application WO2015/142675, filed Mar. 13, 2015, which is incorporated by reference in its entirety.


Telomerase Expression

While not wishing to be bound by any particular theory, in some embodiments, a therapeutic T cell has short term persistence in a patient, due to shortened telomeres in the T cell; accordingly, transfection with a telomerase gene can lengthen the telomeres of the T cell and improve persistence of the T cell in the patient. See Carl June, “Adoptive T cell therapy for cancer in the clinic”, Journal of Clinical Investigation, 117:1466-1476 (2007). Thus, in an embodiment, an immune effector cell, e.g., a T cell, ectopically expresses a telomerase subunit, e.g., the catalytic subunit of telomerase, e.g., TERT, e.g., hTERT. In some aspects, this disclosure provides a method of producing a CAR-expressing cell, comprising contacting a cell with a nucleic acid encoding a telomerase subunit, e.g., the catalytic subunit of telomerase, e.g., TERT, e.g., hTERT. The cell may be contacted with the nucleic acid before, simultaneous with, or after being contacted with a construct encoding a CAR.


In one aspect, the disclosure features a method of making a population of immune effector cells (e.g., T cells or NK cells). In an embodiment, the method comprises: providing a population of immune effector cells (e.g., T cells or NK cells), contacting the population of immune effector cells with a nucleic acid encoding a CAR; and contacting the population of immune effector cells with a nucleic acid encoding a telomerase subunit, e.g., hTERT, under conditions that allow for CAR and telomerase expression.


In an embodiment, the nucleic acid encoding the telomerase subunit is DNA. In an embodiment, the nucleic acid encoding the telomerase subunit comprises a promoter capable of driving expression of the telomerase subunit.


In an embodiment, hTERT has the amino acid sequence of GenBank Protein ID AAC51724.1 (Meyerson et al., “hEST2, the Putative Human Telomerase Catalytic Subunit Gene, Is Up-Regulated in Tumor Cells and during Immortalization” Cell Volume 90, Issue 4, 22 Aug. 1997, Pages 785-795). The amino acid and nucleic acid sequences of hTERT are disclosed in International Application WO 2016/164731 filed on Apr. 8, 2016, the entire contents of which are hereby incorporated by reference.


Activation and Expansion of Immune Effector Cells (e.g., T Cells)

Immune effector cells such as T cells may be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005.


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


The method of expansion of immune effector cells, and methods of introducing CAR nucleic acid molecules into immune effector cells, and method of detecting CAR expression is described on pages 236-246 of in International Application WO 2016/164731 filed on Apr. 8, 2016, which is incorporated by reference in its entirety.


Other assays, including those described in the Example section herein as well as those that are known in the art can also be used to evaluate the CARs described herein.


Alternatively, or in combination to the methods disclosed herein, methods and compositions for one or more of detection and/or quantification of CAR-expressing cells (e.g., in vitro or in vivo (e.g., clinical monitoring)), immune cell expansion and/or activation, and/or CAR-specific selection, that involve the use of a CAR ligand, are disclosed. In one exemplary embodiment, the CAR ligand is an antibody that binds to the CAR molecule, e.g., binds to the extracellular antigen binding domain of CAR (e.g., an antibody that binds to the antigen binding domain, e.g., an anti-idiotypic antibody; or an antibody that binds to a constant region of the extracellular binding domain). In other embodiments, the CAR ligand is a CAR antigen molecule (e.g., a CAR antigen molecule as described herein).


In yet other embodiments, a method for depleting (e.g., reducing and/or killing) a CAR expressing cell is provided. The method includes contacting the CAR expressing cell with a CAR ligand as described herein; and targeting the cell on the basis of binding of the CAR ligand thereby reducing the number, and/or killing, the CAR-expressing cell. In one embodiment, the CAR ligand is coupled to a toxic agent (e.g., a toxin or a cell ablative drug). In another embodiment, the anti-idiotypic antibody can cause effector cell activity, e.g., ADCC or ADC activities.


Exemplary anti-CAR antibodies that can be used in the methods disclosed herein are described, e.g., in WO 2014/190273 and by Jena et al., “Chimeric Antigen Receptor (CAR)-Specific Monoclonal Antibody to Detect CD19-Specific T cells in Clinical Trials”, PLOS March 2013 8:3 e57838, the contents of which are incorporated by reference. In some aspects and embodiments, the compositions and methods herein are optimized for a specific subset of T cells, e.g., as described in US Serial No. PCT/US2015/043219 filed Jul. 31, 2015, the contents of which are incorporated herein by reference in their entirety. In some embodiments, the optimized subsets of T cells display an enhanced persistence compared to a control T cell, e.g., a T cell of a different type (e.g., CD8+ or CD4+) expressing the same construct.


In some embodiments, a CD4+ T cell comprises a CAR described herein, which CAR comprises an intracellular signaling domain suitable for (e.g., optimized for, e.g., leading to enhanced persistence in) a CD4+ T cell, e.g., an ICOS domain. In some embodiments, a CD8+ T cell comprises a CAR described herein, which CAR comprises an intracellular signaling domain suitable for (e.g., optimized for, e.g., leading to enhanced persistence of) a CD8+ T cell, e.g., a 4-1BB domain, a CD28 domain, or another costimulatory domain other than an ICOS domain. In some embodiments, the CAR described herein comprises an antigen binding domain described herein, e.g., a CAR comprising an antigen binding domain.


In an aspect, described herein is a method of treating a subject, e.g., a subject having cancer. The method includes administering to said subject, an effective amount of:


1) a CD4+ T cell comprising a CAR (the CARCD4+) comprising:


an antigen binding domain, e.g., an antigen binding domain described herein;


a transmembrane domain; and


an intracellular signaling domain, e.g., a first costimulatory domain, e.g., an ICOS domain; and


2) a CD8+ T cell comprising a CAR (the CARCD8+) comprising:


an antigen binding domain, e.g., an antigen binding domain described herein;


a transmembrane domain; and


an intracellular signaling domain, e.g., a second costimulatory domain, e.g., a 4-1BB domain, a CD28 domain, or another costimulatory domain other than an ICOS domain;


wherein the CARCD4+ and the CARCD8+ differ from one another.


Optionally, the method further includes administering:


3) a second CD8+ T cell comprising a CAR (the second CARCD8+) comprising:


an antigen binding domain, e.g., an antigen binding domain described herein;


a transmembrane domain; and


an intracellular signaling domain, wherein the second CARCD8+ comprises an intracellular signaling domain, e.g., a costimulatory signaling domain, not present on the CARCD8+, and, optionally, does not comprise an ICOS signaling domain.


Methods of Manufacture/Production

In some embodiments, the methods disclosed herein further include administering a T cell depleting agent after treatment with the cell (e.g., an immune effector cell as described herein, e.g., an immune effector cell expressing CAR driven by a truncated PGK1 promoter), thereby reducing (e.g., depleting) the CAR-expressing cells (e.g., the CD19CAR-expressing cells). Such T cell depleting agents can be used to effectively deplete CAR-expressing cells (e.g., CD19CAR-expressing cells) to mitigate toxicity. In some embodiments, the CAR-expressing cells were manufactured according to a method herein, e.g., assayed (e.g., before or after transfection or transduction) according to a method herein.


In some embodiments, the T cell depleting agent is administered one, two, three, four, or five weeks after administration of the cell, e.g., the population of immune effector cells, described herein.


In one embodiment, the T cell depleting agent is an agent that depletes CAR-expressing cells, e.g., by inducing antibody dependent cell-mediated cytotoxicity (ADCC) and/or complement-induced cell death. For example, CAR-expressing cells described herein may also express an antigen (e.g., a target antigen) that is recognized by molecules capable of inducing cell death, e.g., ADCC or complement-induced cell death. For example, CAR expressing cells described herein may also express a target protein (e.g., a receptor) capable of being targeted by an antibody or antibody fragment. Examples of such target proteins include, but are not limited to, EpCAM, VEGFR, integrins (e.g., integrins ανβ3, α4, αI¾β3, α4β7, α5β1, ανβ3, αν), members of the TNF receptor superfamily (e.g., TRAIL-R1, TRAIL-R2), PDGF Receptor, interferon receptor, folate receptor, GPNMB, ICAM-1, HLA-DR, CEA, CA-125, MUC1, TAG-72, IL-6 receptor, 5T4, GD2, GD3, CD2, CD3, CD4, CD5, CD11, CD11a/LFA-1, CD15, CD18/ITGB2, CD19, CD20, CD22, CD23/lgE Receptor, CD25, CD28, CD30, CD33, CD38, CD40, CD41, CD44, CD51, CD52, CD62L, CD74, CD80, CD125, CD147/basigin, CD152/CTLA-4, CD154/CD40L, CD195/CCR5, CD319/SLAMF7, and EGFR, and truncated versions thereof (e.g., versions preserving one or more extracellular epitopes but lacking one or more regions within the cytoplasmic domain).


In some embodiments, the CAR expressing cell co-expresses the CAR and the target protein, e.g., naturally expresses the target protein or is engineered to express the target protein. For example, the cell, e.g., the population of immune effector cells, can include a nucleic acid (e.g., vector) comprising the CAR nucleic acid (e.g., a CAR nucleic acid as described herein) and a nucleic acid encoding the target protein.


In one embodiment, the T cell depleting agent is a CD52 inhibitor, e.g., an anti-CD52 antibody molecule, e.g., alemtuzumab.


In further embodiments of any of the aforesaid methods, the methods further include transplanting a cell, e.g., a hematopoietic stem cell, or a bone marrow, into the mammal.


In another aspect, the invention features a method of conditioning a mammal prior to cell transplantation. The method includes administering to the mammal an effective amount of the cell comprising a CAR nucleic acid or polypeptide, e.g., a CD19 CAR nucleic acid or polypeptide. In some embodiments, the cell transplantation is a stem cell transplantation, e.g., a hematopoietic stem cell transplantation, or a bone marrow transplantation. In other embodiments, conditioning a subject prior to cell transplantation includes reducing the number of target-expressing cells in a subject, e.g., CD19-expressing normal cells or CD19-expressing cancer cells.


Biopolymer Delivery Methods

In some embodiments, one or more CAR-expressing cells as disclosed herein can be administered or delivered to the subject via a biopolymer scaffold, e.g., a biopolymer implant. Biopolymer scaffolds can support or enhance the delivery, expansion, and/or dispersion of the CAR-expressing cells described herein. A biopolymer scaffold comprises a biocompatible (e.g., does not substantially induce an inflammatory or immune response) and/or a biodegradable polymer that can be naturally occurring or synthetic. Exemplary biopolymers are described, e.g., in paragraphs 1004-1006 of International Application WO2015/142675, filed Mar. 13, 2015, which is herein incorporated by reference in its entirety.


Therapeutic Applications

CD19 or CD22 Associated Diseases and/or Disorders


In one aspect, the invention provides methods for treating a disease associated with CD19 or CD22 expression. In one aspect, the invention provides methods for treating a disease wherein part of the cancer is negative for CD19 and part of the cancer is positive for CD19. In one aspect, the invention provides methods for treating a disease wherein part of the cancer is negative for CD22 and part of the cancer is positive for CD22. For example, the methods and compositions of the invention are useful for treating subjects that have undergone treatment for a disease associated with expression of CD19, wherein the subject that has undergone treatment related to CD19 expression, e.g., treatment with a CD19 CAR, exhibits a disease associated with expression of CD19.


In another aspect, the invention provides methods for treating a disease associated with expression of a B-cell antigen, e.g., CD22. In one aspect, the invention provides methods for treating a disease wherein part of the tumor is negative for the B-cell antigen and part of the tumor is positive for B-cell antigen. For example, the compositions and methods of the invention are useful for treating subjects that have undergone treatment for a disease associated with expression of the B-cell antigen, wherein the subject that has undergone treatment related to expression of a B-cell antigen, e.g., treatment with a CAR targeting a B-cell antigen, exhibits a disease associated with expression of the B-cell antigen. In an aspect, the invention provides methods for treating a disease associated with expression of the B-cell antigen, e.g., associated with the expression of CD19 and one or more other B-cell antigens.


In one aspect, the invention pertains to a vector comprising CD19 CAR or CD22 CAR operably linked to promoter for expression in mammalian cells, e.g., T cells or NK cells. In one aspect, the invention provides a recombinant cell, e.g., a T cell or NK cell, expressing the CD19 CAR or CD22 CAR for use in treating tumor antigen expressing cancers, e.g., CD19-expressing cancers or CD22 expressing cancers. In some embodiments, the recombinant T cell expressing the CD19 CAR is termed a CD19 CART. In one aspect, the CD19 CART described herein, is capable of contacting a cancer cell with at least one CD19 CAR expressed on its surface such that the CART targets the cancer cell and growth of the cancer is inhibited. In some embodiments, the recombinant T cell expressing the CD22 CAR is termed a CD22 CART. In one aspect, the CD22 CART described herein, is capable of contacting a cancer cell with at least one CD22 CAR expressed on its surface such that the CART targets the cancer cell and growth of the cancer is inhibited.


In one aspect the invention pertains to a CD22 inhibitor which is a CD22 CART, e.g., a T cell, expressing the CD22 CAR for use in treating CD22-expressing tumors in combination with CD19 CARTS, wherein the recombinant T cell expressing the CD22 CAR is termed a CD22 CART. In one aspect, the CD22 CART described herein, is capable of contacting a tumor cell with at least one CD22 CAR expressed on its surface such that the CD22 CART targets the tumor cell and growth of the tumor is inhibited.


In one aspect, the invention pertains to a method of inhibiting growth of a tumor antigen expressing cancer cell, e.g., CD19-expressing cancer cell and/or CD22-expressing cancer cell, comprising contacting the cancer cell with a CD19 CAR expressing cell, e.g., a CD19 CART cell, and a CD22 CAR-expressing cell, e.g., a CD22 CART cell, such that the one or more CART are activated in response to the antigen, e.g., CD19 and/or CD22, and targets the cancer cell, wherein the growth of the cancer is inhibited. The CD19 CAR-expressing cell, e.g., T cell, is administered in combination with a CD22 CAR-expressing cell.


In some embodiments, the CD19 inhibitor (e.g., one or more cells that express a CAR molecule that binds CD19, e.g., a CAR molecule that binds CD19 described herein) and the CD22 inhibitor, e.g., CD22 CAR-expressing cell are administered simultaneously. In some embodiments, the CD19 inhibitor and the CD22 cell inhibitor are infused into a subject simultaneously, e.g., are admixed in the same infusion volume. In other embodiments, the simultaneous administration comprises separate administration of the CD19 inhibitor and the CD22 cell inhibitor, e.g., administration of each is initiated within a predetermined time interval (e.g., within 15, 30, or 45 minutes of each other).


In some embodiments, the start of CD19 inhibitor delivery and the start of CD22 inhibitor delivery are within 1, 2, 3, 4, 6, 12, 18, or 24 hours of each other, or within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 60, 80, or 100 days of each other. In some embodiments, the end of CD19 inhibitor delivery and the end of CD22 inhibitor delivery are within 1, 2, 3, 4, 6, 12, 18, or 24 hours of each other, or within 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 60, 80, or 100 days of each other. In some embodiments, the overlap in terms of administration between the CD19 inhibitor delivery (e.g., infusion) and the end of CD22 inhibitor delivery (e.g., infusion) is at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, or 45 minutes.


In some embodiments, the one or more cells that express a CAR molecule that binds CD22 is administered while the one or more cells that express a CAR molecule that binds CD19 are present (e.g., undergoing expansion) in the subject. In some embodiments, the CD19 inhibitor is administered while the one or more cells that express a CAR molecule that binds CD22, are present (e.g., undergoing expansion) in the subject.


In one embodiment, the CAR expressing cell, e.g., T cell, is administered to a subject that has received a previous stem cell transplantation, e.g., autologous stem cell transplantation.


In one embodiment, the CAR expressing cell, e.g., T cell, is administered to a subject that has received chemotherapy, e.g., lymphodepleting chemotherapy, e.g., as described herein.


In one embodiment, the CAR expressing cell, e.g., T cell, is administered to a subject that has received chemotherapy, e.g., bridging chemotherapy, e.g., as described herein.


In some embodiments, the CAR expressing cell, e.g., CD22 CAR22-expressing cell, is administered to a subject who has previously received stem cell transplantation (SCT) therapy, e.g., allogeneic or autologous SCT. In some embodiments, the CAR expressing cell, e.g., CD22 CAR22-expressing cell, is administered to a subject who is not eligible for SCT therapy, e.g., e.g., allogeneic or autologous SCT. In some embodiments, the subject has previously received SCT therapy and the subject has, or is identified as having relapsed from the SCT therapy. In some embodiments, the relapse from SCT therapy comprises a blood or bone marrow relapse and/or the relapse occurs at least 1-12 months, e.g., at least 6 months, after SCT therapy.


In some embodiments, the CAR expressing cell, e.g., CD22 CAR-expressing cell, is administered to a subject who has, or is identified as having, a relapse, e.g., a second or greater relapse, e.g., bone marrow relapse, e.g., as described herein.


In some embodiments, the CAR expressing cell, e.g., CD22 CAR-expressing cell, is administered to a subject who has, or is identified as having relapsed from a CAR-expressing cell therapy, e.g., a CAR-expressing cell therapy other than a CAR22-expressing cell therapy or a CAR19-expressing cell therapy.


In some embodiments, the CAR expressing cell, e.g., CD22 CAR-expressing cell, is administered to a subject who has, or is identified as not having a response, e.g., a complete response or partial response, in response to one or more (e.g., 2, 3, 4, 5, 6, 7, or 8) chemotherapeutic agents, e.g., as described herein.


The invention includes (among other things) a type of cellular therapy where T cells are genetically modified to express a chimeric antigen receptor (CAR) and the CAR T cell is infused to a recipient in need thereof. The infused cell is able to kill tumor cells in the recipient. Unlike antibody therapies, CAR-modified T cells are able to replicate in vivo resulting in long-term persistence that can lead to sustained tumor control. In various aspects, the T cells administered to the patient, or their progeny, persist in the patient for at least four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, thirteen months, fourteen month, fifteen months, sixteen months, seventeen months, eighteen months, nineteen months, twenty months, twenty-one months, twenty-two months, twenty-three months, two years, three years, four years, or five years after administration of the T cell to the patient.


The invention also includes a type of cellular therapy where immune effector cells, e.g., NK cells or T cells are modified, e.g., by in vitro transcribed RNA, to transiently express a chimeric antigen receptor (CAR) and the CAR-expressing (e.g., CAR T) cell is infused to a recipient in need thereof. The infused cell is able to kill cancer cells in the recipient. Thus, in various aspects, the CAR-expressing cells, e.g., T cells, administered to the patient, is present for less than one month, e.g., three weeks, two weeks, one week, after administration of the CAR-expressing cell, e.g., T cell, to the patient.


Without wishing to be bound by any particular theory, the anti-cancer immunity response elicited by the CAR-modified T cells may be an active or a passive immune response, or alternatively may be due to a direct vs indirect immune response. In one aspect, the CAR (e.g., CD19-CAR) transduced T cells exhibit specific proinflammatory cytokine secretion and potent cytolytic activity in response to human cancer cells expressing the target antigen (e.g., CD19), resist soluble target antigen inhibition, mediate bystander killing and mediate regression of an established human cancer. For example, antigen-less cancer cells within a heterogeneous field of target antigen-expressing cancer may be susceptible to indirect destruction by target antigen-redirected T cells that has previously reacted against adjacent antigen-positive cancer cells.


In one aspect, the CAR-modified cells of the invention, e.g., fully human CAR T cells, may be a type of vaccine for ex vivo immunization and/or in vivo therapy in a mammal. In one aspect, the mammal is a human.


With respect to ex vivo immunization, at least one of the following occurs in vitro prior to administering the cell into a mammal: i) expansion of the cells, ii) introducing a nucleic acid encoding a CAR to the cells or iii) cryopreservation of the cells.


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


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


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


Generally, the cells activated and expanded as described herein may be utilized in the treatment and prevention of diseases that arise in individuals who are immunocompromised. In particular, the CAR-expressing cells described herein are used in the treatment of diseases, disorders and conditions associated with expression of one or more B-cell antigen. In certain aspects, the cells are used in the treatment of patients at risk for developing diseases, disorders and conditions associated with expression of one or more B-cell antigen. Thus, the present invention provides (among other things) methods for the treatment or prevention of diseases, disorders and conditions associated with expression of a B-cell antigen comprising administering to a subject in need thereof, a therapeutically effective amount of the CD19 CAR-expressing cells described herein, in combination with one or more of B-cell inhibitor described herein.


The present invention also provides methods for inhibiting the proliferation or reducing a tumor antigen expressing cell population, e.g., a CD19-expressing cell population and/or a CD22-expressing cell population, the methods comprising contacting a population of cells comprising a CD19-expressing cell, and/or a CD22-expressing cell with an anti-CD19 CAR-expressing cell, and/or a CD22-CAR expressing cell described herein, wherein the CD19-expressing cell binds to the CD19 expressing cell and/or the CD22-expressing cell, and the CD22-expressing cell binds to the CD19 expressing cell and/or the CD22-expressing cell.


