Chimeric antigen receptor (CAR) T cells have substantial activity against CD19-expressing B cell malignancies in humans1,2. These receptors fuse the single-chain variable fragment (scFv) of an antibody specific to a tumor target with the signaling molecules of an effector T cell. This provides these cells with the specificity of an antibody and the effector function of T cells3,4.
Treatment of patients with solid tumors using CAR T cell therapy, however, has been less successful5,6. Potential barriers to CAR T cell efficacy against solid tumors include the suboptimal migration and persistence of CAR-T cells in the tumor microenvironment (TME), impaired function mediated by the immunosuppressive TME, and CAR T cell exhaustion. Previous work has shown that Th17 and Tc17 cells have enhanced persistence in solid tumors as compared to conventional Th1 cells7,8. It would be beneficial to determine if CAR T cells generated from Th/Tc17 cells are capable of persisting in the immunosuppressive TME of solid tumors.
Although CAR T cells targeting CD19 have been successful in treating patients with B cell malignancies1,2, the clinical activity of CAR T cells in patients with solid tumors has been modest5,6. Using a rational and sequential approach, a combinatorial immunotherapy strategy was implemented based on CAR T cells polarized to a Th/Tc17 phenotype, anti-PD1 mAb, STING agonist DMXAA and anti-GR-1 mAb.
Described herein are improved methods for enhancing CAR T cell trafficking and persistence in combination with methods of altering the immunosuppressive tumor microenvironment (TME) to sustain anti-tumor effects of the CAR T cells in solid tumors.
Described herein are methods of treating a disease. The methods include administering to a subject a chimeric antigen receptor modified T cell (CAR T cell) and a STING agonist.
In some embodiments, the CAR T cell is a second generation CAR T cell or a third generation CAR T cell. In some embodiments, the CAR T cell is a Th/Tc17 CAR T cell or a 7/15 CAR T cell.
In some embodiments, the STING agonist is a viral or bacterial product, such as DMXAA or cGAMP. In some embodiments, the CAR T cell and STING agonist are administered concurrently. In other embodiments, the CAR T cell and STING agonist are administered sequentially.
In some embodiments, the methods described herein further include administering to the subject a myeloid derived suppressor cell (MDSC) depleting agent. In some embodiments, the MDSC depleting agent is an antibody, e.g., an anti-Gr1 antibody. In some embodiments, the MDSC depleting agent is administered concurrently with the CAR T cell and the STING agonist. In other embodiments, the MDSC depleting agent is administered sequentially with the CAR T cell and the STING agonist.
In some embodiments, the methods described herein further include administering to the subject a PD-1 inhibitor. In some embodiments, the PD-1 inhibitor is an anti-PD-1 antibody. In some embodiments, the PD-1 inhibitor is administered concurrently with the CAR T cell and the STING agonist. In other embodiments, the PD-1 inhibitor is administered sequentially with the CAR T cell and the STING agonist. In some embodiments, the methods described herein further include inactivating PD-1 in the CAR T cell using gene editing, e.g., using at least one of CRISPR, TALEN, or ZFN.
In some embodiments, the disease is cancer, e.g., breast cancer. In some embodiments, the disease is selected from the group consisting of a sarcoma, a carcinoma, and a lymphoma. In certain embodiments, the disease is a solid tumor.
Also described herein are methods of treating cancer. The methods administering to a subject a Th17 CAR T cell, a STING agonist, a MDSC depleting agent, and a PD-1 inhibitor.
In some embodiments, the Th17 CAR T cell, STING agonist, MDSC depleting agent, and the PD-1 inhibitor are administered concurrently. In alternative embodiments, the Th17 CAR T cell, STING agonist, MDSC depleting agent, and the PD-1 inhibitor are administered sequentially.
In some embodiments, the STING agonist is DMXAA. In some embodiments, the MDSC depleting agent is an antibody. In some embodiments, the MDSC depleting agent is an anti-Gr-1 antibody. In some embodiments, the PD-1 inhibitor is an anti-PD-1 antibody.
In some embodiments, the cancer is selected from the group consisting of a sarcoma, a carcinoma, and a lymphoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is a solid tumor. In some embodiments, the subject exhibits symptoms of a cytokine-release like syndrome, and further comprising administering to the subject an anti-IL-6 antibody.
Also disclosed herein are methods of producing Th/Tc17 CAR T cells. The methods include transducing CD4 and CD8 activated T cells with LH28z virus; culturing the transduced T cells with Th17 cytokine cocktail; reactivating the cultured T cells; and culturing the reactivated T cells in Th17 cytokine cocktail plus IL-23.
In some embodiments, the Th17 cytokine cocktail comprises TGF-β1, IL-1β, TNF, IL-6, anti-IFNγ, and anti-IL-2. In some embodiments, the T cells are activated using anti-CD3 and anti-CD28. In some embodiments, the cultured T cells are reactivated using anti-CD3 and anti-CD28. In some embodiments, the LH28z virus is prepared from the LH28z construct comprising a scFv, hinge and transmembrane domain from CD8, and intracellular domains from CD28 and CD3ζ.
The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee.
Chimeric antigen receptor T (CAR-T) cells have substantial activity against CD19-expressing B cell malignancies. However, treatment of patients with solid tumors using CAR-T cell therapy has been less than effective for a variety of reasons including suboptimal migration and persistence of CAR-T cells in the tumor microenvironment (TME), impaired function mediated by the immunosuppressive TME, and CAR-T cell exhaustion. Described herein are improved chimeric antigen receptor T (CAR-T) cells and methods of producing the same. Also described herein are methods of treating a disease, e.g., a solid tumor, by administering the improved CAR-T cells.
CAR T cells are produced by obtaining T cells, such as from a subject in need thereof or from a donor subject and manipulating the cells such that they include chimeric antigen receptors (CARs). The CARs provide the ability to target specific proteins on cancer cells and include an antigen recognition domain, an extracellular hinge region, a transmembrane domain, and an intracellular T cell signaling domain. CAR T cells may be classified as first generation, second generation, third generation, or fourth generation. First generation CARs were engineered with only the CD3ζ domain. Second generation CARs were engineered with the CD3ζ domain and a costimulatory signaling domain (e.g., CD28 or 4-1BB). Third generation CARs are engineered to include the CD3ζ (domain in addition to two costimulatory signaling domains (e.g., both CD28 and CD137). Finally, fourth generation CARs, also referred to as T-cells redirected for universal cytokine-mediated killing (TRUCKs) are engineered to include the CD3ζ domain, two costimulatory signaling domains (e.g., both CD28 and CD137), and some additional genetic modification, such as the addition of transgenes for cytokine secretion or additional costimulatory signaling domains.
Described herein are CAR-T cells developed to enhance CAR-T cell function in solid tumors. The CAR-T cells may be second and/or third generation CAR-T cells. In some embodiments, a CAR-T cell includes the costimulatory domain from CD28 or CD137. In one embodiment, a CAR-T cell includes the costimulatory domains from CD28 and CD137. In some embodiments, a CAR-T cell includes the transmembrane domain from CD8. In some embodiments, the CAR-T cells are 7/15 CAR-T cells. In some embodiments, the CAR-T cells are Th/Tc17 CAR-T cells. A population of CAR-T cells may include a roughly equal ratio of CD4+ to CD8+ cells. In some aspects, the CD4+ and CD8+ T cells are present in a ratio of about 2:