In a specific aspect, the present invention provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19 and/or CD22, the methods comprising contacting the population of cells comprising CD19-expressing cancer cell and/or CD22-expressing cells with an anti-CD19 CAR-expressing cell described herein, and/or a CD22 CAR-expressing cell described herein, wherein the CD19-expressing cell binds to the CD19 expressing cell and/or the CD22-expressing cell, and the CD22-expressing cell binds to the CD19 expressing cell and/or the CD22-expressing cell.


In one aspect, the present invention provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19 and/or CD22, the methods comprising contacting the population of cells comprising CD19-expressing cancer cell, and/or CD22-expressing cance cells with an anti-CD19 CAR-expressing cell described herein, and/or a CD22 CAR-expressing cell described herein, wherein the CD19-expressing cell binds to the CD19 expressing cell and/or the CD22-expressing cell, and the CD22-expressing cell binds to the CD19 expressing cell and/or the CD22-expressing cell.


In certain aspects, the combination of the anti-CD19 CAR-expressing cell described herein and the anti-CD22 CAR-expressing cell described herein reduces the quantity, number, amount or percentage of cells, e.g., cells expressing CD19 and/or CD22, and/or cancer cells, e.g., cancer cells expressing CD19 and/or CD22, by at least 25%, at least 30%, at least 40%, at least 50%, at least 65%, at least 75%, at least 85%, at least 95%, or at least 99% in a subject with or animal model for a hematological cancer or another cancer associated with CD19-expressing cells, and/or CD22 expressing cells relative to a negative control. In one aspect, the subject is a human.


The present invention also provides methods for inhibiting the proliferation or reducing a cell population comprising CD19-expressing cells, and/or CD22-expressing cells. In one aspect, CD19 and CD22 antigen are expressed by the same cells within the population. In another aspect, CD19 and CD22 antigen are expressed by distinct subsets of cells within the population. In another aspect, CD19 and CD22 antigen are expressed by overlapping subsets of cells within the population, such that some cells express CD19 and CD22 antigen, some cells express CD19, and some cells express the CD22.


The present invention also provides methods for inhibiting the proliferation or reducing a cell population expressing CD19 and CD22, the methods comprising (i) contacting a population of cells comprising a CD19-expressing cell with an anti-CD19 CAR-expressing cell described herein that binds to the CD19-expressing cell, and (ii) contacting the CD22-expressing cell with a second CAR-expressing cell described herein that binds to the CD22-expressing cell. In a specific aspect, the present invention provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19 and a CD22, the methods comprising (i) contacting the CD19-expressing cancer cell population with an anti-CD19 CAR-expressing cell described herein that binds to the CD19-expressing cell, and (ii) contacting the CD22-expressing cell population with a second CAR-expressing cell described herein that binds to CD22 cell expressing. In one aspect, the present invention provides methods for inhibiting the proliferation or reducing the population of cancer cells expressing CD19 and/or a CD22, the methods comprising (i) contacting the CD19-expressing cancer cell population with an anti-CD19 CAR-expressing cell described herein that binds to the CD19-expressing cell and (ii) contacting the CD22-expressing cell population with a second CAR-expressing cell described herein that binds to the CD22 cell expressing the CD22. In certain aspects, the combination of the anti-CD19 CAR-expressing cell described herein and the anti-CD22 CAR-expressing cell described herein, reduces the quantity, number, amount or percentage of cells and/or cancer cells by at least 25%, at least 30%, at least 40%, at least 50%, at least 65%, at least 75%, at least 85%, at least 95%, or at least 99% in a subject with or animal model for a hematological cancer or another cancer associated with CD19 and/or CD22-expressing cells relative to a negative control. In one aspect, the subject is a human.


The present invention also provides methods for preventing, treating and/or managing a disease associated with CD19-expressing cells and/or CD22 expressing cells (e.g., a hematologic cancer or atypical cancer expressing CD19 and/or CD22), the methods comprising administering to a subject in need an anti-CD19 CAR-expressing cell that binds to the CD19-expressing cell and administering an anti-CD22 CAR-expressing cell that binds to the CD22-expressing cells. In one aspect, the subject is a human. Non-limiting examples of disorders associated with CD19-expressing cells and/or CD22-expressing cells include autoimmune disorders (such as lupus), inflammatory disorders (such as allergies and asthma) and cancers (such as hematological cancers or atypical cancers expressing CD19 and/or CD22, e.g., B-cell ALL, e.g., relapsed and/or refractory B-cell ALL).


In one aspect, the invention pertains to a method of treating cancer in a subject. The method comprises administering to the subject a CD19 CAR-expressing cell, e.g., T cell, described herein, in combination with a CD22 CAR-expressing cell, e.g., T cell, such that the cancer is treated in the subject. An example of a cancer that is treatable by the methods described herein is a cancer associated with expression of CD19 and/or CD22, e.g., B-cell ALL, e.g., relapsed and/or refractory ALL. In one embodiment, the disease is a solid or liquid tumor. In one embodiment, the disease is a hematologic cancer, e.g., as described herein.


Non-cancer related indications associated with expression of CD19 and/or CD22 include, but are not limited to, e.g., autoimmune disease, (e.g., lupus), inflammatory disorders (allergy and asthma) and transplantation.


The CAR-expressing cells described herein, e.g., CD19 CAR-expressing cells and/or CD22 CAR-expressing cells, may be administered either alone, or as a pharmaceutical composition in combination with diluents and/or with other components such as IL-2 or other cytokines or cell populations.


Hematologic Cancers

Hematological cancer conditions are the types of cancer such as leukemia, lymphoma and malignant lymphoproliferative conditions that affect blood, bone marrow and the lymphatic system.


In one embodiment, the hematologic cancer is leukemia, e.g., a relapsed or refractory leukemia. In one embodiment, the cancer is selected from the group consisting of one or more acute leukemias including but not limited to B-cell acute lymphoid leukemia (BALL), T-cell acute lymphoid leukemia (TALL), small lymphocytic leukemia (SLL), acute lymphoid leukemia (ALL); one or more chronic leukemias including but not limited to chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL); additional hematologic cancers or hematologic conditions including, but not limited to mantle cell lymphoma (MCL), B cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, Marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia” which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells. Diseases associated with CD19, or CD22 expression include, but not limited to atypical and/or non-classical cancers, malignancies, precancerous conditions or proliferative diseases expressing CD19, or CD22; and any combination thereof.


Leukemia can be classified as acute leukemia and chronic leukemia. Acute leukemia can be further classified as acute myelogenous leukemia (AML) and acute lymphoid leukemia (ALL). Chronic leukemia includes chronic myelogenous leukemia (CML) and chronic lymphoid leukemia (CLL). Other related conditions include myelodysplastic syndromes (MDS, formerly known as “preleukemia”) which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells and risk of transformation to AML.


Lymphoma is a group of blood cell tumors that develop from lymphocytes. Exemplary lymphomas include non-Hodgkin lymphoma and Hodgkin lymphoma.


In an aspect, the invention pertains to a method of treating a mammal having Hodgkin lymphoma, comprising administering to the mammal an effective amount of the cells expressing a CD19 CAR molecule, e.g., a CD19 CAR molecule described herein and a CD22 CAR molecule.


In one aspect, the compositions and CART cells or CAR expressing NK cells of the present invention are particularly useful for treating B cell malignancies, such as non-Hodgkin lymphomas, e.g., DLBCL, Follicular lymphoma, or CLL.


Non-Hodgkin lymphoma (NHL) is a group of cancers of lymphocytes, formed from either B or T cells. NHLs occur at any age and are often characterized by lymph nodes that are larger than normal, weight loss, and fever. Different types of NHLs are categorized as aggressive (fast-growing) and indolent (slow-growing) types. B-cell non-Hodgkin lymphomas include Burkitt lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), diffuse large B-cell lymphoma (DLBCL), follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, and mantle cell lymphoma. Examples of T-cell non-Hodgkin lymphomas include mycosis fungoides, anaplastic large cell lymphoma, and precursor T-lymphoblastic lymphoma. Lymphomas that occur after bone marrow or stem cell transplantation are typically B-cell non-Hodgkin lymphomas. See, e.g., Maloney. NEJM. 366.21(2012):2008-16.


Diffuse large B-cell lymphoma (DLBCL) is a form of NHL that develops from B cells. DLBCL is an aggressive lymphoma that can arise in lymph nodes or outside of the lymphatic system, e.g., in the gastrointestinal tract, testes, thyroid, skin, breast, bone, or brain. Three variants of cellular morphology are commonly observed in DLBCL: centroblastic, immunoblastic, and anaplastic. Centroblastic morphology is most common and has the appearance of medium-to-large-sized lymphocytes with minimal cytoplasm. There are several subtypes of DLBCL. For example, primary central nervous system lymphoma is a type of DLBCL that only affects the brain is called and is treated differently than DLBCL that affects areas outside of the brain. Another type of DLBCL is primary mediastinal B-cell lymphoma, which often occurs in younger patients and grows rapidly in the chest. Symptoms of DLBCL include a painless rapid swelling in the neck, armpit, or groin, which is caused by enlarged lymph nodes. For some subjects, the swelling may be painful. Other symptoms of DLBCL include night sweats, unexplained fevers, and weight loss. Although most patients with DLBCL are adults, this disease sometimes occurs in children. Treatment for DLBCL includes chemotherapy (e.g., cyclophosphamide, doxorubicin, vincristine, prednisone, etoposide), antibodies (e.g., Rituxan), radiation, or stem cell transplants.


Follicular lymphoma a type of non-Hodgkin lymphoma and is a lymphoma of follicle center B-cells (centrocytes and centroblasts), which has at least a partially follicular pattern. Follicular lymphoma cells express the B-cell markers CD10, CD19, CD20, and CD22. Follicular lymphoma cells are commonly negative for CD5. Morphologically, a follicular lymphoma tumor is made up of follicles containing a mixture of centrocytes (also called cleaved follicle center cells or small cells) and centroblasts (also called large noncleaved follicle center cells or large cells). The follicles are surrounded by non-malignant cells, mostly T-cells. The follicles contain predominantly centrocytes with a minority of centroblasts. The World Health Organization (WHO) morphologically grades the disease as follows: grade 1 (<5 centroblasts per high-power field (hpf); grade 2 (6-15 centroblasts/hpf); grade 3 (>15 centroblasts/hpf). Grade 3 is further subdivided into the following grades: grade 3A (centrocytes still present); grade 3B (the follicles consist almost entirely of centroblasts). Treatment of follicular lymphoma includes chemotherapy, e.g., alkyating agents, nucleoside analogs, anthracycline-containing regimens, e.g., a combination therapy called CHOP-cyclophosphamide, doxorubicin, vincristine, prednisone/prednisolone, antibodies (e.g., rituximab), radioimmunotherapy, and hematopoietic stem cell transplantation.


CLL is a B-cell malignancy characterized by neoplastic cell proliferation and accumulation in bone morrow, blood, lymph nodes, and the spleen. The median age at time of diagnosis of CLL is about 65 years. Current treatments include chemotherapy, radiation therapy, biological therapy, or bone marrow transplantation. Sometimes symptoms are treated surgically (e.g., splenectomy removal of enlarged spleen) or by radiation therapy (e.g., de-bulking swollen lymph nodes). Chemotherapeutic agents to treat CLL include, e.g., fludarabine, 2-chlorodeoxyadenosine (cladribine), chlorambucil, vincristine, pentostatin, cyclophosphamide, alemtuzumab (Campath-1H), doxorubicin, and prednisone. Biological therapy for CLL includes antibodies, e.g., alemtuzumab, rituximab, and ofatumumab; as well as tyrosine kinase inhibitor therapies. A number of criteria can be used to classify stage of CLL, e.g., the Rai or Binet system. The Rai system describes CLL has having five stages: stage 0 where only lymphocytosis is present; stage I where lymphadenopathy is present; stage II where splenomegaly, lymphadenopathy, or both are present; stage III where anemia, organomegaly, or both are present (progression is defined by weight loss, fatigue, fever, massive organomegaly, and a rapidly increasing lymphocyte count); and stage IV where anemia, thrombocytopenia, organomegaly, or a combination thereof are present. Under the Binet staging system, there are three categories: stage A where lymphocytosis is present and less than three lymph nodes are enlarged (this stage is inclusive of all Rai stage 0 patients, one-half of Rai stage I patients, and one-third of Rai stage II patients); stage B where three or more lymph nodes are involved; and stage C wherein anemia or thrombocytopenia, or both are present. These classification systems can be combined with measurements of mutation of the immunoglobulin genes to provide a more accurate characterization of the state of the disease. The presence of mutated immunoglobulin genes correlates to improved prognosis.


ALL and Response Criteria

Acute lymphoblastic leukemia (ALL) is a B-cell malignancy characterized by neoplastic cell proliferation and accumulation in bone morrow, blood, lymph nodes, and the spleen. ALL can arise in adults or in pediatric populations, and can progress rapidly and can be fatal if left untreated. ALL includes relapsed and/or refractory ALL (r/r ALL). For relapsed and/or refractory ALL, treatment options include high-dose chemotherapy with subsequent allogeneic stem cell transplantation (SCT), standard chemo-immunotherapy, targeted treatment with small molecule pathway inhibitors, or supportive care with non-curative palliative goals. Allogeneic SCT is the only potentially curative option for r/r pediatric ALL, but outcomes are suboptimal. Among relapsed and/or refractory pediatric ALL patients who received allogeneic SCT in third or later remission, received allogeneic SCT with active disease or received allogeneic SCT after relapse from previous allogeneic SCT, the 1-year overall survival (OS) rates are in 25 to 55% range and 5-year OS rates are generally in 20 to 45% range.


For ALL patients presenting who are positive for the Philadelphia chromosome (Ph+), dasatinib (Sprycel) was approved in 2006 for the treatment of adult patients with resistance or intolerance to prior therapy. Ponatinib (Iclusig) was approved in 2013 for the treatment of adult patients with Ph+ ALL who are resistant to or intolerant of dasatinib. Blincyto (blinatumomab), a bispecific anti-CD3/CD19 monoclonal antibody, has been approved for the treatment of adults with Ph-relapsed or refractory B-precursor ALL. Despite the current treatment modalities, maintaining a remission in relapsed ALL patients is difficult, and the patients are being hospitalized for a long periods of time with a poor quality of life. The prognosis of patients with relapsed and/or refractory disease still remains poor.


In some embodiments, a subject having ALL, e.g., relapsed and/or refractory ALL, has or is identified as having a complete response to a CAR-expressing cell therapy, e.g., a CD22 CAR-expressing cell therapy. In some embodiments, a complete response comprises one or more, e.g., all, of the following characteristics:

    • (i) bone marrow, e.g., having trilineage hematopoiesis (TLH) and <5% blasts
    • (ii) peripheral blood having, e.g., Neutrophils >1.0×109/L; Platelets >100×109/L; or Circulating blasts <1%;
    • (iii) lack of evidence of extramedullary disease (e.g., lack of CNS disease, mediastinal disease CR, or other extramedullary sites involvement); or
    • (iv) transfusion independency, e.g., platelet and/or neutrophil transfusions within 1 week before peripheral blood sample for disease assessment.


In some embodiments, a subject having ALL, e.g., relapsed and/or refractory ALL, has or is identified as having a complete response with incomplete blood count recovery (CRi) to a CAR-expressing cell therapy, e.g., a CD22 CAR-expressing cell therapy. In some embodiments, a CRi comprises one or more, e.g., all, of the following characteristics:

    • (i) bone marrow, e.g., having trilineage hematopoiesis (TLH) and <5% blasts
    • (ii) peripheral blood having, e.g., Neutrophils <1.0×109/L; Platelets <100×109/L; or Circulating blasts <1%;
    • (iii) lack of evidence of extramedullary disease (e.g., lack of CNS disease, mediastinal disease CR, or other extramedullary sites involvement); or
    • (iv) platelet and/or neutrophil transfusions within 1 week before peripheral blood sample for disease assessment.


In some embodiments, a subject having ALL, e.g., relapsed and/or refractory ALL, has or is identified as having a complete response with residual mediastinal disease to a CAR-expressing cell therapy, e.g., a CD22 CAR-expressing cell therapy.


In some embodiments, a subject having ALL, e.g., relapsed and/or refractory ALL, has or is identified as having no response to a CAR-expressing cell therapy, e.g., a CD22 CAR-expressing cell therapy.


In some embodiments, a subject having ALL, e.g., relapsed and/or refractory ALL, has or is identified as having relapsed disease to a CAR-expressing cell therapy, e.g., a CD22 CAR-expressing cell therapy. In some embodiments, a relapsed disease comprises one or more, e.g., all, of the following characteristics:

    • (i) reappearance of blasts in the blood, e.g., more than 1%;
    • (ii) reappearance of blasts in the bone marrow, e.g., more than 5%; or
    • (iii) appearance or reappearance of extramedullary disease after a complete response, e.g., as described herein.


In another embodiment, the CAR expressing cells of the present invention are used to treat cancers or leukemias, e.g., with leukemia stem cells. For example, the leukemia stem cells are CD34+/CD38 leukemia cells.


In some embodiments, the subject is a non-responder to CD19 CAR therapy. In some embodiments, the subject is a partial responder to CD19 CAR therapy. In some embodiments, the subject is a complete responder to CD19 CAR therapy. In some embodiments, the subject is a non-relapser to CD19 CAR therapy. In some embodiments, the subject is a partial relapser to CD19 CAR therapy. In some embodiments, the subject is a complete relapser to CD19 CAR therapy.


In some embodiments, a cancer or other condition that was previously responsive to treatment with CD19 CAR-expressing cells does not express CD19. In some embodiments, a cancer or other condition that was previously responsive to treatment with CD19 CAR-expressing cells has a 10%, 20%, 30%, 40%, 50% or more reduction in CD19 expression levels relative to when the cancer or other condition was responsive to treatment with CD19 CAR-expressing cells. In some embodiments, a cancer or other condition that was previously responsive to treatment with CD19 CAR-expressing cells expresses CD22.


In some embodiments, the CD22 CAR-expressing cell of the invention is administered post-relapse of a cancer or other condition previously treated with CD19 CAR-expressing cell. In some embodiments, a CD19 CAR-expressing cell and a CD22 CAR-expressing cell are administered concurrently, as described herein.


Dosing Regimens

In some embodiments, a CAR-expressing cell, e.g., a CD19 CAR-expressing cell described herein or a CD22 CAR-expressing cell described herein, is administered to the subject according to a dosing regimen comprising a dose, e.g., a total dose, of cells administered to the subject by dose fractionation (e.g., split dosing), e.g., one, two, three or more separate administration of a partial dose, e.g., one, two, three, four, five or six partial doses. In embodiments, a dose of cells expressing a CAR molecule, e.g., a total dose of cells expressing a CAR molecule, comprises one or more partial doses, e.g., one, two, three, four, five or six partial doses, e.g., three partial doses. In embodiments, a total dose of cells expressing a CAR molecule comprises three partial doses. In embodiments, a first percentage of the total dose, e.g., a first partial dose, is administered on a first day of treatment, a second percentage of the total dose, e.g., a second partial dose, is administered on a subsequent (e.g., second, third, fourth, fifth, sixth, or seventh or later) day of treatment, and a third percentage (e.g., the remaining percentage) of the total dose, e.g., a third partial dose, is administered on a yet subsequent (e.g., third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, or later) day of treatment.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15% (e.g., about 5-10%, 10-15%, 6-14%, 7-13%, 8-12%, or 9-11%), of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15%, e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15%, of the total dose. In an embodiment, a first percentage of a total dose, e.g., a first partial dose, comprises about 5-15% of the total dose, e.g., about 0.1-1.0×106 cells/kg of the CAR-expressing cells. In some embodiments, a first percentage of a total dose, e.g., a first partial dose, comprises about 10% of the total dose (e.g., about 0.2×106 cells/kg) of the CAR-expressing cells when the total dose is about 2.0×106 cells/kg.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% (e.g., about 25-30%, 30-35%, 26-34%, 27-33%, 28-32%, or 29-31%) of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35%, e.g., about 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35%, of the total dose. In an embodiment, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% of the total dose, e.g., about 0.2-6.0×106 cells/kg, of the CAR expressing cells. In some embodiments, a second percentage of a total dose, e.g., a second partial dose, comprises about 30% of the total dose (e.g., about 0.6×106 cells/kg) of the CAR-expressing cells when the total dose is about 2.0×10 6 cells/kg. In some embodiments, a second percentage of a total dose, e.g., a second partial dose, is administered, e.g., delivered or infused, on the second day, e.g., second consecutive day.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% (e.g., about 50-55%, 55-60%, 56-64, 57-63, 58-62, or 59-61%) of the total dose. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65%, e.g., about 55, 56, 57, 58, 59, 60, 61, 62, 63, 64 or 65%, of the total dose. In an embodiment, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% of the total dose, e.g., about 0.5-12×106 cells/kg of the CAR expressing cells. In some embodiments a third percentage of a total dose, e.g., a third partial dose, comprises about 60% of the total dose (e.g., about 1.2×106 cells/kg) of the CAR-expressing cells when the total dose is about 2.0×106 cells/kg. In some embodiments, a third percentage of a total dose, e.g., a third partial dose, is administered, e.g., delivered or infused on the third da, e.g., third consecutive day


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, about 10% of the total dose of cells is delivered on the first day, about 30% of the total dose of cells is delivered on the second day (e.g., second consecutive day), and the remaining about 60% of the total dose of cells is delivered on the third day of treatment, e.g., third consecutive day of treatment.


In an embodiment, a dose of CAR-expressing cells, e.g., a total dose of CAR-expressing cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5-20×106 cells/kg, e.g., about 0.5-1×106 cells/kg, 1-5×106 cells/kg, 5-10×106 cells/kg, 10-15×106 cells/kg, 15-20×106 cells/kg, 0.6-24×106 cells/kg, 0.7-23×106 cells/kg, 0.8-22×106 cells/kg, 0.9-21×106 cells/kg, 1-20×106 cells/kg, 2-19×106 cells/kg, 3-18×106 cells/kg, 4-17×106 cells/kg, 5-16×106 cells/kg, 6-15×106 cells/kg, 7-14×106 cells/kg, 8-13×106 cells/kg, 9-12×106 cells/kg, or 10-11×106 cells/kg. In an embodiment, a dose of cells, e.g., a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5-20×106 cells/kg, e.g., about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, or 20×106 cells/kg. In an embodiment, a dose of CAR-expressing cells, e.g., a total cell dose (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 1-5×106 cells/kg, e.g., about 1-5×106 cells/kg, 1.5-4×106 cells/kg, 1.8-3.5×106 cells/kg, or about 1×106 cells/kg, 1.5×106 cells/kg, 2×106 cells/kg, 3×106 cells/kg, 4×106 cells/kg, or 5×106 cells/kg, e.g., about 2.0×106 cells/kg.


In an embodiment, a dose of CAR-expressing cells, e.g., a total dose of CAR-expressing cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5-20×108 cells, e.g., about 0.5-1×108 cells, 1-5×108 cells, 5-10×108 cells, 10-15×108 cells, 15-20×108 cells, 0.6-24×108 cells, 0.7-23×108 cells, 0.8-22×108 cells, 0.9-21×108 cells, 1-20×108 cells, 2-19×108 cells, 3-18×108 cells, 4-17×108 cells, 5-16×108 cells, 6-15×108 cells, 7-14×108 cells, 8-13×108 cells, 9-12×108 cells, or 10-11×108 cells. In an embodiment, a dose of cells, e.g., a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5-20×108 cells, e.g., about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10, 10.5, 11, 11.5, 12, 12.5, 13, 13.5, 14, 14.5, 15, 15.5, 16, 16.5, 17, 17.5, 18, 18.5, 19, 19.5, or 20×108 cells. In an embodiment, a dose of CAR-expressing cells, e.g., a total cell dose (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 1-5×108 cells, e.g., about 1-5×108 cells, 1.5-4×108 cells, 1.8-3.5×108 cells, or about 1×108 cells, 1.5×108 cells, 2×108 cells, 3×108 cells, 4×108 cells, or 5×108 cells, e.g., about 2.0×108 cells.


In an embodiment, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about about 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.02×107 to 1×107, about 1×107 to 1.5×107, about 1.5×107 to 2×107, about 2×107 to 2.5×107, about 2.5×107 to 3×107, about 3×107 to 3.5×107, about 3.5×107 to 4×107, about 4×107 to 4.5×107, or about 4.5×107 to 5×107 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.02×107, about 0.04×107, about 0.06×107, about 0.08×107, about 1×107, about 1.5×107, about 2×107, about 2.5×107, about 3×107, about 3.5×107, about 4×107, about 4.5×107, or about 5×107 CAR-expressing cells/kg, e.g., CD22 CAR-expressing cells/kg. In some embodiments, the subject weighs less than 50 kg.


In an embodiment, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5×108 to 1×108, about 1×108 to 1.5×108, about 1.5×108 to 2×108, about 2×108 to 2.5×108, about 2.5×108 to 3×108, about 3×108 to 3.5×108, about 3.5×108 to 4×108, about 4×108 to 4.5×108, about 4.5×108 to 5×108, about 5×108 to 6×108, about 6×108 to 7×108, about 7×108 to 8×108, about 8×108 to 9×108, or about 9×108 to 10×108 CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, a total dose of cells (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 0.5×108, about 1×108, about 1.5×108, about 2×108, about 2.5×108, about 3×108, about 3.5×108, about 4×108, about 4.5×108, about 5×108, about 6×108, about 7×108, about 8×108, about 9×108, about 10×108 CAR-expressing cells, e.g., CD22 CAR-expressing cells. In some embodiments, the subject weighs ≥50 kg.


In one embodiment, the total cell dose is about 1-5×106 cells/kg, e.g., about 2.0×106 cells/kg, e.g., 2.0×106 cells/kg of a CAR expressing cell, e.g., a CD19 CAR expressing cell or a CD22 CAR expressing cell.


In one embodiment, the total cell dose, e.g., combined dose of CD19 CAR expressing cell and a CD22 CAR expressing cell, is about 5-7×106 cells/kg, e.g., about 4.0×106 cells/kg. In one embodiment, the total cell dose, e.g., combined dose of CD19 CAR expressing cell and a CD22 CAR expressing cell, is about 1-5×108 CD22 CAR-expressing cells, and about 5-7×106 CD19 CAR expressing cells/kg, e.g., about 4.0×106 cells/kg. In one embodiment, the CD22 CAR-expressing cells, e.g., one or more doses of the CD22 CAR-expressing cells, e.g., according to a dosing regimen described herein, are administered before, after or concurrently with the administration of the CD19 CAR-expressing cells, e.g., one or more doses (e.g., all, e.g., total dose as a single infusion) of the CD19 CAR-expressing cells.


In one embodiment, the subject administered a CAR expressing cell according to a dosing regimen described herein, e.g., dose fractionation or split-dosing, has received chemotherapy, e.g., lymphodepleting chemotherapy comprising cyclophosphamide and fludarabine, e.g., according to a dosing regimen described herein. In one embodiment, the subject has ALL, e.g., relapsed and/or refractory ALL. In one embodiment, the subject is a young adult or a pediatric subject, e.g., aged about 1-29 years, e.g., aged 1-29 years. In one embodiment, the subject has failed, e.g., relapsed or not responded to, one or more (e.g., two, three, four or five) previous therapies, e.g., chemotherapies, e.g., standard of care, e.g., as described herein.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15% (e.g., about 5-10%, 10-15%, 6-14%, 7-13%, 8-12%, or 9-11%), of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15%, e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15%, of the total dose. In an embodiment, when a subject weighs less than 50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 5-15% of the total dose. In some embodiments, when a subject weighs less than 50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 10% of the total dose (e.g., about 0.2×106 to 1×106) of the CAR-expressing cells when the total dose is about 0.2×107 to 1.0×107 cells/kg. In some embodiments, a first percentage of a total dose, e.g., a first partial dose, is administered, e.g., delivered or infused, on the first day. In some embodiments, when a subject weighs less than 50 kg, a first partial dose, e.g., about 10% of the total dose (e.g., about 0.2×106 to 1×106 cells/kg), comprises CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% (e.g., about 25-30%, 30-35%, 26-34%, 27-33%, 28-32%, or 29-31%) of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35%, e.g., about 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35%, of the total dose. In an embodiment, when a subject weighs less than 50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% of the total dose. In some embodiments, when a subject weighs less than 50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 30% of the total dose (e.g., about 0.6×106 to 3×106 cells/kg) of the CAR-expressing cells when the total dose is about 0.2×107 to 1.0×107 cells/kg. In some embodiments, a second percentage of a total dose, e.g., a second partial dose, is administered, e.g., delivered or infused, on the second day, e.g., second consecutive day. In some embodiments, when a subject weighs less than 50 kg, a second partial dose, e.g., about 30% of the total dose (e.g., about 0.6×106 to 3×106 cells/kg), comprises CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% (e.g., about 50-55%, 55-60%, 56-64, 57-63, 58-62, 59-61%) of the total dose. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65%, e.g., about 55, 56, 57, 58, 59, 60, 61, 62, 63, 64 or 65%, of the total dose. In an embodiment, when a subject weighs less than 50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% of the total dose. In some embodiments, when a subject weighs less than 50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 60% of the total dose (e.g., about 1.2×106 to 6×106 cells/kg) of the CAR-expressing cells when the total dose is about 0.2×107 to 1.0×107 cells/kg. In some embodiments, when a subject weighs less than 50 kg a third percentage of a total dose, e.g., a third partial dose, is administered, e.g., delivered or infused on the third day, e.g., third consecutive day. In some embodiments, when a subject weighs less than 50 kg a third partial dose, e.g., about 60% of the total dose (e.g., about 1.2×106 to 6×106 cells/kg), comprises CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15% (e.g., about 5-10%, 10-15%, 6-14%, 7-13%, 8-12%, or 9-11%), of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a first percentage of the total dose, e.g., a first partial dose, comprises about 5-15%, e.g., about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15%, of the total dose. In an embodiment, when a subject weighs >50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 5-15% of the total dose. In some embodiments, when a subject weighs ≥50 kg, a first percentage of a total dose, e.g., a first partial dose, comprises about 10% of the total dose (e.g., about 1×107 to 5×107 cells) of the CAR-expressing cells when the total dose is about 1×108 to 5×108 cells. In some embodiments, a first percentage of a total dose, e.g., a first partial dose, is administered, e.g., delivered or infused, on the first day. In some embodiments, when a subject weighs ≥50 kg, a first partial dose, e.g., about 10% of the total dose (e.g., about 1×107 to 5×107 cells), comprises CAR-expressing cells, e.g., CD22 CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% (e.g., about 25-30%, 30-35%, 26-34%, 27-33%, 28-32%, or 29-31%) of the total dose of cells. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35%, e.g., about 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35%, of the total dose. In an embodiment, when a subject weighs ≥50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 25-35% of the total dose. In some embodiments, when a subject weighs ≥50 kg, a second percentage of a total dose, e.g., a second partial dose, comprises about 30% of the total dose (e.g., about 0.3×107 to 1.5×108 cells) of the CAR-expressing cells when the total dose is about 1×108 to 5×108 cells. In some embodiments, a second percentage of a total dose, e.g., a second partial dose, is administered, e.g., delivered or infused, on the second day, e.g., second consecutive day. In some embodiments, when a subject weighs ≥50 kg, a second partial dose, e.g., about 30% of the total dose (e.g., about 0.3×107 to 1.5×108 cells), comprises CAR-expressing cells, e.g., CD22 CAR-expressing cells.


In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% (e.g., about 50-55%, 55-60%, 56-64, 57-63, 58-62, or 59-61%) of the total dose. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65%, e.g., about 55, 56, 57, 58, 59, 60, 61, 62, 63, 64 or 65%, of the total dose. In an embodiment, when a subject weighs ≥50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 55-65% of the total dose. In some embodiments, when a subject weighs ≥50 kg a third percentage of a total dose, e.g., a third partial dose, comprises about 60% of the total dose (e.g., about 0.6×108 to 3×108 cells) of the CAR-expressing cells when the total dose is about 1×108 to 5×108cells. In some embodiments, when a subject ≥50 kg a third percentage of a total dose, e.g., a third partial dose, is administered, e.g., delivered or infused on the third day, e.g., third consecutive day. In some embodiments, when a subject weighs ≥50 kg a third partial dose, e.g., about 60% of the total dose (e.g., about 0.6×108 to 3×108 cells), comprises CAR-expressing cells, e.g., CD22 CAR-expressing cells.


In one embodiment, a CD19 CAR-expressing cell described herein is administered to the subject according to a dosing regimen comprising a total dose of CD19 CAR-expressing cells administered to the subject by dose fractionation (e.g., split dosing), e.g., three separate administrations of a partial dose of the CD19 CAR-expressing cells. In one embodiment, the total cell dose of the CD19 CAR-expressing cells is about 2.0×106 cells/kg. In an embodiment, of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, about 10% of the total dose (e.g., about 0.2×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the first day, about 30% of the total dose (e.g., about 0.6×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day), and the remaining about 60% of the total dose (e.g., about 1.2×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive day of treatment. In one embodiment, the subject has received chemotherapy, e.g., lymphodepleting chemotherapy comprising cyclophosphamide and fludarabine, e.g., according to a dosing regimen described herein. In one embodiment, the subject has ALL, e.g., relapsed and/or refractory ALL. In one embodiment, the subject is a young adult or a pediatric subject, e.g., is at about 1-29 years of age. In one embodiment, the subject has failed, e.g., relapsed or not responded to, one or more (e.g., two, three, four or five) previous therapies, e.g., chemotherapies, e.g., standard of care, e.g., as described herein.


In one embodiment, a CD22 CAR-expressing cell described herein is administered to the subject according to a dosing regimen comprising a total dose of CD22 CAR-expressing cells administered to the subject by dose fractionation (e.g., split dosing), e.g., three separate administrations of a partial dose of the CD22 CAR-expressing cells. In one embodiment, the total cell dose of the CD22 CAR-expressing cells is 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CD22 CAR-expressing cells/kg or about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CD22 CAR-expressing cells. In an embodiment, of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, about 10% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the first day, about 30% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day), and the remaining about 60% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive day of treatment. In one embodiment, the subject has received chemotherapy, e.g., lymphodepleting chemotherapy comprising cyclophosphamide and fludarabine, e.g., according to a dosing regimen described herein. In one embodiment, the subject has ALL, e.g., relapsed and/or refractory ALL. In one embodiment, the subject is a young adult or a pediatric subject, e.g., aged about 1-29 years, e.g., aged 1-29 years. In one embodiment, the subject has failed, e.g., relapsed or not responded to, one or more (e.g., two, three, four or five) previous therapies, e.g., chemotherapies, e.g., standard of care, e.g., as described herein.


In one embodiment, a CD22 CAR-expressing cell described herein, and a CD19 CAR-expressing cell described herein is administered to the subject according to a dosing regimen comprising a total dose of CD22 CAR-expressing cells and CD19 CAR-expressing cells administered to the subject by dose fractionation (e.g., split dosing), e.g., three separate administrations of a partial dose of each of the CD22 CAR-expressing cells and CD19 CAR-expressing cells. In one embodiment, the total cell dose of the CD22 CAR-expressing cells is 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CD22 CAR-expressing cells/kg or about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CD22 CAR-expressing cells. In one embodiment, the total cell dose of the CD19 CAR-expressing cells is about 2.0×106 cells/kg. In one embodiment, the CD22-CAR expressing cells, administered according to a dosing regimen described herein, e.g., a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein are administered before the administration of CD19 CAR-expressing cells. In one embodiment, the total dose of CD22 CAR-expressing cells comprising three separate administrations of a partial dose of CD22-CAR expressing cells is administered prior to the administration of CD19 CAR-expressing cells. In one embodiment, the total cell dose of the CD22 CAR-expressing cells is 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CD22 CAR-expressing cells/kg or about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CD22 CAR-expressing cells. In an embodiment, of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, about 10% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the first day, about 30% of the total of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day), and the remaining about 60% of the total dose of the CD22 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive day of treatment. In an embodiment, the CD19 CAR-expressing cell is administered after the administration of all three partial doses of the CD22 CAR-expressing cell, e.g., according to a dose fractionation regimen described herein. In one embodiment, the total cell dose of the CD19 CAR-expressing cells is about 2.0×106 cells/kg. In an embodiment, of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, about 10% of the total dose (e.g., about 0.2×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the first day, about 30% of the total dose (e.g., about 0.6×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the second day (e.g., second consecutive day), and the remaining about 60% of the total dose (e.g., about 1.2×106 cells/kg) of the CD19 CAR-expressing cells is administered, e.g., delivered or infused, on the third day of treatment, e.g., third consecutive day of treatment. In one embodiment, the subject has received chemotherapy, e.g., lymphodepleting chemotherapy comprising cyclophosphamide and fludarabine, e.g., according to a dosing regimen described herein. In one embodiment, the CD22 CAR-expressing cells, e.g., one or more doses of the CD22 CAR-expressing cells, e.g., according to a dosing regimen described herein, are administered before, after or concurrently with the administration of the CD19 CAR-expressing cells, e.g., one or more doses (e.g., all, e.g., total dose as a single infusion) of the CD19 CAR-expressing cells.


In one embodiment, the CD22 CAR-expressing cells, e.g., one or more doses of the CD22 CAR-expressing cells, e.g., according to a dosing regimen described herein, are administered before the administration of the CD19 CAR-expressing cells, e.g., at least about 1 week before, e.g., at least about 1-6 days, 2-5 days, 3-4 days before, or at least about 1, 2, 3, 4, 5, 6, or 7 days (e.g., about 23, 22, 20, 21, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or 0.5 hours) before the administration of the CD19 CAR-expressing cells, e.g., one or more doses (e.g., all doses, e.g., total dose as a single infusion) of the CD19 CAR-expressing cells. In one embodiment, the subject has ALL, e.g., relapsed and/or refractory ALL. In one embodiment, the subject is a young adult or a pediatric subject, e.g., is at aged about 1-29 years. In one embodiment, the subject has failed, e.g., relapsed or not responded to, one or more (e.g., two, three, four or five) previous therapies, e.g., chemotherapies, e.g., standard of care, e.g., as described herein.


In one embodiment, the therapy described herein (e.g., a CD22 CAR therapy, and the cells expressing a CD19 CAR molecule, e.g., a CD19 CAR molecule described herein) are administered to a subject as a first line treatment for the disease, e.g., the cancer, e.g., the cancer described herein, e.g., ALL, e.g., B cell ALL, e.g., relapsed or refractory ALL. In another embodiment, the therapy described herein (e.g., a CD22 CAR therapy, and the cells expressing a CD19 CAR molecule, e.g., a CD19 CAR molecule described herein) are administered to a subject as a second, third, fourth, or fifth line treatment for a disease, e.g., a cancer, e.g., a cancer described herein, e.g., ALL, e.g., B cell ALL, e.g., relapsed and/or refractory ALL. In some embodiments, the subject has relapsed or is refractory to a prior line of treatment (e.g., as described herein), e.g., a first, second, or third line of treatment prior to administration of a CAR therapy described herein. In one embodiment, the subject is a young adult or a pediatric subject, e.g., aged about 1-29 years, e.g., aged 1-29 years.


In one embodiment, a CAR expressing cell, e.g., a CD19 CAR expressing cell or a CD22 CAR expressing cell, administered according to a dosing regimen comprising a dose fractionation (e.g., split dosing), is administered to a subject that has received chemotherapy, e.g., lymphodepleting chemotherapy, e.g., as described herein. In one embodiment, the subject has relapsed and/or refractory ALL. In one embodiment, the subject has previously received, e.g., prior to lymphodepleting chemotherapy, one, two, or more lines of therapy, e.g., chemotherapy, e.g., standard of care, and has relapsed or not responded to the previous therapy or therapies.


Bone Marrow Ablation

In one aspect, the present invention provides compositions and methods for bone marrow ablation. For example, in one aspect, the invention provides compositions and methods for eradication of at least a portion of existing bone marrow in a subject. It is described herein that, in certain instances, the CD19, or CD22 CAR expressing cell comprising a CAR of the present invention eradicates CD19, or CD22 positive bone marrow myeloid progenitor cells.


In one aspect, the present invention provides a method of treating cancer comprising bone marrow conditioning, where at least a portion of bone marrow of the subject is eradicated by the CD22 CAR cell (e.g., T cell or NK cell) of the invention. For example, in certain instances, the bone marrow of the subject comprises a malignant precursor cell that can be targeted and eliminated by the activity of the CD22 CAR cell (e.g., T cell or NK cell). In one aspect, a bone marrow conditioning therapy comprises administering a bone marrow or stem cell transplant to the subject following the eradication of native bone marrow. In one aspect, the bone marrow reconditioning therapy is combined with one or more other anti-cancer therapies, including, but not limited to anti-tumor CAR therapies, chemotherapy, radiation, and the like.


In one aspect, eradication of the administered CD19, or CD22 CAR expressing cells may be required prior to infusion of bone marrow or stem cell transplant. Eradication of the CD19, or CD22 expressing CAR cell (e.g., T cell or NK cell) may be accomplished using any suitable strategy or treatment, including, but not limited to, use of a suicide gene, limited CAR persistence using RNA encoded CARs, or anti-T cell modalities including antibodies or chemotherapy.


Combination Therapies

The combination of a CAR as described herein (e.g., a CD20 CAR, a CD22 CAR, or a CD19 CAR-expressing cell described herein e.g., and one or more B-cell inhibitors, e.g., as described herein) may be used in combination with other known agents and therapies.


B cell inhibitors, combination therapies comprising the same and uses thereof are described in International Application WO 2016/164731 filed on Apr. 8, 2016, which is incorporated by reference in its entirety.


In further aspects, the combination of the CAR-expressing cell described herein (e.g., a CD22 CAR, or a CD19 CAR-expressing cell, optionally in combination with one or more B-cell inhibitor) may be used in a treatment regimen in combination with surgery, chemotherapy, radiation, an mTOR pathway inhibitor, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, cytokines, and irradiation. peptide vaccine, such as that described in Izumoto et al. 2008 J Neurosurg 108:963-971.


In one embodiment, the combination of a CD19, or CD22 CAR-expressing cell described herein (e.g., and one or more B-cell inhibitor) can be used in combination with a chemotherapeutic agent. Exemplary chemotherapeutic agents include an anthracycline (e.g., doxorubicin (e.g., liposomal doxorubicin)); a vinca alkaloid (e.g., vinblastine, vincristine, vindesine, vinorelbine); an alkylating agent (e.g., cyclophosphamide, decarbazine, melphalan, ifosfamide, temozolomide); an immune cell antibody (e.g., alemtuzamab, gemtuzumab, rituximab, tositumomab); an antimetabolite (including, e.g., folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors (e.g., fludarabine)); a TNFR glucocorticoid induced TNFR related protein (GITR) agonist; a proteasome inhibitor (e.g., aclacinomycin A, gliotoxin or bortezomib); an immunomodulator such as thalidomide or a thalidomide derivative (e.g., lenalidomide).


General Chemotherapeutic agents considered for use in combination therapies include anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC-Dome®), dactinomycin (Actinomycin D, Cosmegan), daunorubicin hydrochloride (Cerubidine®), daunorubicin citrate liposome injection (DaunoXome®), dexamethasone, docetaxel (Taxotere®), doxorubicin hydrochloride (Adriamycin®, Rubex®), etoposide (Vepesid®), fludarabine phosphate (Fludara®), 5-fluorouracil (Adrucil®, Efudex®), flutamide (Eulexin®), tezacitibine, gemcitabine (difluorodeoxycitidine), hydroxyurea (Hydrea®), Idarubicin (Idamycin®), ifosfamide (IFEX®), irinotecan (Camptosar®), L-asparaginase (ELSPAR®), leucovorin calcium, melphalan (Alkeran®), 6-mercaptopurine (Purinethol®), methotrexate (Folex®), mitoxantrone (Novantrone®), mylotarg, paclitaxel (Taxol®), nab-paclitaxel (Abraxane®), phoenix (Yttrium90/MX-DTPA), pentostatin, polifeprosan 20 with carmustine implant (Gliadel®), tamoxifen citrate (Nolvadex®), teniposide (Vumon®), 6-thioguanine, thiotepa, tirapazamine (Tirazone®), topotecan hydrochloride for injection (Hycamptin®), vinblastine (Velban®), vincristine (Oncovin®), and vinorelbine (Navelbine®).


Treatment with a combination of a chemotherapeutic agent and a cell expressing a CAR molecule described herein can be used to treat a hematologic cancer described herein, e.g., AML. In embodiments, the combination of a chemotherapeutic agent and a CAR-expressing cell is useful for targeting, e.g., killing, cancer stem cells, e.g., leukemic stem cells, e.g., in subjects with AML. In embodiments, the combination of a chemotherapeutic agent and a CAR-expressing cell is useful for treating minimal residual disease (MRD). MRD refers to the small number of cancer cells that remain in a subject during treatment, e.g., chemotherapy, or after treatment. MRD is often a major cause for relapse. The present invention provides a method for treating cancer, e.g., MRD, comprising administering a chemotherapeutic agent in combination with a CAR-expressing cell, e.g., as described herein.


In an embodiment, the chemotherapeutic agent is administered prior to administration of the cell expressing a CAR molecule, e.g., a CAR molecule described herein. In chemotherapeutic regimens where more than one administration of the chemotherapeutic agent is desired, the chemotherapeutic regimen is initiated or completed prior to administration of a cell expressing a CAR molecule, e.g., a CAR molecule described herein. In embodiments, the chemotherapeutic agent is administered at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 20 days, 25 days, or 30 days prior to administration of the cell expressing the CAR molecule. In embodiments, the chemotherapeutic regimen is initiated or completed at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 20 days, 25 days, or 30 days prior to administration of the cell expressing the CAR molecule. In embodiments, the chemotherapeutic agent is a chemotherapeutic agent that increases expression of CD19, CD20, or CD22 on the cancer cells, e.g., the tumor cells, e.g., as compared to expression on normal or non-cancer cells. Expression can be determined, for example, by immunohistochemical staining or flow cytometry analysis. For example, the chemotherapeutic agent is cytarabine (Ara-C).


Anti-cancer agents of particular interest for combinations with the compounds of the present invention include: antimetabolites; drugs that inhibit either the calcium dependent phosphatase calcineurin or the p70S6 kinase FK506) or inhibit the p70S6 kinase; alkylating agents; mTOR inhibitors; immunomodulators; anthracyclines; vinca alkaloids; proteosome inhibitors; GITR agonists; protein tyrosine phosphatase inhibitors; a CDK4 kinase inhibitor; a BTK kinase inhibitor; a MKN kinase inhibitor; a DGK kinase inhibitor; or an oncolytic virus.


Exemplary antimetabolites include, without limitation, folic acid antagonists (also referred to herein as antifolates), pyrimidine analogs, purine analogs and adenosine deaminase inhibitors): methotrexate (Rheumatrex®, Trexall®), 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), cytarabine (Cytosar-U®, Tarabine PFS), 6-mercaptopurine (Puri-Nethol®)), 6-thioguanine (Thioguanine Tabloid®), fludarabine phosphate (Fludara®), pentostatin (Nipent®), pemetrexed (Alimta®), raltitrexed (Tomudex®), cladribine (Leustatin®), clofarabine (Clofarex®, Clolar®), mercaptopurine (Puri-Nethol®), capecitabine (Xeloda®), nelarabine (Arranon®), azacitidine (Vidaza®) and gemcitabine (Gemzar®). Preferred antimetabolites include, e.g., 5-fluorouracil (Adrucil®, Efudex®, Fluoroplex®), floxuridine (FUDF®), capecitabine (Xeloda®), pemetrexed (Alimta®), raltitrexed (Tomudex®) and gemcitabine (Gemzar®).


Exemplary alkylating agents include, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes): uracil mustard (Aminouracil Mustard®, Chlorethaminacil®, Demethyldopan®, Desmethyldopan®, Haemanthamine®, Nordopan®, Uracil nitrogen mustard®, Uracillost®, Uracilmostaza®, Uramustin®, Uramustine®), chlormethine (Mustargen®), cyclophosphamide (Cytoxan®, Neosar®, Clafen®, Endoxan®, Procytox®, Revimmune™), ifosfamide (Mitoxana®), melphalan (Alkeran®), Chlorambucil (Leukeran®), pipobroman (Amedel®, Vercyte®), triethylenemelamine (Hemel®, Hexalen®, Hexastat®), triethylenethiophosphoramine, Temozolomide (Temodar®), thiotepa (Thioplex®), busulfan (Busilvex®, Myleran®), carmustine (BiCNU®), lomustine (CeeNU®), streptozocin (Zanosar®), and Dacarbazine (DTIC-Dome®). Additional exemplary alkylating agents include, without limitation, Oxaliplatin (Eloxatin®); Temozolomide (Temodar® and Temodal®); Dactinomycin (also known as actinomycin-D, Cosmegen®); Melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, Alkeran®); Altretamine (also known as hexamethylmelamine (HMM), Hexalen®); Carmustine (BiCNU®); Bendamustine (Treanda®); Busulfan (Busulfex® and Myleran®); Carboplatin (Paraplatin®); Lomustine (also known as CCNU, CeeNU®); Cisplatin (also known as CDDP, Platinol® and Platinol®-AQ); Chlorambucil (Leukeran®); Cyclophosphamide (Cytoxan® and Neosar®); Dacarbazine (also known as DTIC, DIC and imidazole carboxamide, DTIC-Dome®); Altretamine (also known as hexamethylmelamine (HMM), Hexalen®); Ifosfamide (Ifex®); Prednumustine; Procarbazine (Matulane®); Mechlorethamine (also known as nitrogen mustard, mustine and mechloroethamine hydrochloride, Mustargen®); Streptozocin (Zanosar®); Thiotepa (also known as thiophosphoamide, TESPA and TSPA, Thioplex®); Cyclophosphamide (Endoxan®, Cytoxan®, Neosar®, Procytox®, Revimmune®); and Bendamustine HCl (Treanda®).


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with fludarabine, cyclophosphamide, and/or rituximab. In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with fludarabine, cyclophosphamide, and rituximab (FCR). In embodiments, the subject has CLL. For example, the subject has a deletion in the short arm of chromosome 17 (del(17p), e.g., in a leukemic cell). In other examples, the subject does not have a del(17p). In embodiments, the subject comprises a leukemic cell comprising a mutation in the immunoglobulin heavy-chain variable-region (IgVH) gene. In other embodiments, the subject does not comprise a leukemic cell comprising a mutation in the immunoglobulin heavy-chain variable-region (IgVH) gene. In embodiments, the fludarabine is administered at a dosage of about 10-50 mg/m2 (e.g., about 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-45, or 45-50 mg/m2), e.g., intravenously. In embodiments, the cyclophosphamide is administered at a dosage of about 200-300 mg/m2 (e.g., about 200-225, 225-250, 250-275, or 275-300 mg/m2), e.g., intravenously. In embodiments, the rituximab is administered at a dosage of about 400-600 mg/m2 (e.g., 400-450, 450-500, 500-550, or 550-600 mg/m2), e.g., intravenously.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with bendamustine and rituximab. In embodiments, the subject has CLL. For example, the subject has a deletion in the short arm of chromosome 17 (del(17p), e.g., in a leukemic cell). In other examples, the subject does not have a del(17p). In embodiments, the subject comprises a leukemic cell comprising a mutation in the immunoglobulin heavy-chain variable-region (IgVH) gene. In other embodiments, the subject does not comprise a leukemic cell comprising a mutation in the immunoglobulin heavy-chain variable-region (IgVH) gene. In embodiments, the bendamustine is administered at a dosage of about 70-110 mg/m2 (e.g., 70-80, 80-90, 90-100, or 100-110 mg/m2), e.g., intravenously. In embodiments, the rituximab is administered at a dosage of about 400-600 mg/m2 (e.g., 400-450, 450-500, 500-550, or 550-600 mg/m2), e.g., intravenously.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with rituximab, cyclophosphamide, doxorubicine, vincristine, and/or a corticosteroid (e.g., prednisone). In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with rituximab, cyclophosphamide, doxorubicine, vincristine, and prednisone (R-CHOP). In embodiments, the subject has diffuse large B-cell lymphoma (DLBCL). In embodiments, the subject has nonbulky limited-stage DLBCL (e.g., comprises a tumor having a size/diameter of less than 7 cm). In embodiments, the subject is treated with radiation in combination with the R-CHOP. For example, the subject is administered R-CHOP (e.g., 1-6 cycles, e.g., 1, 2, 3, 4, 5, or 6 cycles of R-CHOP), followed by radiation. In some cases, the subject is administered R-CHOP (e.g., 1-6 cycles, e.g., 1, 2, 3, 4, 5, or 6 cycles of R-CHOP) following radiation.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and/or rituximab. In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab (EPOCH-R). In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with dose-adjusted EPOCH-R (DA-EPOCH-R). In embodiments, the subject has a B cell lymphoma, e.g., a Myc-rearranged aggressive B cell lymphoma.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with rituximab and/or lenalidomide. Lenalidomide ((RS)-3-(4-Amino-1-oxo 1,3-dihydro-2H-isoindol-2-yl)piperidine-2,6-dione) is an immunomodulator. In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with rituximab and lenalidomide. In embodiments, the subject has follicular lymphoma (FL) or mantle cell lymphoma (MCL). In embodiments, the subject has FL and has not previously been treated with a cancer therapy. In embodiments, lenalidomide is administered at a dosage of about 10-20 mg (e.g., 10-15 or 15-20 mg), e.g., daily. In embodiments, rituximab is administered at a dosage of about 350-550 mg/m2 (e.g., 350-375, 375-400, 400-425, 425-450, 450-475, or 475-500 mg/m2), e.g., intravenously.


Exemplary mTOR inhibitors include, e.g., temsirolimus; ridaforolimus (formally known as deferolimus, (1R,2R,4S)-4-[(2R)-2 [(1R,9S,12S,15R,16E,18R,19R,21R, 23S,24E,26E,28Z,30S,32S,35R)-1,18-dihydroxy-19,30-dimethoxy-15,17,21,23, 29,35-hexamethyl-2,3,10,14,20-pentaoxo-11,36-dioxa-4-azatricyclo[30.3.1.04.9] hexatriaconta-16,24,26,28-tetraen-12-yl]propyl]-2-methoxycyclohexyl dimethylphosphinate, also known as AP23573 and MK8669, and described in PCT Publication No. WO 03/064383); everolimus (Afinitor® or RAD001); rapamycin (AY22989, Sirolimus®); simapimod (CAS 164301-51-3); emsirolimus, (5-{2,4-Bis[(3S)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidin-7-yl}-2-methoxyphenyl)methanol (AZD8055); 2-Amino-8-[trans-4-(2-hydroxyethoxy)cyclohexyl]-6-(6-methoxy-3-pyridinyl)-4-methyl-pyrido[2,3-d]pyrimidin-7(8H)-one (PF04691502, CAS 1013101-36-4); and N2-[1,4-dioxo-4-[[4-(4-oxo-8-phenyl-4H-1-benzopyran-2-yl)morpholinium-4-yl]methoxy]butyl]-L-arginylglycyl-L-α-aspartylL-serine-, inner salt (SF1126, CAS 936487-67-1) (SEQ ID NO: 1316), and XL765.


Exemplary immunomodulators include, e.g., afutuzumab (available from Roche®); pegfilgrastim (Neulasta®); lenalidomide (CC-5013, Revlimid®); thalidomide (Thalomid®), actimid (CC4047); and IRX-2 (mixture of human cytokines including interleukin 1, interleukin 2, and interferon γ, CAS 951209-71-5, available from IRX Therapeutics).


Exemplary anthracyclines include, e.g., doxorubicin (Adriamycin® and Rubex®); bleomycin (Lenoxane®); daunorubicin (dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, Cerubidine®); daunorubicin liposomal (daunorubicin citrate liposome, DaunoXome®); mitoxantrone (DHAD, Novantrone®); epirubicin (Ellence™); idarubicin (Idamycin®, Idamycin PFS®); mitomycin C (Mutamycin®); geldanamycin; herbimycin; ravidomycin; and desacetylravidomycin.


Exemplary vinca alkaloids include, e.g., vinorelbine tartrate (Navelbine®), Vincristine (Oncovin®), and Vindesine (Eldisine®)); vinblastine (also known as vinblastine sulfate, vincaleukoblastine and VLB, Alkaban-AQ® and Velban®); and vinorelbine (Navelbine®).


Exemplary proteosome inhibitors include bortezomib (Velcade®); carfilzomib (PX-171-007, (S)-4-Methyl-N—((S)-1-(((S)-4-methyl-1-((R)-2-methyloxiran-2-yl)-1-oxopentan-2-yl)amino)-1-oxo-3-phenylpropan-2-yl)-2-((S)-2-(2-morpholinoacetamido)-4-phenylbutanamido)-pentanamide); marizomib (NPI-0052); ixazomib citrate (MLN-9708); delanzomib (CEP-18770); and O-Methyl-N-[(2-methyl-5-thiazolyl)carbonyl]-L-seryl-O-methyl-N-[(1S)-2-[(2R)-2-methyl-2-oxiranyl]-2-oxo-1-(phenylmethyl)ethyl]-L-serinamide (ONX-0912).


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with brentuximab. Brentuximab is an antibody-drug conjugate of anti-CD30 antibody and monomethyl auristatin E. In embodiments, the subject has Hodgkin's lymphoma (HL), e.g., relapsed or refractory HL. In embodiments, the subject comprises CD30+ HL. In embodiments, the subject has undergone an autologous stem cell transplant (ASCT). In embodiments, the subject has not undergone an ASCT. In embodiments, brentuximab is administered at a dosage of about 1-3 mg/kg (e.g., about 1-1.5, 1.5-2, 2-2.5, or 2.5-3 mg/kg), e.g., intravenously, e.g., every 3 weeks.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with brentuximab and dacarbazine or in combination with brentuximab and bendamustine. Dacarbazine is an alkylating agent with a chemical name of 5-(3,3-Dimethyl-1-triazenyl)imidazole-4-carboxamide. Bendamustine is an alkylating agent with a chemical name of 4-[5-[Bis(2-chloroethyl)amino]-1-methylbenzimidazol-2-yl]butanoic acid. In embodiments, the subject has Hodgkin's lymphoma (HL). In embodiments, the subject has not previously been treated with a cancer therapy. In embodiments, the subject is at least 60 years of age, e.g., 60, 65, 70, 75, 80, 85, or older. In embodiments, dacarbazine is administered at a dosage of about 300-450 mg/m2 (e.g., about 300-325, 325-350, 350-375, 375-400, 400-425, or 425-450 mg/m2), e.g., intravenously. In embodiments, bendamustine is administered at a dosage of about 75-125 mg/m2 (e.g., 75-100 or 100-125 mg/m2, e.g., about 90 mg/m2), e.g., intravenously. In embodiments, brentuximab is administered at a dosage of about 1-3 mg/kg (e.g., about 1-1.5, 1.5-2, 2-2.5, or 2.5-3 mg/kg), e.g., intravenously, e.g., every 3 weeks.


In some embodiments, a CAR-expressing cell described herein is administered to a subject in combination with a CD20 inhibitor, e.g., an anti-CD20 antibody (e.g., an anti-CD20 mono- or bispecific antibody) or a fragment thereof. Exemplary anti-CD20 antibodies include but are not limited to rituximab, ofatumumab, ocrelizumab, veltuzumab, obinutuzumab, TRU-015 (Trubion Pharmaceuticals), ocaratuzumab, and Pro131921 (Genentech). See, e.g., Lim et al. Haematologica. 95.1 (2010): 135-43.


Dosing regimens comprising CD20 inhibitors are described in International Application WO 2016/164731 filed on Apr. 8, 2016, which is incorporated by reference in its entirety.


In some embodiments, one or more CAR-expressing cells described herein is administered in combination with an oncolytic virus. In embodiments, oncolytic viruses are capable of selectively replicating in and triggering the death of or slowing the growth of a cancer cell. In some cases, oncolytic viruses have no effect or a minimal effect on non-cancer cells. An oncolytic virus includes but is not limited to an oncolytic adenovirus, oncolytic Herpes Simplex Viruses, oncolytic retrovirus, oncolytic parvovirus, oncolytic vaccinia virus, oncolytic Sinbis virus, oncolytic influenza virus, or oncolytic RNA virus (e.g., oncolytic reovirus, oncolytic Newcastle Disease Virus (NDV), oncolytic measles virus, or oncolytic vesicular stomatitis virus (VSV)).


In some embodiments, the oncolytic virus is a virus, e.g., recombinant oncolytic virus, described in US2010/0178684 A1, which is incorporated herein by reference in its entirety. In some embodiments, a recombinant oncolytic virus comprises a nucleic acid sequence (e.g., heterologous nucleic acid sequence) encoding an inhibitor of an immune or inflammatory response, e.g., as described in US2010/0178684 A1, incorporated herein by reference in its entirety. In embodiments, the recombinant oncolytic virus, e.g., oncolytic NDV, comprises a pro-apoptotic protein (e.g., apoptin), a cytokine (e.g., GM-CSF, interferon-gamma, interleukin-2 (IL-2), tumor necrosis factor-alpha), an immunoglobulin (e.g., an antibody against ED-B firbonectin), tumor associated antigen, a bispecific adapter protein (e.g., bispecific antibody or antibody fragment directed against NDV HN protein and a T cell co-stimulatory receptor, such as CD3 or CD28; or fusion protein between human IL-2 and single chain antibody directed against NDV HN protein). See, e.g., Zamarin et al. Future Microbiol. 7.3(2012):347-67, incorporated herein by reference in its entirety. In some embodiments, the oncolytic virus is a chimeric oncolytic NDV described in U.S. Pat. No. 8,591,881 B2, US 2012/0122185 A1, or US 2014/0271677 A1, each of which is incorporated herein by reference in their entireties.


In some embodiments, an oncolytic virus described herein is administering by injection, e.g., subcutaneous, intra-arterial, intravenous, intramuscular, intrathecal, or intraperitoneal injection. In embodiments, an oncolytic virus described herein is administered intratumorally, transdermally, transmucosally, orally, intranasally, or via pulmonary administration.


In an embodiment, cells expressing a CAR described herein are administered to a subject in combination with a molecule that decreases the Treg cell population. Methods that decrease the number of (e.g., deplete) Treg cells are known in the art and include, e.g., CD25 depletion, cyclophosphamide administration, modulating GITR function. Without wishing to be bound by theory, it is believed that reducing the number of Treg cells in a subject prior to apheresis or prior to administration of a CAR-expressing cell described herein reduces the number of unwanted immune cells (e.g., Tregs) in the tumor microenvironment and reduces the subject's risk of relapse.


In an embodiment, a CAR-expressing cell described herein is administered to a subject in combination with a molecule that decreases the Treg cell population. Methods that decrease the number of (e.g., deplete) Treg cells are known in the art and include, e.g., CD25 depletion, cyclophosphamide administration, and modulating GITR function. Without wishing to be bound by theory, it is believed that reducing the number of Treg cells in a subject prior to apheresis or prior to administration of a CAR-expressing cell described herein reduces the number of unwanted immune cells (e.g., Tregs) in the tumor microenvironment and reduces the subject's risk of relapse. In one embodiment, CAR-expressing cells described herein are administered to a subject in combination with a molecule targeting GITR and/or modulating GITR functions, such as a GITR agonist and/or a GITR antibody that depletes regulatory T cells (Tregs). In one embodiment, CAR-expressing cells described herein are administered to a subject in combination with cyclophosphamide. In one embodiment, the GITR binding molecule and/or molecule modulating GITR function (e.g., GITR agonist and/or Treg depleting GITR antibodies) is administered prior to the CAR-expressing cells. For example, in one embodiment, the GITR agonist can be administered prior to apheresis of the cells. In embodiments, cyclophosphamide is administered to the subject prior to administration (e.g., infusion or re-infusion) of the CAR-expressing cell or prior to apheresis of the cells. In embodiments, cyclophosphamide and an anti-GITR antibody are administered to the subject prior to administration (e.g., infusion or re-infusion) of the CAR-expressing cell or prior to apheresis of the cells. In one embodiment, the subject has cancer (e.g., a solid cancer or a hematological cancer such as ALL or CLL). In one embodiment, the subject has CLL. In embodiments, the subject has a solid cancer, e.g., a solid cancer described herein.


In one embodiment, a CAR expressing cell described herein is administered to a subject in combination with a GITR agonist, e.g., a GITR agonist described herein. In one embodiment, the GITR agonist is administered prior to the CAR-expressing cell. For example, in one embodiment, the GITR agonist can be administered prior to apheresis of the cells.


Exemplary GITR agonists include, e.g., GITR fusion proteins and anti-GITR antibodies (e.g., bivalent anti-GITR antibodies) such as, e.g., a GITR fusion protein described in U.S. Pat. No. 6,111,090, European Patent No.: 090505B1, U.S. Pat. No. 8,586,023, PCT Publication Nos.: WO 2010/003118 and 2011/090754, or an anti-GITR antibody described, e.g., in U.S. Pat. No. 7,025,962, European Patent No.: 1947183B1, U.S. Pat. Nos. 7,812,135, 8,388,967, 8,591,886, European Patent No.: EP 1866339, PCT Publication No.: WO 2011/028683, PCT Publication No.: WO 2013/039954, PCT Publication No.: WO2005/007190, PCT Publication No.: WO 2007/133822, PCT Publication No.: WO2005/055808, PCT Publication No.: WO 99/40196, PCT Publication No.: WO 2001/03720, PCT Publication No.: WO99/20758, PCT Publication No.: WO2006/083289, PCT Publication No.: WO 2005/115451, U.S. Pat. No. 7,618,632, and PCT Publication No.: WO 2011/051726.


In one embodiment, a CAR-expressing cell described herein is administered in combination with a kinase inhibitor. Exemplary kinase inhibitors and uses thereof, are described in International Application WO 2016/164731 filed on Apr. 8, 2016, which is incorporated by reference in its entirety.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with an indoleamine 2,3-dioxygenase (IDO) inhibitor. The IDO an emzyme and uses thereof are described on pages 292-293 of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with a modulator of myeloid-derived suppressor cells (MDSCs). MDSCs and compositions that can be used to modulate MDSCs are described on pages 293-294 of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


In embodiments, a CAR-expressing cell described herein is administered to a subject in combination with a CD19 CART cell (e.g., CTL019, e.g., as described in WO2012/079000, incorporated herein by reference). In embodiments, a CD19 CART is administered to the subject prior to, concurrently with, or after administration (e.g., infusion) of a non-CD19 CAR-expressing cell, e.g., a non-CD19 CAR-expressing cell described herein.


Without being bound by theory, it is believed that administering a CD19 CAR-expressing cell in combination with a CAR-expressing cell improves the efficacy of a CAR-expressing cell described herein by targeting early lineage cancer cells, e.g., cancer stem cells, modulating the immune response, depleting regulatory B cells, and/or improving the tumor microenvironment. For example, a CD19 CAR-expressing cell targets cancer cells that express early lineage markers, e.g., cancer stem cells and CD19-expressing cells, while the CAR-expressing cell described herein targets cancer cells that express later lineage markers, e.g., CLL-1. This preconditioning approach can improve the efficacy of the CAR-expressing cell described herein. In such embodiments, the CD19 CAR-expressing cell is administered prior to, concurrently with, or after administration (e.g., infusion) of a CAR-expressing cell described herein.


In embodiments, a CAR-expressing cell described herein also expresses a CAR targeting CD19, e.g., a CD19 CAR. In an embodiment, the cell expressing a CAR described herein and a CD19 CAR is administered to a subject for treatment of a cancer described herein, e.g., ALL. In an embodiment, the configurations of one or both of the CAR molecules comprise a primary intracellular signaling domain and a costimulatory signaling domain. In another embodiment, the configurations of one or both of the CAR molecules comprise a primary intracellular signaling domain and two or more, e.g., 2, 3, 4, or 5 or more, costimulatory signaling domains. In such embodiments, the CAR molecule described herein and the CD19 CAR may have the same or a different primary intracellular signaling domain, the same or different costimulatory signaling domains, or the same number or a different number of costimulatory signaling domains. Alternatively, the CAR described herein and the CD19 CAR are configured as a split CAR, in which one of the CAR molecules comprises an antigen binding domain and a costimulatory domain (e.g., 4-1BB), while the other CAR molecule comprises an antigen binding domain and a primary intracellular signaling domain (e.g., CD3 zeta).


In some embodiments, a CAR-expressing cell described herein is administered to a subject in combination with a interleukin-15 (IL-15) polypeptide, a interleukin-15 receptor alpha (IL-15Ra) polypeptide, or a combination of both a IL-15 polypeptide and a IL-15Ra polypeptide e.g., hetIL-15 (Admune Therapeutics, LLC). hetIL-15 is a heterodimeric non-covalent complex of IL-15 and IL-15Ra. hetIL-15 is described in, e.g., U.S. Pat. No. 8,124,084, U.S. 2012/0177598, U.S. 2009/0082299, U.S. 2012/0141413, and U.S. 2011/0081311, incorporated herein by reference. In embodiments, het-IL-15 is administered subcutaneously. In embodiments, the subject has a cancer, e.g., solid cancer, e.g., melanoma or colon cancer. In embodiments, the subject has a metastatic cancer.


In embodiments, a subject having a disease described herein, e.g., a hematological disorder, e.g., ALL, is administered a CAR-expressing cell described herein in combination with an agent described on pages 296-297 of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


Lymphodepletion

In embodiments, lymphodepletion is performed on a subject, e.g., prior to administering one or more cells that express a CAR described herein. In embodiments, the lymphodepletion regimen comprises administering one or more of melphalan, cytoxan, bendamustine, cyclophosphamide, and fludarabine. In some embodiments, the lymphodepletion regimen is also referred to as a lymphodepleting chemotherapy or a lymphodepleting therapy. In some embodiments, a subject is administered lymphodepleting chemotherapy after administration of bridging chemotherapy, e.g., as described herein. In some embodiments, a subject is administered lymphodepleting chemotherapy without prior administration of bridging chemotherapy.


In embodiments, the lymphodepletion regimen comprises administering cyclophosphamide. In embodiments, cyclophosphamide is administered daily, e.g., for 2 or 3 days, at a dosage of about 200-700 mg/m2 (e.g., 250-650, 300-600, 350-550, 400-500, 200-300, 400-600, or 450-550 mg/m2, e.g., about 250 mg/m2 or 500 mg/m2), e.g., intravenously. In some embodiments, cyclophosphamide is administered at a dosage of about 250 mg/m2 per day, for 3 days. In some embodiments, cyclophosphamide is administered at a dosage of about 500 mg/m2 per day, for 2 days.


In embodiments, the lymphodepletion regimen comprises administering fludarabine. In embodiments, fludarabine is administered daily, e.g., for 3 or 4 days, at a dosage of about 10-50 mg/m2 (e.g., 20-30, 25-40 or 25-35 mg/m2, e.g., about 25 mg/m2 or 30 mg/m2), e.g., intravenously. In some embodiments, fludarabine is administered at a dosage of about 30 mg/m2 per day, for 4 days. In some embodiments, fludarabine is administered at a dosage of about 25 mg/m2 per day, for 3 days.


In embodiments, the lymphodepletion regimen comprises administering cyclophosphamide and fludarabine. In some embodiments, the lymphodepletion comprises administering 500 mg/m2 cyclophosphamide daily for 2 days and 30 mg/m2 fludarabine daily for 4 days. In some embodiments, the lymphodepletion regimen comprises administering 500 mg/m2 cyclophosphamide daily for 2 days, e.g., 2 doses, and 30 mg/m2 fludarabine daily for 4 days, e.g., 4 doses. In some embodiments, the subject has a cancer, e.g., a hematological cancer as described herein. In some embodiments, the hematological cancer is a leukemia or a lymphoma, e.g., a relapsed and/or refractory leukemia or lymphoma. In some embodiments, the leukemia, e.g., ALL, e.g., relapsed and/or refractory ALL. In some embodiments, the subject is a young adult or a pediatric subject and the leukemia is relapsed and/or refractory ALL. In some embodiments, the subject is aged about 1-29 years, e.g., aged 1-29 years, and the leukemia is a relapsed and/or refractory ALL. In some embodiments, the lymphodepletion regimen is initiated with the administration of the first dose of fludarabine. In some embodiments, cyclophosphamide and fludarabine are administered on the same day. In some embodiments, cyclophosphamide and fludarabine are not administered on the same day. In some embodiments, the daily dosages are administered on consecutive days. In embodiments, the subject is administered CAR-expressing cells about 1-14 days, e.g., 2-13, 3-12, 4-11, 5-10, 2-11, 2-6, or 1-4 days, after completion of the lymphodepletion regimen. In some embodiments, the lymphodepletion regimen is administered to the subject about 1 week, e.g., about 6, 5, 4, 3, 2, or 1 days, prior to administration of CAR-expressing cells. In some embodiments, the lymphodepletion regimen is completed, about 1 week, e.g., about 6, 5, 4, 3, 2, or 1 days, prior to administration of CAR-expressing cells.


In some embodiments, when the subject has ALL, e.g., relapsed or refractory ALL, the lymphodepletion regimen comprises administering 500 mg/m2 cyclophosphamide daily for 2 days, e.g., 2 doses, and 30 mg/m2 fludarabine daily for 4 days, e.g., 4 doses, starting with the first dose of fludarabine.


In embodiments, the lymphodepletion regimen comprises administering bendamustine. In some embodiments, bendamustine is administered daily, e.g., for 2 days, at a dosage of about 75-125 mg/m2 (e.g., 75-100 or 100-125 mg/m2, e.g., about 90 mg/m2), e.g., intravenously. In some embodiments, bendamustine is administered at a dosage of 90 mg/m2 daily, e.g., for 2 days. In some embodiments, the subject has a cancer, e.g., a hematological cancer as described herein. In some embodiments, the hematological cancer is a leukemia or a lymphoma. In some embodiments, the leukemia is ALL, e.g., a relapsed/refractory ALL (e.g., r/r ALL), e.g., a CD 19+ r/r ALL. In some embodiments, the subject is a young adult or a pediatric subject and the leukemia is an r/r ALL. In some embodiments, the subject is a young adult or a pediatric subject, e.g., aged about 1-29 years, and the leukemia is a relapsed or refractory ALL. In embodiments, the subject is administered CAR-expressing cells about 1-14 days, e.g., 2-13, 3-12, 4-11, 5-10, 2-11, or 2-6 days, after completion of the lymphodepletion regimen. In some embodiments, the lymphodepletion regimen is administered to the subject about 1 week, e.g., about 10, 9, 8, 7, or 6 days, prior to administration of CAR-expressing cells.


In embodiments, the subject is administered a first lymphodepletion regimen and/or a second lymphodepletion regimen. In embodiments, the first lymphodepletion regimen is administered before the second lymphodepletion regimen. In embodiments, the second lymphodepletion regimen is administered before the first lymphodepletion regimen. In embodiments, the first lymphodepletion regimen comprises cyclophosphamide and fludarabine, e.g., 250 mg/m2 cyclophosphamide daily for 3 days, and 25 mg/m2 fludarabine daily for 3 days. In embodiments, the second lymphodepletion regimen comprises bendamustine, e.g., 90 mg/m2 daily, e.g., for 2 days. In embodiments, the second lymphodepletion regimen is administered as an alternate lymphodepletion regimen. In some embodiments, the second lymphodepletion regimen, e.g., comprising bendamustine, is administered as an alternate lymphodepletion regimen, e.g., if a subject has experienced adverse effects, e.g., hemorrhagic cystitis (e.g., Grade 4 hemorrhagic cystitis), to a lymphodepletion regimen comprising cyclophosphamide, or if a subject shows or has shown resistance to a cyclophoshamide-containing regimen. In some embodiments, the lymphoma is a DLBCL, e.g., a relapsed or refractory DLBCL (e.g., r/r DLBCL), e.g., a CD19+ r/r DLBCL. In some embodiments, the subject is an adult and the lymphoma is an r/r DLBCL. In some embodiments, the lymphoma is a follicular lymphoma (FL), e.g., relapsed or refractory FL. In some embodiments, the subject is an adult, e.g., at least 18 years of age, and the lymphoma is a FL, e.g., relapsed or refractory FL.


In embodiments, the lymphodepletion comprises administering bendamustine (e.g., at about 90 mg/m2, e.g., daily×2), cyclophosphamide and fludarabine (e.g., at about 200 mg/m2 cyclophosphamide and about 20 mg/m2 fludarabine, e.g., daily×3), XRT and cyclophosphamide (e.g., at about 400 cGy XRT and about 1 g/m2 cyclophosphamide), cyclophosphamide (e.g., about 1 g/m2 or 1.2 g/m2 cyclophosphamide, e.g., over 4 days), carboplatin and gemcitabine, or modified EPOCH.


In some embodiments, a subject is not administered a lymphodepletion regimen, e.g., lymphodepleting chemotherapy, if the patient has a white blood cell count (WBC) of less than about 5-0.5×109 cells/L, e.g., about 4-0.4, 3-0.3, 2-0.2 or or 1.5-0.5×109 cells/L, e.g., about 1×109 cells/L. In some embodiments, the WBC count is obtained, e.g., within 1 week, e.g., 6, 5, 4, 3, 2, or 1 days, prior to CAR cell administration, e.g., infusion. In some embodiments, a subject is not administered a lymphodepletion regimen, e.g., as described herein, if the subject has cytopenia, e.g., WBC of <1000 cells/μl, or absolute lymphocyte count (ALC) of <200/0. In some embodiments, the leukemia is an ALL, e.g., a relapsed or refractory ALL (e.g., r/r ALL). In some embodiments, the subject is a young adult or a pediatric subject and the leukemia is relapsed and/or refractory ALL. In some embodiments, the subject is a young adult or a pediatric subject, e.g., aged about 1-29 years, and the leukemia is a relapsed or refractory ALL.


In embodiments, a lymphodepleting chemotherapy is administered to the subject prior to, concurrently with, or after administration (e.g., infusion) of CAR cells, e.g., cells described herein. In an example, the lymphodepleting chemotherapy is administered to the subject prior to administration of CAR cells. For example, the lymphodepleting chemotherapy ends 1˜4 days (e.g., 1, 2, 3, or 4 days) prior to CAR cell infusion. In embodiments, multiple doses of CAR cells are administered, e.g., as described herein, e.g., according to a dose fractionation or split-dosing regimen described herein, e.g., for a total dose of 2.0×106 cells/kg of each CAR molecule expressing cell, e.g., CD19 CAR molecule expressing cell or CD22 CAR molecule expressing cell. In embodiments, a lymphodepleting chemotherapy is administered to the subject prior to, concurrently with, or after administration (e.g., infusion) of a CAR-expressing cell described herein.


In one embodiment, the one or more doses of the cells are administered after one or more lymphodepleting therapies, e.g., a lymphodepleting chemotherapy. In one embodiment, the lymphodepleting therapy includes a chemotherapy (e.g., cyclophosphamide).


In one embodiment, the one or more doses is followed by a cell transplant, e.g., an allogeneic hematopoietic stem cell transplant. For example, the allogeneic hematopoietic stem cell transplant occurs between about 20 to about 35 days, e.g., between about 23 and 33 days.


Inhibitory Molecule Inhibitors/Checkpoint Inhibitors

In one embodiment, the subject can be administered an agent which enhances the activity of a CAR-expressing cell. For example, in one embodiment, the agent can be an agent which inhibits an inhibitory molecule, e.g., the agent is a checkpoint inhibitor. Inhibitory or checkpoint molecules, e.g., Programmed Death 1 (PD1), can, in some embodiments, decrease the ability of a CAR-expressing cell to mount an immune effector response. Examples of inhibitory molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAGS, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, GALS, adenosine, and TGF (e.g., TGFR beta). In embodiments, the CAR-expressing cell described herein comprises a switch costimulatory receptor, e.g., as described in WO 2013/019615, which is incorporated herein by reference in its entirety.


The methods described herein can include administration of a CAR-expressing cell in combination with a checkpoint inhibitor. Methods comprising the administration of CAR-expressing cell in combination with checkpoint inhibitors is disclosed on pages 299-308 of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


Antibodies, antibody fragments, and other inhibitors of PD1, PD-L1 and PD-L2 are available in the art and may be used combination with a CD19 CAR described herein. For example, nivolumab (also referred to as BMS-936558 or MDX1106; Bristol-Myers Squibb) is a fully human IgG4 monoclonal antibody which specifically blocks PD1. Nivolumab (clone 5C4) and other human monoclonal antibodies that specifically bind to PD1 are disclosed in U.S. Pat. No. 8,008,449 and WO2006/121168. Pidilizumab (CT-011; Cure Tech) is a humanized IgG1k monoclonal antibody that binds to PD1. Pidilizumab and other humanized anti-PD1 monoclonal antibodies are disclosed in WO2009/101611. Pembrolizumab (formerly known as lambrolizumab, and also referred to as Keytruda, MK03475; Merck) is a humanized IgG4 monoclonal antibody that binds to PD1. Pembrolizumab and other humanized anti-PD1 antibodies are disclosed in U.S. Pat. No. 8,354,509 and WO2009/114335. MEDI4736 (Medimmune) is a human monoclonal antibody that binds to PDL1, and inhibits interaction of the ligand with PD1. MDPL3280A (Genentech/Roche) is a human Fc optimized IgG1 monoclonal antibody that binds to PD-L1. MDPL3280A and other human monoclonal antibodies to PD-L1 are disclosed in U.S. Pat. No. 7,943,743 and U.S Publication No.: 20120039906. Other anti-PD-L1 binding agents include YW243.55.570 (heavy and light chain variable regions are shown in SEQ ID NOs 20 and 21 in WO2010/077634) and MDX-1 105 (also referred to as BMS-936559, and, e.g., anti-PD-L1 binding agents disclosed in WO2007/005874). AMP-224 (B7-DCIg; Amplimmune; e.g., disclosed in WO2010/027827 and WO2011/066342), is a PD-L2 Fc fusion soluble receptor that blocks the interaction between PD1 and B7-H1. Other anti-PD1 antibodies include AMP 514 (Amplimmune), among others, e.g., anti-PD1 antibodies disclosed in U.S. Pat. No. 8,609,089, US 2010028330, and/or US 20120114649.


In some embodiments, a PD1 inhibitor described herein (e.g., a PD1 antibody, e.g., a PD1 antibody described herein) is used combination with a CD19 CAR described herein to treat a disease associated with expression of CD19. In some embodiments, a PD-L1 inhibitor described herein (e.g., a PD-L1 antibody, e.g., a PD-L1 antibody described herein) is used combination with a CD19 CAR described herein to treat a disease associated with expression of CD19. In some embodiments, the subject has, or is identified as having, at least 5%, 6%, 7%, 8%, 9%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of cancer cells, e.g., which are CD3+/PD1+. In some embodiments, the subject has, or is identified as having, substantially non-overlapping populations of CD19+ cells and PD-L1+ cells in a cancer, e.g., the cancer microenvironment. For instance, in some embodiments, less than 20%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% of cells in the cancer, e.g., cancer microenvironment, are double positive for CD19 and PD-L1.


In some embodiments, the subject is treated with a combination of a CD19 CAR, a PD1 inhibitor, and a PD-L1 inhibitor. In some embodiments, the subject is treated with a combination of a CD19 CAR, a PD1 inhibitor, and a CD3 inhibitor. In some embodiments, the subject is treated with a combination of a CD19 CAR, a PD1 inhibitor, a PD-L1 inhibitor, and a CD3 inhibitor.


In some embodiments, the methods herein include a step of assaying cells in a biological sample, e.g., a sample comprising DLBCL cells, for CD3 and/or PD-1 (e.g., CD3 and/or PD-1 expression). In some embodiments, the methods include a step of assaying cells in a biological sample, e.g., a sample comprising DLBCL cells, for CD19 and/or PD-L1 (e.g., CD19 and/or PD-L1 expression). In some embodiments, the methods include, e.g., providing a sample comprising cancer cells and performing a detection step, e.g., by immunohistochemistry, for one or more of CD3, PD-1, CD19, or PD-L1. The methods may comprise a further step of recommending or administering a treatment, e.g., a treatment comprising a CD19 CAR.


In one embodiment, the anti-PD-1 antibody or fragment thereof is an anti-PD-1 antibody molecule as described in US 2015/0210769, entitled “Antibody Molecules to PD-1 and Uses Thereof,” incorporated by reference in its entirety. In one embodiment, the anti-PD-1 antibody molecule includes at least one, two, three, four, five or six CDRs (or collectively all of the CDRs) from a heavy and light chain variable region from an antibody chosen from any of BAP049-hum01, BAP049-hum02, BAP049-hum03, BAP049-hum04, BAP049-hum05, BAP049-hum06, BAP049-hum07, BAP049-hum08, BAP049-hum09, BAP049-hum10, BAP049-hum11, BAP049-hum12, BAP049-hum13, BAP049-hum14, BAP049-hum15, BAP049-hum16, BAP049-Clone-A, BAP049-Clone-B, BAP049-Clone-C, BAP049-Clone-D, or BAP049-Clone-E; or as described in Table 1 of US 2015/0210769, or encoded by the nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or closely related CDRs, e.g., CDRs which are identical or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions).


In yet another embodiment, the anti-PD-1 antibody molecule comprises at least one, two, three or four variable regions from an antibody described herein, e.g., an antibody chosen from any of BAP049-hum01, BAP049-hum02, BAP049-hum03, BAP049-hum04, BAP049-hum05, BAP049-hum06, BAP049-hum07, BAP049-hum08, BAP049-hum09, BAP049-hum10, BAP049-hum11, BAP049-hum12, BAP049-hum13, BAP049-hum14, BAP049-hum15, BAP049-hum16, BAP049-Clone-A, BAP049-Clone-B, BAP049-Clone-C, BAP049-Clone-D, or BAP049-Clone-E; or as described in Table 1 of US 2015/0210769, or encoded by the nucleotide sequence in Table 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.


TIM3 (T cell immunoglobulin-3) also negatively regulates T cell function, particularly in IFN-g-secreting CD4+ T helper 1 and CD8+ T cytotoxic 1 cells, and plays a critical role in T cell exhaustion. Inhibition of the interaction between TIM3 and its ligands, e.g., galectin-9 (Gal9), phosphatidylserine (PS), and HMGB1, can increase immune response. Antibodies, antibody fragments, and other inhibitors of TIM3 and its ligands are available in the art and may be used combination with a CD19 CAR described herein. For example, antibodies, antibody fragments, small molecules, or peptide inhibitors that target TIM3 binds to the IgV domain of TIM3 to inhibit interaction with its ligands. Antibodies and peptides that inhibit TIM3 are disclosed in WO2013/006490 and US20100247521. Other anti-TIM3 antibodies include humanized versions of RMT3-23 (disclosed in Ngiow et al., 2011, Cancer Res, 71:3540-3551), and clone 8B.2C12 (disclosed in Monney et al., 2002, Nature, 415:536-541). Bi-specific antibodies that inhibit TIM3 and PD-1 are disclosed in US20130156774.


In one embodiment, the anti-TIM3 antibody or fragment thereof is an anti-TIM3 antibody molecule as described in US 2015/0218274, entitled “Antibody Molecules to TIM3 and Uses Thereof,” incorporated by reference in its entirety. In one embodiment, the anti-TIM3 antibody molecule includes at least one, two, three, four, five or six CDRs (or collectively all of the CDRs) from a heavy and light chain variable region from an antibody chosen from any of ABTIM3, ABTIM3-hum01, ABTIM3-hum02, ABTIM3-hum03, ABTIM3-hum04, ABTIM3-hum05, ABTIM3-hum06, ABTIM3-hum07, ABTIM3-hum08, ABTIM3-hum09, ABTIM3-hum10, ABTIM3-hum11, ABTIM3-hum12, ABTIM3-hum13, ABTIM3-hum14, ABTIM3-hum15, ABTIM3-hum16, ABTIM3-hum17, ABTIM3-hum18, ABTIM3-hum19, ABTIM3-hum20, ABTIM3-hum21, ABTIM3-hum22, ABTIM3-hum23; or as described in Tables 1-4 of US 2015/0218274; or encoded by the nucleotide sequence in Tables 1-4; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences, or closely related CDRs, e.g., CDRs which are identical or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions).


In yet another embodiment, the anti-TIM3 antibody molecule comprises at least one, two, three or four variable regions from an antibody described herein, e.g., an antibody chosen from any of ABTIM3, ABTIM3-hum01, ABTIM3-hum02, ABTIM3-hum03, ABTIM3-hum04, ABTIM3-hum05, ABTIM3-hum06, ABTIM3-hum07, ABTIM3-hum08, ABTIM3-hum09, ABTIM3-hum10, ABTIM3-hum11, ABTIM3-hum12, ABTIM3-hum13, ABTIM3-hum14, ABTIM3-hum15, ABTIM3-hum16, ABTIM3-hum17, ABTIM3-hum18, ABTIM3-hum19, ABTIM3-hum20, ABTIM3-hum21, ABTIM3-hum22, ABTIM3-hum23; or as described in Tables 1-4 of US 2015/0218274; or encoded by the nucleotide sequence in Tables 1-4; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.


In other embodiments, the agent which enhances the activity of a CAR-expressing cell is a CEACAM inhibitor (e.g., CEACAM-1, CEACAM-3, and/or CEACAM-5 inhibitor). In one embodiment, the inhibitor of CEACAM is an anti-CEACAM antibody molecule. Exemplary anti-CEACAM-1 antibodies are described in WO 2010/125571, WO 2013/082366 WO 2014/059251 and WO 2014/022332, e.g., a monoclonal antibody 34B1, 26H7, and 5F4; or a recombinant form thereof, as described in, e.g., US 2004/0047858, U.S. Pat. No. 7,132,255 and WO 99/052552. In other embodiments, the anti-CEACAM antibody binds to CEACAM-5 as described in, e.g., Zheng et al. PLoS One. 2010 Sep. 2; 5(9). pii: e12529 (DOI:10:1371/journal.pone.0021146), or crossreacts with CEACAM-1 and CEACAM-5 as described in, e.g., WO 2013/054331 and US 2014/0271618.


Without wishing to be bound by theory, carcinoembryonic antigen cell adhesion molecules (CEACAM), such as CEACAM-1 and CEACAM-5, are believed to mediate, at least in part, inhibition of an anti-tumor immune response (see e.g., Markel et al. J Immunol. 2002 Mar. 15; 168(6):2803-10; Markel et al. J Immunol. 2006 Nov. 1; 177(9):6062-71; Markel et al. Immunology. 2009 February; 126(2):186-200; Markel et al. Cancer Immunol Immunother. 2010 February; 59(2):215-30; Ortenberg et al. Mol Cancer Ther. 2012 June; 11(6):1300-10; Stern et al. J Immunol. 2005 Jun. 1; 174(11):6692-701; Zheng et al. PLoS One. 2010 Sep. 2; 5(9). pii: e12529). For example, CEACAM-1 has been described as a heterophilic ligand for TIM-3 and as playing a role in TIM-3-mediated T cell tolerance and exhaustion (see e.g., WO 2014/022332; Huang, et al. (2014) Nature doi:10.1038/nature13848). In embodiments, co-blockade of CEACAM-1 and TIM-3 has been shown to enhance an anti-tumor immune response in xenograft colorectal cancer models (see e.g., WO 2014/022332; Huang, et al. (2014), supra). In other embodiments, co-blockade of CEACAM-1 and PD-1 reduce T cell tolerance as described, e.g., in WO 2014/059251. Thus, CEACAM inhibitors can be used with the other immunomodulators described herein (e.g., anti-PD-1 and/or anti-TIM-3 inhibitors) to enhance an immune response against a cancer, e.g., a melanoma, a lung cancer (e.g., NSCLC), a bladder cancer, a colon cancer, an ovarian cancer, and other cancers as described herein.


LAG3 (lymphocyte activation gene-3 or CD223) is a cell surface molecule expressed on activated T cells and B cells that has been shown to play a role in CD8+ T cell exhaustion. Antibodies, antibody fragments, and other inhibitors of LAG3 and its ligands are available in the art and may be used combination with a CD19 CAR described herein. For example, BMS-986016 (Bristol-Myers Squib) is a monoclonal antibody that targets LAG3. IMP701 (Immutep) is an antagonist LAG3 antibody and 1MP731 (Immutep and GlaxoSmithKline) is a depleting LAG3 antibody. Other LAG3 inhibitors include IMP321 (Immutep), which is a recombinant fusion protein of a soluble portion of LAG3 and Ig that binds to MHC class II molecules and activates antigen presenting cells (APC). Other antibodies are disclosed, e.g., in WO2010/019570.


In one embodiment, the anti-LAG3 antibody or fragment thereof is an anti-LAG3 antibody molecule as described in US 2015/0259420, entitled “Antibody Molecules to LAG3 and Uses Thereof,” incorporated by reference in its entirety. In one embodiment, the anti-LAG3 antibody molecule includes at least one, two, three, four, five or six CDRs (or collectively all of the CDRs) from a heavy and light chain variable region from an antibody chosen from any of BAP050-hum01, BAP050-hum02, BAP050-hum03, BAP050-hum04, BAP050-hum05, BAP050-hum06, BAP050-hum07, BAP050-hum08, BAP050-hum09, BAP050-hum10, BAP050-hum11, BAP050-hum12, BAP050-hum13, BAP050-hum14, BAP050-hum15, BAP050-hum16, BAP050-hum17, BAP050-hum18, BAP050-hum19, BAP050-hum20, huBAP050(Ser) (e.g., BAP050-hum01-Ser, BAP050-hum02-Ser, BAP050-hum03-Ser, BAP050-hum04-Ser, BAP050-hum05-Ser, BAP050-hum06-Ser, BAP050-hum07-Ser, BAP050-hum08-Ser, BAP050-hum09-Ser, BAP050-hum10-Ser, BAP050-hum11-Ser, BAP050-hum12-Ser, BAP050-hum13-Ser, BAP050-hum14-Ser, BAP050-hum15-Ser, BAP050-hum18-Ser, BAP050-hum19-Ser, or BAP050-hum20-Ser), BAP050-Clone-F, BAP050-Clone-G, BAP050-Clone-H, BAP050-Clone-I, or BAP050-Clone-J; or as described in Table 1 of US 2015/0259420; or encoded by the nucleotide sequence in Table 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences, or closely related CDRs, e.g., CDRs which are identical or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions).


In yet another embodiment, the anti-LAG3 antibody molecule comprises at least one, two, three or four variable regions from an antibody described herein, e.g., an antibody chosen from any of BAP050-hum01, BAP050-hum02, BAP050-hum03, BAP050-hum04, BAP050-hum05, BAP050-hum06, BAP050-hum07, BAP050-hum08, BAP050-hum09, BAP050-hum10, BAP050-hum11, BAP050-hum12, BAP050-hum13, BAP050-hum14, BAP050-hum15, BAP050-hum16, BAP050-hum17, BAP050-hum18, BAP050-hum19, BAP050-hum20, huBAP050(Ser) (e.g., BAP050-hum01-Ser, BAP050-hum02-Ser, BAP050-hum03-Ser, BAP050-hum04-Ser, BAP050-hum05-Ser, BAP050-hum06-Ser, BAP050-hum07-Ser, BAP050-hum08-Ser, BAP050-hum09-Ser, BAP050-hum10-Ser, BAP050-hum11-Ser, BAP050-hum12-Ser, BAP050-hum13-Ser, BAP050-hum14-Ser, BAP050-hum15-Ser, BAP050-hum18-Ser, BAP050-hum19-Ser, or BAP050-hum20-Ser), BAP050-Clone-F, BAP050-Clone-G, BAP050-Clone-H, BAP050-Clone-I, or BAP050-Clone-J; or as described in Table 1 of US 2015/0259420; or encoded by the nucleotide sequence in Tables 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.


In some embodiments, the agent which enhances the activity of a CAR-expressing cell can be, e.g., a fusion protein comprising a first domain and a second domain, wherein the first domain is an inhibitory molecule, or fragment thereof, and the second domain is a polypeptide that is associated with a positive signal, e.g., a polypeptide comprising an intracellular signaling domain as described herein. In some embodiments, the polypeptide that is associated with a positive signal can include a costimulatory domain of CD28, CD27, ICOS, e.g., an intracellular signaling domain of CD28, CD27 and/or ICOS, and/or a primary signaling domain, e.g., of CD3 zeta, e.g., described herein. In one embodiment, the fusion protein is expressed by the same cell that expressed the CAR. In another embodiment, the fusion protein is expressed by a cell, e.g., a T cell that does not express an anti-CD19 CAR.


In one embodiment, the agent which enhances activity of a CAR-expressing cell described herein is miR-17-92.


In one embodiment, the agent which enhances activity of a CAR-described herein is a cytokine. Cytokines have important functions related to T cell expansion, differentiation, survival, and homeostasis. Cytokines that can be administered to the subject receiving a CAR-expressing cell described herein include: IL-2, IL-4, IL-7, IL-9, IL-15, IL-18, and IL-21, or a combination thereof. In embodiments, the cytokine administered is IL-7, IL-15, or IL-21, or a combination thereof. The cytokine can be administered once a day or more than once a day, e.g., twice a day, three times a day, or four times a day. The cytokine can be administered for more than one day, e.g. the cytokine is administered for 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, or 4 weeks. For example, the cytokine is administered once a day for 7 days.


In embodiments, the cytokine is administered in combination with CAR-expressing cells. The cytokine can be administered simultaneously or concurrently with the CAR-expressing cells, e.g., administered on the same day. The cytokine may be prepared in the same pharmaceutical composition as the CAR-expressing cells, or may be prepared in a separate pharmaceutical composition. Alternatively, the cytokine can be administered shortly after administration of the CAR-expressing T cells, e.g., 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or 7 days after administration of the CAR-expressing cells. In embodiments where the cytokine is administered in a dosing regimen that occurs over more than one day, the first day of the cytokine dosing regimen can be on the same day as administration with the CAR-expressing cells, or the first day of the cytokine dosing regimen can be 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or 7 days after administration of the CAR-expressing T cells. In one embodiment, on the first day, the CAR-expressing cells are administered to the subject, and on the second day, a cytokine is administered once a day for the next 7 days. In an embodiment, the cytokine to be administered in combination with the CAR-expressing cells is IL-7, IL-15, and/or IL-21.


In other embodiments, the cytokine is administered a sufficient period of time after administration of the CAR-expressing cells, e.g., at least 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 10 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 1 year or more after administration of CAR-expressing cells. In one embodiment, the cytokine is administered after assessment of the subject's response to the CAR-expressing cells. For example, the subject is administered CAR-expressing cells according to the dosage and regimens described herein. The response of the subject to CART therapy is assessed at 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 10 weeks, 12 weeks, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, or 1 year or more after administration of CAR-expressing cells, using any of the methods described herein, including inhibition of tumor growth, reduction of circulating tumor cells, or tumor regression. Subjects that do not exhibit a sufficient response to CART therapy can be administered a cytokine. Administration of the cytokine to the subject that has sub-optimal response to the CART therapy improves CART efficacy and/or anti-tumor activity. In an embodiment, the cytokine administered after administration of CAR-expressing cells is IL-7.


Further combination therapies may include anti-allergenic agents, anti-emetics, analgesics, adjunct therapies,


The above-mentioned compounds, which can be used in combination with a compound of the present invention, can be prepared and administered as described in the art, such as in the documents cited above.


In one embodiment, the present invention provides pharmaceutical compositions comprising at least one compound of the present invention (e.g., a compound of the present invention) or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier suitable for administration to a human or animal subject, either alone or together with other anti-cancer agents.


In one embodiment, the present invention provides methods of treating human or animal subjects suffering from a cellular proliferative disease, such as cancer. The present invention provides methods of treating a human or animal subject in need of such treatment, comprising administering to the subject a therapeutically effective amount of a compound of the present invention (e.g., a compound of the present invention) or a pharmaceutically acceptable salt thereof, either alone or in combination with other anti-cancer agents.


In particular, compositions will either be formulated together as a combination therapeutic or administered separately.


In combination therapy, the compound of the present invention and other anti-cancer agent(s) may be administered either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient.


In a preferred embodiment, the compound of the present invention and the other anti-cancer agent(s) is generally administered sequentially in any order by infusion or orally. The dosing regimen may vary depending upon the stage of the disease, physical fitness of the patient, safety profiles of the individual drugs, and tolerance of the individual drugs, as well as other criteria well-known to the attending physician and medical practitioner(s) administering the combination. The compound of the present invention and other anti-cancer agent(s) may be administered within minutes of each other, hours, days, or even weeks apart depending upon the particular cycle being used for treatment. In addition, the cycle could include administration of one drug more often than the other during the treatment cycle and at different doses per administration of the drug.


In another aspect of the present invention, kits that include one or more compound of the present invention and a combination partner as disclosed herein are provided. Representative kits include (a) a compound of the present invention or a pharmaceutically acceptable salt thereof, (b) at least one combination partner, e.g., as indicated above, whereby such kit may comprise a package insert or other labeling including directions for administration.


A compound of the present invention may also be used to advantage in combination with known therapeutic processes, for example, the administration of hormones or especially radiation. A compound of the present invention may in particular be used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.


Combination with a Low, Immune-Enhancing Dose of an mTOR Inhibitor


In one embodiment, the cells expressing a CAR molecule, e.g., a CAR molecule described herein, are administered in combination with a low, immune enhancing dose of an mTOR inhibitor. Low, immune enhancing doses of MTOR inhibitor and uses thereof are described in International Application WO 2016/164731 filed on Apr. 8, 2016, which is incorporated by reference in its entirety.


Methods and Biomarkers for Evaluating CAR-Effectiveness or Sample Suitability

The present disclosure provides, among other things, gene signatures that indicate whether a cancer patient treated with a CAR therapy is likely to relapse, or has relapsed. Methods and Biomarkers for Evaluating CAR-Effectiveness or Sample Suitability are described in the section titled “Methods and Biomarkers for Evaluating CAR-Effectiveness or Sample Suitability” on pages 320-328 of International Application WO 2016/164731 filed on 8 Apr. 2016, which is hereby incorporated by reference.


Personalized Medicine (Theranostics)

CD19 Characteristics, e.g. Mutations


Without wishing to be bound by theory, some cancer patients show an initial response to a CD19 inhibitor such as a CD19 CAR-expressing cell, and then relapse. In some embodiments, the relapse is caused (at least in part) by a frameshift and/or premature stop codon in CD19 in the cancer cells, or other change in the expression (including expression levels) of CD19 which reduces the ability of a CD19 CAR-expressing cell to target the cancer cells. Such a mutation can reduce the effectiveness of the CD19 therapy and contribute to the patient's relapse. Accordingly, in some embodiments, it can be beneficial when a CD19 therapy is supplemented or replaced with a therapy directed to a second, different target, e.g., a target expressed in B-cells, e.g., CD22. Various exemplary combination therapies of this type are disclosed herein.


This application discloses, among other things, methods for treating a subject having cancer comprising one or more of: (1) determining if a subject has a difference, e.g., statistically significant difference, in a characteristic of CD19 relative to a reference characteristic, and (2) if there is a difference between the determined characteristic and reference characteristic, administering to the subject a therapeutically effective dose of a CAR therapy, e.g., CART, thereby treating the subject. The patient may be, e.g., a patient who has relapsed after treatment with a CD19 inhibitor, e.g., a CD19 CAR expressing cell. The patient may be a patient who has received or is receiving a CD19 CAR therapy and is at risk of relapse. The patient may be a non-responder to a CD19 CAR therapy.


The characteristic can be, e.g., a CD19 sequence, e.g., protein or nucleic acid sequence. The sequence can be determined, e.g., as described in the Examples, by high throughput nucleic acid sequencing, or by mass spectrometry of proteins. As described in the Example herein, a patient may relapse after CD19 CART therapy because of mutations in CD19, e.g., in exon 2 of CD19, e.g., a mutation that causes a frameshift and a premature stop codon in CD19. In embodiments, the insertion or deletion does not cause one or both of a frameshift and a premature stop codon. The mutation may be, e.g., an insertion, a deletion, a substitution, a translocation, or a combination of any of the foregoing. The insertion, deletion, or substitution may involve, e.g., at least 1, 2, 3, 4, 5, 10, 15, 20, 20, or 50 nucleotides. The insertion, deletion, or substitution may involve, e.g., at most 2, 3, 4, 5, 10, 15, 20, 20, 50, or 100 nucleotides. In some cases, a population of cells will comprise more than one mutation. In such cases, the mutations can be in overlapping or non-overlapping sub-populations of cells.


In some cases a patient is identified as having a CD19 characteristic that reduces CD19's ability to engage with a CD19 inhibitor such as a CD19 CAR expressing cell. Such a characteristic may be, e.g., a frameshift mutation, a premature stop codon, an alteration in nucleic acid sequence or an alteration in the structure of the primary mRNA transcript. The characteristic may be, e.g., a departure from normal production of CD19 that occurs earlier than splicing. The characteristic may be, e.g., a characteristic other than exon skipping. Such patients may be treated with an inhibitor of another target, e.g., a B-cell inhibitor, for example a CAR expressing cell directed against another epitope, e.g., an epitope of CD22.


In some cases, a patient is identified as having a CD19 characteristic that reduces CD19's ability to engage with a CD19 inhibitor, such as a CD19 CAR expressing cell, but does not reduce or abrogate CD19's ability to engage with a second CD19 inhibitor, such as a CD19 inhibitor that binds to a different region on CD19. Such a characteristic may be, e.g., a mutation that does not cause one or both of a frameshift mutation or a premature stop codon. Such a characteristic may be, e.g., an alteration in nucleic acid sequence or an alteration in the structure of the primary mRNA transcript, a departure from normal production of CD19 that occurs earlier than splicing, or a characteristic other than exon skipping. Such patients may be treated with an inhibitor of CD19, e.g., a B-cell inhibitor directed against an intact region of CD19, e.g., a wild-type portion of CD19. For instance, if a mutation is present in exon 2, the second CD19 inhibitor may bind to an exon other than exon 2, or a part of exon 2 that lacks the mutation. The second CD19 inhibitor may be, e.g., a CD19 inhibitor described herein.


TEFF and TREG Signatures


Methods herein can include steps of determining a TREG signature or determining the levels of TEFF cells or TREG cells, e.g., in a patient or in a population of cells e.g., immune cells. Methods of reducing the level of TREG cells, or decreasing a TREG signature, in a patient or in a population of cells is described in International Application WO 2016/164731 filed on Apr. 8, 2016, which is incorporated by reference in its entirety.


Pharmaceutical Compositions, Dosage and Treatments

Pharmaceutical compositions of the present invention may comprise, in some aspects, a CAR-expressing cell, e.g., a plurality of CAR-expressing cells, as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. Such compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives. Compositions of the present invention are in one aspect formulated for intravenous administration.


Pharmaceutical compositions of the present invention may be administered in a manner appropriate to the disease to be treated (or prevented). The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.


In one embodiment, the pharmaceutical composition is substantially free of, e.g., there are no detectable levels of a contaminant, e.g., selected from the group consisting of endotoxin, mycoplasma, replication competent lentivirus (RCL), p24, VSV-G nucleic acid, HIV gag, residual anti-CD3/anti-CD28 coated beads, mouse antibodies, pooled human serum, bovine serum albumin, bovine serum, culture media components, vector packaging cell or plasmid components, a bacterium and a fungus. In one embodiment, the bacterium is at least one selected from the group consisting of Alcaligenes faecalis, Candida albicans, Escherichia coli, Haemophilus influenza, Neisseria meningitides, Pseudomonas aeruginosa, Staphylococcus aureus, Streptococcus pneumonia, and Streptococcus pyogenes group A.


When “an immunologically effective amount,” “an anti-tumor effective amount,” “a tumor-inhibiting effective amount,” or “therapeutic amount” is indicated, the precise amount of the compositions of the present invention to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient (subject). In some embodiments, a pharmaceutical composition comprising the cells, e.g., T cells described herein may be administered at a dosage of 104 to 109 cells/kg body weight, in some instances 105 to 106 cells/kg body weight, including all integer values within those ranges. In some embodiments, the cells, e.g., T cells described herein may be administered at 3×104, 1×106, 3×106, or 1×107 cells/kg body weight. The 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).


In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 1×105, 2×105, 5×105, 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises at least about 1×105, 2×105, 5×105, 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises up to about 1×105, 2×105, 5×105, 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.1×106-1.8×107 cells/kg, about 0.1×106 to 3.0×106 cells/kg, about 0.5×106 to about 2.5×106 cells/kg, about 8×105-3.0×106 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5×106 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.2×106 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.6×106 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 1.2×106 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 2.0×106 cells/kg. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises at least about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises up to about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose includes a total dose. In some embodiment, a dose include a partial dose, e.g., a partial dose of a total dose, e.g., as administered according to a dosing regimen described herein, e.g., a dose fractionation or split-dosing regimen.


In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 1×105, 2×105, 5×105, 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises at least about 1×105, 2×105, 5×105, 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises up to about 1×105, 2×105, 5×105, 1×106, 1.1×106, 2×106, 3.6×106, 5×106, 1×107, 1.8×107, 2×107, 5×107, 1×108, 2×108, or 5×108 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.1×106-1.8×107 cells, about 0.1×106 to 3.0×106 cells, about 0.5×106 to about 2.5×106 cells, about 8×105-3.0×106 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5×106 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.2×106 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 0.6×106 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 1.2×106 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 2.0×106 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises at least about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose of CAR cells (e.g., CD19, or CD22 CAR cells) comprises up to about 1×107, 2×107, 5×107, 1×108, 2×108, 5×108, 1×109, 2×109, or 5×109 cells. In some embodiments, a dose includes a total dose. In some embodiment, a dose include a partial dose, e.g., a partial dose of a total dose, e.g., as administered according to a dosing regimen described herein, e.g., a dose fractionation or split-dosing regimen.


A dosing regimen can include dose fractionation, e.g., where a certain percentage of the total dose of cells is delivered on a first day of treatment, a different percentage of the total dose of cells is delivered on a subsequent day of treatment, and a different percentage of the total dose of cells is delivered on a yet subsequent day of treatment. For example, 10% of the total dose of cells is delivered on the first day, 30% of the total dose of cells is delivered on the second day, and the remaining 60% of the total dose of cells is delivered on the third day of treatment. For example, a total cell dose includes 1 to 5×106 cells/kg, 5 to 10×106 cells/kg, 1 to 5×107, cells/kg, 5 to 10×107 cells/kg or 1 to 5×108 cells of CAR expressing cells, e.g., CD19 CAR expressing cells or CD22 CAR expressing cells.


In some embodiments, a CAR-expressing cell, e.g., a CD19 CAR-expressing cell described herein or a CD22 CAR-expressing cell described herein, is administered to the subject according to a dosing regimen comprising a total dose of cells administered to the subject by dose fractionation (e.g., split dosing), e.g., one, two, three or more separate administration of a partial dose. In embodiments, a first percentage of the total dose is administered on a first day of treatment, a second percentage of the total dose is administered on a subsequent (e.g., second, third, fourth, fifth, sixth, or seventh or later) day of treatment, and a third percentage (e.g., the remaining percentage) of the total dose is administered on a yet subsequent (e.g., third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, or later) day of treatment. In an embodiment of a dose fractionation regimen (e.g., split-dosing regimen) disclosed herein, about 10% of the total dose of cells is delivered on the first day, about 30% of the total dose of cells is delivered on the second day (e.g., second consecutive day), and the remaining about 60% of the total dose of cells is delivered on the third day of treatment, e.g., third consecutive day of treatment. In an embodiment, a total cell dose (e.g., administered according to a dosing regimen disclosed herein, e.g., dose fractionation, e.g., split-dosing) comprises about 1-5×106 cells/kg, e.g., about 1-5×106 cells/kg, 1.5-4×106 cells/kg, 1.8-3.5×106 cells/kg, or about 1×106 cells/kg, 1.5×106 cells/kg, 2×106 cells/kg, 3×106 cells/kg, 4×106 cells/kg, or 5×106 cells/kg, e.g., about 2.0×106 cells/kg. In one embodiment, the total cell dose is about 2.0×106 cells/kg, e.g., 2.0×106 cells/kg of a CAR expressing cell, e.g., a CD19 CAR expressing cell or a CD22 CAR expressing cell.


In certain aspects, it may be desired to administer activated cells, e.g., T cells or NK cells, to a subject and then subsequently redraw blood (or have an apheresis performed), activate the cells therefrom according to the present invention, and reinfuse the patient with these activated and expanded cells. This process can be carried out multiple times every few weeks. In certain aspects, cells, e.g., T cells or NK cells, can be activated from blood draws of from 10 cc to 400 cc. In certain aspects, cells, e.g., T cells or NK 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.


The administration of the subject compositions 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 trans arterially, subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally. In one aspect, the cell compositions, e.g., T cell or NK cell compositions, of the present invention are administered to a patient by intradermal or subcutaneous injection. In one aspect, the cell compositions e.g., T cell or NK cell compositions, of the present invention are administered by i.v. injection. The compositions of cells e.g., T cell or NK cell compositions, may be injected directly into a tumor, lymph node, or site of infection.


In a particular exemplary aspect, subjects may undergo leukapheresis, wherein leukocytes are collected, enriched, or depleted ex vivo to select and/or isolate the cells of interest, e.g., T cells. These cell isolates, e.g., T cell or NK cell isolates, may be expanded by methods known in the art and treated such that one or more CAR constructs of the invention may be introduced, thereby creating a CAR-expressing cell, e.g., CAR T cell of the invention. Subjects in need thereof may subsequently undergo standard treatment with high dose chemotherapy followed by peripheral blood stem cell transplantation. In certain aspects, following or concurrent with the transplant, subjects receive an infusion of the expanded CAR-expressing cells of the present invention. In an additional aspect, expanded cells are administered before or following surgery.


The dosage of the above treatments to be administered to a patient will vary with the precise nature of the condition being treated and the recipient of the treatment. The scaling of dosages for human administration can be performed according to art-accepted practices. The dose for a therapeutic, e.g., an antibody, e.g., CAMPATH, for example, may be, e.g., in the range 1 to about 100 mg for an adult patient, e.g., administered daily for a period between 1 and 30 days. A suitable daily dose is 1 to 10 mg per day although in some instances larger doses of up to 40 mg per day may be used (described in U.S. Pat. No. 6,120,766).


In one embodiment, the CAR is introduced into cells, e.g., T cells or NK cells, e.g., using in vitro transcription, and the subject (e.g., human) receives an initial administration of CAR-expressing cells, e.g., CAR T cells of the invention, and one or more subsequent administrations of the CAR-expressing cells, e.g., CAR T cells of the invention, wherein the one or more subsequent administrations are administered less than 15 days, e.g., 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 days after the previous administration. In one embodiment, more than one administration of the CAR-expressing cells, e.g., CAR T cells of the invention are administered to the subject (e.g., human) per week, e.g., 2, 3, or 4 administrations of the CAR-expressing cells, e.g., CAR T cells of the invention are administered per week. In one embodiment, the subject (e.g., human subject) receives more than one administration of the CAR-expressing cells, e.g., CAR T cells per week (e.g., 2, 3 or 4 administrations per week) (also referred to herein as a cycle), followed by a week of no CAR-expressing cells, e.g., CAR T cells administrations, and then one or more additional administration of the CAR-expressing cells, e.g., CAR T cells (e.g., more than one administration of the CAR-expressing cells, e.g., CAR T cells per week) is administered to the subject. In another embodiment, the subject (e.g., human subject) receives more than one cycle of CAR-expressing cells, e.g., CAR T cells, and the time between each cycle is less than 10, 9, 8, 7, 6, 5, 4, or 3 days. In one embodiment, the CAR-expressing cells, e.g., CAR T cells are administered every other day for 3 administrations per week. In one embodiment, the CAR-expressing cells, e.g., CAR T cells of the invention are administered for at least two, three, four, five, six, seven, eight or more weeks.


In some embodiments, subjects may be adult subjects (i.e., 18 years of age and older). In certain embodiments, subjects may be between 1 and 30 years of age. In some embodiments, the subjects are 16 years of age or older. In certain embodiments, the subjects are between 16 and 30 years of age.


In some embodiments, the subjects may be a young adult or a pediatric subject (e.g., aged about 1 and 29 years of age, e.g., aged 1-5, 5-10, 10-15, 15-20, 20-25, or 25-29 years).


In one aspect, CAR-expressing cells, e.g., CARTs are generated using lentiviral viral vectors, such as lentivirus. CAR-expressing cells, e.g., CARTs generated that way will have stable CAR expression.


In one aspect, CAR-expressing cells, e.g., CARTs, are generated using a viral vector such as a gammaretroviral vector, e.g., a gammaretroviral vector described herein. CARTs generated using these vectors can have stable CAR expression.


In one aspect, CAR-expressing cells, e.g., CARTs transiently express CAR vectors for 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 days after transduction. Transient expression of CARs can be effected by RNA CAR vector delivery. In one aspect, the CAR RNA is transduced into the cell, e.g., NK cell or T cell, by electroporation.


A potential issue that can arise in patients being treated using transiently expressing CAR T cells (particularly with murine scFv bearing CARTs) is anaphylaxis after multiple treatments.


Without being bound by this theory, it is believed that such an anaphylactic response might be caused by a patient developing humoral anti-CAR response, i.e., anti-CAR antibodies having an anti-IgE isotype. It is thought that a patient's antibody producing cells undergo a class switch from IgG isotype (that does not cause anaphylaxis) to IgE isotype when there is a ten to fourteen day break in exposure to antigen.


If a patient is at high risk of generating an anti-CAR antibody response during the course of transient CAR therapy (such as those generated by RNA transductions), CART infusion breaks should not last more than ten to fourteen days.


EXAMPLES

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


Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the following illustrative examples, make and utilize the compounds of the present invention and practice the claimed methods. The following working examples specifically point out various aspects of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure.


Example 1: Efficacy of Human CART22 Cells in an ALL Animal Model

This example demonstrates the efficacy of the human CART22 cell in an immunodeficient NOD/SCID/IL2rcustom-character−/− (NSG) leukemia mouse model. Leukemia was established in the NSG mice via intravenous (i.v.) injection of 1×106 ALL tumor cells (NALM-6 luciferase positive cells). Baseline tumor engraftment was measured 6 days later by bioluminescence imaging (BLI). Mice then received either untransduced T cells (UTD) or transduced CART22 (75% CAR expression) ranging from 1.25-5×106 total cells/mouse.


Mice were then monitored for tumor burden, blood T cell expansion, and survival. A CART22 dose-dependent, anti-leukemic effect was observed. Tumor burden by BLI was found to inversely correlate with the CART22 dose. Mice receiving 5×106 CART22 cells showed better tumor control than mice that received fewer CART22 cells. CART22 treated mice show a statistically significant better overall survival (OS) in comparison to mice that received UTD cells (p=0.0027). Higher CART22 doses also correlated with higher levels of T cell expansion.


CART22 was compared to CART19 using the NSG mice engrafted with human primary B-ALL blasts, JH331, collected from bone marrow of a relapsing B-ALL patient and containing a click beetle green (CBG) luciferase marker. By flow cytometry, the JH331 B-ALL blasts have both CD22 and CD19 surface expression (FIG. 1A). Leukemia bearing NSG mice received either 5×106 untransduced total T cells (UTD), CART19 or CART22. Transduced CART19 and CART22 cells had 75% CAR expression. Mice were then monitored for tumor burden, blood T cell expansion, and survival (FIG. 1B). Mice receiving CART22 or CART19 showed elimination of the tumor while mice receiving control UTD demonstrated rapid tumor progression. This data shows that CART22 or CART19 are efficacious in preventing tumor growth and disease progression.


A fully human anti-CD22 scFv with a short linker (nicknamed 65s) was then developed and used for testing in the proposed clinical trial described in Example 2. First the in vivo function of CART22-65s was tested. Briefly, one million Nalm6 leukemia cells were injected IV into NSG mice, followed 1 week later by 1×106 T cells that were transduced to express CAR19 (C2137 CTL119), CAR22 with an anti-CD22 antigen binding domain from m971 with various hinge regions (C2270, C3034 or C3042), CART22-65 with longer linkers (C3041 or C7003) or CART22-65s (C7002), as depicted in FIG. 2. Tumor burden was followed by serial bioluminescence imaging. As shown in FIG. 2, CART22-65s (also known as C7002) mediated a superior anti-leukemia effect compared with the previously tested m971 (also known as C3042) and was at least as good as huCART19 (also known as C2137 CTL119). In summary, this data supported the development of CART22-65s as a clinical candidate.


Example 2: Pilot Study of Autologous Anti-CD22 Chimeric Antigen Receptor Redirected T Cells in Pediatric Patients with Chemotherapy Resistant or Refractory Acute Lymphoblastic Leukemia

This Example describes a single center, single arm, dual-cohort, open-label pilot study to determine, e.g., the feasibility and safety of a single dose (administered as split fractions, e.g., split doses) of autologous T cells expressing CD22 chimeric antigen receptors expressing tandem TCRζ and 4-1BB (TCRζ/4-1BB) co-stimulatory domains (referred to as “CART22” and “CART22-65s” cells) in e.g., pediatric patients with relapsed or refractory B-cell acute lymphoblastic leukemia.


Study Design


FIG. 3 depicts the study design. Subjects in Cohort 1 will be administered CART22 cells, and subject in Cohort 2 will be administered CART22-65s cells. The study will consist of four sequential phases: 1) a screening phase, 2) a manufacturing and pre-treatment phase, consisting of apheresis (if applicable) and chemotherapy (if applicable), 3) a treatment phase, consisting of a CART22 (Cohort 1) or CART22-65s (Cohort 2) transduced cell infusion and follow up evaluations, 4) long-term follow-up to monitor subjects for delayed adverse events.


CART22/CART22-65s cells transduced with a lentiviral vector to express anti-CD22 scFv TCRz:41BB administered by IV infusion. Cells will be transduced with the anti-CD22 TCRζ/4-1BB lentiviral vector, expanded in vitro and then frozen for future administration.


In some embodiments, patients will be given conditioning chemotherapy prior to CAR T cell infusion with the intent of lymphodepletion. Additionally, if the subject's white blood cell (WBC) count is ≤1,000/uL, conditioning/lymphodepleting chemotherapy will not be required. The chemotherapy will be planned so that the last dose is completed 1-4 days prior to the planned infusion of CART22/CART22-65s cells. If the period from chemotherapy to CART22/CART22-65s infusion is delayed 4 or more weeks, the patient will, e.g., be re-treated with lymphodepleting chemotherapy prior to CAR T cell infusion.


Subject Inclusion Criteria

A pediatric subject having relapsed or refractory B-cell ALL may be included in this study. In some embodiments, the subject has one or more of the following characteristics:

    • a. a second or greater bone marrow relapse; or
    • b. a marrow relapse after allogeneic HSCT and ≥6 months from SCT at infusion;
    • c. marrow relapse after CAR-modified T cell therapy;
    • d. refractory disease defined as having not achieved a CR after >2 chemotherapy regimens;
    • e. patients with Ph+ ALL are eligible if they are intolerant to or have failed tyrosine kinase inhibitor therapy;
    • f. ineligible for allogeneic SCT, e.g., because of:
      • i. Comorbid disease
      • ii. Other contraindications to allogeneic SCT conditioning regimen
      • iii. Lack of suitable donor; or
      • iv. Prior SCT
      • v. declines allogeneic SCT as a therapeutic option after documented discussion, with expected outcomes, about the role of SCT with a BMT physician not part of the study team; or
    • g. patients with CNS3 disease will be eligible if CNS disease is responsive to therapy.


In some embodiments, a subject with ALL has a documentation of CD22 tumor expression in bone marrow or peripheral blood by flow cytometry at relapse (or a recent marrow in the case of refractory disease). If the patient has received CD22-directed therapy (i.e. inotuzumab), then the marrow should be obtained after this therapy to show CD22 expression.


Treatment Regimen

Subjects <50 kg will receive 0.2-1×107 CART22 cells/kg or CART22-65s cells/kg as a split dose over three days as follows:

    • Day 1, 10% fraction: 0.2-1×106 CAR T cells/kg
    • Day 2, 30% fraction: 0.6-3×106 CAR T cells/kg; OR
    • Day 3, 60% fraction: 1.2-6×106 CAR T cells/kg


Subjects ≥50 kg will receive 1-5×108 CART22 cells or CART22-65s cells as a split dose over three days as follows:

    • Day 1, 10% fraction: 1-5×107 cells
    • Day 2, 30% fraction: 0.3-1.5×108 cells
    • Day 3, 60% fraction: 0.6-3×108 cells


The infusion will be scheduled to occur approximately 1 to 4 days following lymphodepleting chemotherapy but may be delayed. CAR T cells will be given on Days 1, 2 and 3. The first three subjects infused within each cohort will be staggered by 14 days to allow for monitoring of adverse events, including CRS.


Timing and Doses of Additional CART22/CART22-65s Infusions

For patients who have had i) evidence of brief B cell aplasia with subsequent B cell recovery, or ii) fever and other reversible toxicities without evidence of CAR expansion/LGLs, or iii) a partial or temporary response to the initial infusion, it may be appropriate to give more CART22/CART22-65s cells (e.g., additional treatment or CART22/CART22-65s infusions). Additional CART22/CART22-65s cells may be given no earlier than Day 14, if supplemental doses are available. Supplemental doses will generally consist of another 30% dose and should not exceed the 60% fraction (0.6-3×108cells).


Lymphodepleting Chemotherapy

In some embodiments, the subject is administered lymphodepleting chemotherapy, e.g, fludarabine (30 mg/m2/day×4 days) and cyclophosphamide (500 mg/m2/day×2 days). The chemotherapy will be planned so that the last dose is completed 1-4 days prior to the planned infusion of CART22/CART22-65s cells.









TABLE 11







ALL response criteria








Response category
Definition





Complete remission (CR)
All the following criteria are met:



Bone marrow



Trilineage Hematopoiesis (TLH)



and < 5% blasts



Peripheral blood



Neutrophils > 1.0 × 109/L, and



Platelets > 100 × 109/L, and



Circulating blasts < 1%



Extramedullary disease



No evidence of extramedullary disease (no



CNS disease, mediastinal disease CR, no



other extramedullary sites involvement)



Transfusion independency



No platelet and/or neutrophil transfusions



within 1 week before peripheral blood



sample for disease assessment


Complete remission with
All criteria for CR as defined above


incomplete blood
are met, except that the following exist:


count recovery (CRi)
Neutrophils < 1.0 × 109/L, or



Platelets < 100 × 109/L, or



Platelet and/or neutrophil transfusions



within week before peripheral blood sample



for disease assessment


Complete remission (CR)
All criteria for CR or CRi as defined above


with residual mediastinal
are met, except that mediastinal disease as


disease
defined by CRu or PR is observed:


No response (Treatment
Failure to attain the criteria needed for any


failure)
response categories


Relapsed Disease
Only in subjects with a CR or CRi:



Reappearance of blasts in the blood (≥1%),



or



Reappearance of blasts in bone marrow



(≥5%), or



(Re-)appearance of any extramedullary



disease after CR


Unknown
In case the response assessment was not



done, the baseline assessment was not done,



the assessment was incomplete or was not



done within the respective time frame. If



there is evidence of relapse, the overall



response will be assessed as relapse with



the relapsed component alone.









EXAMPLES INCORPORATED BY REFERENCE

The following examples are expressly incorporated by reference from published International Applications WO 2016/164731 and WO 2018/067992 as referenced below.


Example 9 on pages 525-526 of International Application WO 2016/164731 filed on 8 April 2016, titled: “Evaluation of CD22 CAR”, hereby incorporated by reference.


Example 10 on page 527 of International Application WO 2016/164731 filed on 8 Apr. 2016, titled: “Dose escalation study of CD22 CART treatment”, hereby incorporated by reference.


Example 13 on pages 533-535 of International Application WO 2016/164731 filed on 8 April 2016, titled: “Insertion mutations are a mechanism of resistance to CTL019 therapy in B cell Acute Lymphoid Leukemia (B-ALL)”, hereby incorporated by reference.


Example 14 on pages 535-536 of International Application WO 2016/164731 filed on 8 Apr. 2016, titled: “Expression of B-cell antigens in relapsed ALL cancer patients”, hereby incorporated by reference.


Example 15 on pages 536-537 of International Application WO 2016/164731 filed on 8 Apr. 2016, titled: “Expression of B-cell antigens in relapsed CD19-negative cancer patients”, hereby incorporated by reference.


Example 25 on pages 567-568 of International Application WO 2016/164731 filed on 8 Apr. 2016, titled: “Functional assays of CD22 CAR-expressing cells”, hereby incorporated by reference.


Example 8 on pages 535-538 of International Application WO 2018/067992 filed on 6 Oct. 2017, titled: “In vitro activity of CARTs bearing human anti-CD22 scFv with short linkers” hereby incorporated by reference.


Example 9 on pages 538-541 of International Application WO 2018/067992 filed on 6 October 2017, titled: “In vivo activity of CARTs bearing human anti-CD22 scFv with short linkers” hereby incorporated by reference.


Example 10 on pages 541-543 of International Application WO 2018/067992 filed on 6 Oct. 2017, titled: “Clinical efficacy of anti-CD22 CAR T cells for B-cell acute lymphoblastic leukemia correlates with scFv linker length and can be predicted using a xenograft model” hereby incorporated by reference.


Example 11 on pages 543-546 of International Application WO 2018/067992 filed on 6 October 2017, titled: “In vitro activity of CARTs bearing human anti-CD22 scFv with linker variants” hereby incorporated by reference.


Example 12 on pages 546-548 of International Application WO 2018/067992 filed on 6 Oct. 2017, titled: “In vivo activity of CARTs bearing human anti-CD22 scFv with linker variants” hereby incorporated by reference.


Example 13 on pages 548-551 of International Application WO 2018/067992 filed on 6 Oct. 2017, titled: “Generation, expression, and antigen activation of CARTs including tandem anti-CD19 and anti-CD22 scFVs” hereby incorporated by reference.


EQUIVALENTS

The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety. While this invention has been disclosed with reference to specific aspects, it is apparent that other aspects and variations of this invention may be devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims are intended to be construed to include all such aspects and equivalent variations.

Claims
  • 1. A method of treating a young adult or a pediatric subject having a hematological cancer comprising administering to the subject an effective number of cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR, wherein the CD22 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., a split-dosing regimen.
  • 2. A composition comprising cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR, for use in the treatment of a young adult or a pediatric subject having a hematological cancer comprising administering to the subject an effective number of said cells wherein the CD22 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., a split-dosing regimen.
  • 3. The method of claim 1 or the composition for use of claim 2, wherein the CD22 CAR-expressing cells are administered: a. at a dose of about 0.02×107 to 5.0×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg; orb. at a dose of about 0.5×108 to 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.
  • 4. A method of treating a subject having a hematological cancer, comprising administering to the subject an effective number of cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR, wherein the CD22 CAR-expressing cells are administered: a. at a dose of about 0.02×107 to 5.0×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg; orb. at a dose of about 0.5×108 to 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.
  • 5. A composition comprising cells that express a chimeric antigen receptor (CAR) molecule that binds CD22, e.g., a CD22 CAR, for use in the treatment of a subject having a hematological cancer comprising administering to the subject an effective number of said cells wherein the CD22 CAR-expressing cells are administered: a. at a dose of about 0.02×107 to 5.0×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg; orb. at a dose of about 0.5×108 to 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.
  • 6. The method of claim 4 or the composition for use of claim 5, wherein the CD22 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., a split-dosing regimen.
  • 7. The method or the composition for use of any of the preceding claims, wherein the hematological cancer is B cell ALL, e.g., relapsed and/or refractory B cell ALL.
  • 8. The method or the composition for use of any of the preceding claims, wherein the subject is a young adult or a pediatric subject, e.g., aged about 1-29 years, e.g., aged 1-29 years.
  • 9. The method of any of claim 1, 3 or 6, or the composition for use of any of claim 2-3 or 6, wherein the dose fractionation dosing regimen comprises a total dose administered in, e.g., one, two, three, or more separate administrations of a partial dose.
  • 10. The method of any of claim 1, 3, 6 or 9, or the composition for use of any of claim 2-3, 6 or 9, wherein the dose fractionation dosing regimen comprising a total dose comprises three administrations of a partial dose, e.g., a first partial dose, a second partial dose and a third partial dose.
  • 11. The method or composition for use of claim 9 or 10, wherein the first partial dose comprises a first percentage of the total dose and is administered on a first day of treatment.
  • 12. The method or composition for use of claim 11, wherein the first percentage comprising the first partial dose is about 10% (e.g., 10%) of the total dose.
  • 13. The method or composition for use of claim 9 or 10, wherein the second partial dose comprises a second percentage of the total dose and is administered on a second day of treatment.
  • 14. The method or composition for use of claim 13, wherein the second percentage comprising the second partial dose is about 30% (e.g., 30%) of the total dose.
  • 15. The method or composition for use of claim 9 or 10, wherein the third partial dose comprises a third percentage (e.g., the remaining percentage) of the total dose and is administered on a third day of treatment.
  • 16. The method or composition for use of claim 15, wherein the third percentage comprising the third partial dose is about 60% (e.g., 60%) of the total dose.
  • 17. The method or composition for use of any of claims 10-16, wherein the first partial dose, the second partial dose and the third partial dose are administered on consecutive days.
  • 18. The method or composition for use of any of claim 1-2 or 9-17, wherein the total cell dose comprises about 0.02×107 to 5.0×107 (e.g., about 0.2×107 to 1.0×107) CD22 CAR-expressing cells/kg or about 0.5×108 to 10×108 (e.g., about 1×108 to 5×108) CD22 CAR-expressing cells.
  • 19. The method or composition for use of claim 18, wherein the total cell dose comprises about 0.2×107 to 1.0×107 CD22 CAR-expressing cells/kg.
  • 20. The method or composition for use of claim 18 or 19, wherein: a. the first partial dose, e.g., 10% of the total dose, comprises about 0.2×106 to 1×106 CD22 CAR-expressing cells/kg;b. the second partial dose, e.g., 30% of the total dose, comprises about 0.6×106 to 3×106 CD22 CAR-expressing cells/kg; andc. the third partial dose, e.g., 60% of the total dose, comprises about 1.2×106 to 6×106 CD22 CAR-expressing cells/kg.
  • 21. The method or composition for use of any of claims 18-20, wherein the subject weighs less than 50 kg.
  • 22. The method or composition for use of claim 18, wherein the total cell dose comprises about 1×108 to 5×108 CD22 CAR-expressing cells/kg.
  • 23. The method or composition for use of claim 18 or 22, wherein: a. the first partial dose, e.g., 10% of the total dose, comprises about 1×107 to 5×107 CD22 CAR-expressing cells;b. the second partial dose, e.g., 30% of the total dose, comprises about 0.3×107 to 1.5×108 CD22 CAR-expressing cells; andc. the third partial dose, e.g., 60% of the total dose, comprises about 0.6×108 to 3×108 CD22 CAR-expressing cells.
  • 24. The method or composition for use of any of claim 18 or 22-23, wherein the subject weighs 50 kg or more than 50 kg.
  • 25. The method of claim 3 or 4, or the CD22 CAR-expressing cells for use of claim 3 or 5, wherein the CD22 CAR-expressing cells are administered at: a. a dose of about 0.02×107 to 1×107, about 1×107 to 1.5×107, about 1.5×107 to 2×107, about 2×107 to 2.5×107, about 2.5×107 to 3×107, about 3×107 to 3.5×107, about 3.5×107 to 4×107, about 4×107 to 4.5×107, or about 4.5×107 to 5×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg;b. a dose of about 0.02×107, about 0.04×107, about 0.06×107, about 0.08×107, about 1×107, about 1.5×107, about 2×107, about 2.5×107, about 3×107, about 3.5×107, about 4×107, about 4.5×107, or about 5×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg;c. a dose of about 0.5×108 to 1×108, about 1×108 to 1.5×108, about 1.5×108 to 2×108, about 2×108 to 2.5×108, about 2.5×108 to 3×108, about 3×108 to 3.5×108, about 3.5×108 to 4×108, about 4×108 to 4.5×108, about 4.5×108 to 5×108, about 5×108 to 6×108, about 6×108 to 7×108, about 7×108 to 8×108, about 8×108 to 9×108, or about 9×108 to 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg; ord. a dose of about 0.5×108, about 1×108, about 1.5×108, about 2×108, about 2.5×108, about 3×108, about 3.5×108, about 4×108, about 4.5×108, about 5×108, about 6×108, about 7×108, about 8×108, about 9×108, about 10×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.
  • 26. The method of any of claim 3-4 or 25, or the CD22 CAR-expressing cells for use of any of claim 3, 5 or 25, wherein the CAR-expressing cells are administered a. at a dose of about 0.2×107 to 1×107 viable CD22 CAR-expressing cells/kg, when the subject weighs <50 kg; orb. at a dose of about 1×108 to 5×108 viable CD22 CAR-expressing cells, when the subject weighs ≥50 kg.
  • 27. The method of any of claim 6-14, or 25-26, or the CD22 CAR-expressing cells for use of any of claim 6-14 or 25-26, wherein when the subject weighs <50 kg, the total dose comprises about 0.2×107 to 1×107 viable CD22 CAR-expressing cells/kg, and wherein a. the first partial dose, e.g., 10% of the total dose, comprises about 0.2×106 to 1×106 CD22 CAR-expressing cells/kg;b. the second partial dose, e.g., 30% of the total dose, comprises about 0.6×106 to 3×106 CD22 CAR-expressing cells/kg; andc. the third partial dose, e.g., 60% of the total dose, comprises about 1.2×106 to 6×106 CD22 CAR-expressing cells/kg.
  • 28. The method of any of claim 6-14, or 25-26, or the CD22 CAR-expressing cells for use of any of claim 6-14 or 25-26, wherein when the subject weighs ≥50 kg, the total dose comprises about 1×108 to 5×108 viable CD22 CAR-expressing cells, and wherein a. the first partial dose, e.g., 10% of the total dose, comprises about 1×107 to 5×107 CD22 CAR-expressing cells;b. the second partial dose, e.g., 30% of the total dose, comprises about 0.3×107 to 1.5×108 CD22 CAR-expressing cells; andc. the third partial dose, e.g., 60% of the total dose, comprises about 0.6×108 to 3×108 CD22 CAR-expressing cells.
  • 29. The method or composition for use of claim 9, comprising administering an additional dose of CD22 CAR-expressing cells.
  • 30. The method or composition for use of claim 29, wherein the additional dose, e.g., supplemental dose, of CD22 CAR-expressing: a. comprises about 0.6×106 to 3×106 cells/kg or about 0.3×108 to 1.5×108 cells;b. comprises less than about 1.2×106 to 6×106 cells/kg or about 0.6×108 to 3×108 cells; orc. is administered at least 13 days after administration of the first partial dose of CD22 CAR-expressing cells, e.g., administered on day 14 or later.
  • 31. The method or composition for use of any of the preceding claims, further comprising administering to the subject an effective number of cells that express a CAR molecule that binds CD19, e.g., a CD19 CAR.
  • 32. The method or composition for use of claim 31, wherein the CD22 CAR-expressing cells are administered before, after or concurrently with the administration of the CD19 CAR-expressing cells.
  • 33. The method or composition for use of claim 31 or 32, wherein the CD19 CAR-expressing cells are administered as a single dose infusion, e.g., a total dose is administered in a single infusion, e.g., at a dose of about 0.2×106 to 10×106 CD19 CAR-expressing cells/kg, e.g., about 2.0×106 CD19 CAR-expressing cells/kg.
  • 34. The method or composition for use of claim 31 or 32, wherein the CD19 CAR-expressing cells are administered according to a dose fractionation dosing regimen, e.g., split-dosing regimen described herein, wherein the dose fractionation dosing regimen comprises a total dose administered in, e.g., one, two, three, or more separate administrations of a partial dose.
  • 35. The method or compositions for use of claim 34, wherein the dose fractionation dosing regimen comprising a total dose comprises three administrations of a partial dose, e.g., a first partial dose, a second partial dose and a third partial dose.
  • 36. The method or composition for use of claim 35, wherein: a. the first partial dose comprises a first percentage (e.g., about 10%) of the total dose; and is administered on a first day of treatment;b. the second partial dose comprises a second percentage (e.g., about 30%) of the total dose; and is administered on a second day of treatment; andc. the third partial dose comprises a third percentage (e.g., about 60%) of the total dose; and is administered on a third day of treatment.
  • 37. The method or composition for use of claim 35 or 36, wherein the first partial dose, the second partial dose and the third partial dose are administered on consecutive days.
  • 38. The method or composition for use of any of claims 35-37, wherein the total cell dose comprises about 0.2×106 to 10×106 CD19 CAR-expressing cells/kg, e.g., about 2.0×106 CD19 CAR-expressing cells/kg.
  • 39. The method or composition for use of any of claims 35-38, wherein the first partial dose comprises about 0.2×106 CD19 CAR-expressing cells/kg, the second partial dose comprises about 0.6×106 CD19 CAR-expressing cells/kg, and the third partial dose comprises about 1.2×106 CD19 CAR-expressing cells/kg.
  • 40. The method or composition for use of any of the preceding claims, wherein the subject has been administered lymphodepleting chemotherapy.
  • 41. The method or composition for use of any of the preceding claims, wherein the subject has not been administered lymphodepleting chemotherapy.
  • 42. The method or composition for use of any of claims 31-41, wherein the subject has not relapsed to treatment with cells that express a CAR molecule, e.g., a CD19 CAR or a CD22 CAR therapy (e.g., a CD19 CAR monotherapy or a CD22 CAR monotherapy).
  • 43. The method or composition for use of any of claims 31-42, administration of the combination comprising a CD19 CAR and CD22 CAR prevents relapse in the subject, relative to a CD19 CAR monotherapy or a CD22 CAR monotherapy, or relative to a subject that has not received the combination.
  • 44. The method or composition for use of any of the preceding claims, wherein the cancer, e.g., a sample from the subject containing cancer cells, comprises cells that express CD19 and/or CD22.
  • 45. The method or composition for use of any of the preceding claims, wherein the CD22 CAR molecule comprises a CD22 binding domain, a transmembrane domain, and an intracellular signaling domain, and wherein said CD22 binding domain comprises one or more of light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of any CD22 light chain binding domain amino acid sequence listed in Table 6, 8 or 10, and one or more of heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of any CD22 heavy chain binding domain amino acid sequence listed in Table 6, 7, or 9.
  • 46. The method or composition for use of any of the preceding claims, wherein the CD22 CAR molecule comprises the amino acid sequence of SEQ ID NO: 835, or an amino acid sequence with at least 95% identity thereto.
  • 47. The method or composition for use of any of the preceding claims, wherein the CD22 CAR molecule comprises a CD22 binding domain comprising a light chain variable region of SEQ ID NO: 1333, or an amino acid sequence with at least 95% identity thereto, and a heavy chain variable region of SEQ ID NO: 1332, or an amino acid sequence with at least 95% identity thereto.
  • 48. The method or composition for use of any of claims 31-43, wherein the CD19 CAR molecule comprises a CD19 binding domain, a transmembrane domain, and an intracellular signaling domain, and wherein said CD19 binding domain comprises one or more of (e.g., all three of) light chain complementarity determining region 1 (LC CDR1), light chain complementarity determining region 2 (LC CDR2), and light chain complementarity determining region 3 (LC CDR3) of any CD19 scFv or light chain binding domain amino acid sequence listed in Tables 2 or 3, and one or more of (e.g., all three of) heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and heavy chain complementarity determining region 3 (HC CDR3) of any CD19 scFv or heavy chain binding domain amino acid sequence listed in Tables 2 or 3.
  • 49. The method or composition for use of any of claim 31-43 or 47, wherein the CD19 CAR molecule comprises any light chain variable region of a scFv listed in Tables 2 or 3 and any heavy chain variable region of a scFv listed Tables 2 or 3.
  • 50. The method or composition for use of claim 48, wherein the CD19 CAR molecule comprises a CD19 binding domain comprising the amino acid sequence of SEQ ID NO:2, or SEQ ID NO: 59, or an amino acid sequence with at least 95% identity thereto.
  • 51. The method or composition for use of any of the preceding claims, wherein the CAR molecule comprises a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154.
  • 52. The method or composition for use of any of claims 45-51, wherein the antigen binding domain is connected to the transmembrane domain by a hinge region.
  • 53. The method or composition for use of any of claims 45-52, wherein the hinge region comprises SEQ ID NO:14, or an amino acid sequence with at least 95% identity thereto.
  • 54. The method or composition for use of any of claims 45-53, wherein the intracellular signaling domain: a. comprises a costimulatory domain and/or a primary signaling domain;b. comprises a costimulatory domain comprising a functional signaling domain obtained from a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137),c. comprises a costimulatory domain comprising the amino acid sequence of SEQ ID NO:16 or SEQ ID NO:51;d. comprises a functional signaling domain of 4-1BB and/or a functional signaling domain of CD3 zeta; ore. comprises the amino acid sequence of SEQ ID NO: 16 and/or the amino acid sequence of SEQ ID NO:17 or SEQ ID NO:43.
  • 55. The method or composition for use of any of claims 45-54, wherein the CAR molecule further comprises a leader sequence, wherein, optionally, the leader sequence comprises SEQ ID NO: 13.
  • 56. The method or composition for use of any of the preceding claims, wherein the CAR-expressing cells comprise T cells (e.g., CD8+ or CD4+ T cells) or NK cells.
  • 57. The method or composition for use of any of the preceding claims, further comprising administering an additional agent, e.g., a checkpoint inhibitor, e.g., as described herein.
  • 58. The method or composition for use of any of claim 1-6 or 7-57, wherein the hematological cancer: a. is associated with expression of CD22 or CD19;b. is a leukemia or lymphoma;c. is a relapsed and/or refractory cancer; ord. is chosen from B-cell acute Lymphoid Leukemia (BALL), T-cell acute Lymphoid Leukemia (TALL), acute lymphoid leukemia (ALL), Prolymphocytic leukemia, Chronic myeloid leukemia (CML), blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma, Follicular lymphoma, Hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma (MCL), Marginal zone lymphoma, multiple myeloma, acute myeloid leukemia (AML), myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, or plasmablastic lymphoma.
  • 59. The method or composition for use of any of the preceding claims, wherein the subject: a. has previously received stem cell transplantation (SCT) therapy, e.g., allogeneic or autologous SCT; orb. is not eligible for SCT therapy, e.g., allogeneic or autologous SCT.
  • 60. The method or composition for use of claim 59, wherein the subject has previously received SCT therapy and the subject has, or is identified as having relapsed from the SCT therapy.
  • 61. The method or composition for use of claim 60, wherein the relapse from SCT therapy comprises a blood or bone marrow relapse and/or the relapse occurs at least 1-12 months, e.g., at least 6 months, after SCT therapy.
  • 62. The method or composition for use of any of the preceding claims, wherein the subject has, or is identified as having, a relapse, e.g., a second or greater relapse, e.g., bone marrow relapse, e.g., as described herein.
  • 63. The method or composition for use of any of the preceding claims, wherein the subject has, or is identified as having relapsed from a CAR-expressing cell therapy, e.g., a CAR-expressing cell therapy other than a CAR22-expressing cell therapy or a CAR19-expressing cell therapy.
  • 64. The method or composition for use of any of the preceding claims, wherein the subject has, or is identified as not having a response, e.g., a complete response or partial response, in response to one or more (e.g., 2, 3, 4, 5, 6, 7, or 8) chemotherapeutic agents, e.g., as described herein.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application 62/738,387 filed on Sep. 28, 2018, and U.S. Provisional Application 62/744,947 filed on Oct. 12, 2018, the entire contents of each of which are hereby incorporated by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/053601 9/27/2019 WO 00
Provisional Applications (2)
Number Date Country
62744947 Oct 2018 US
62738387 Sep 2018 US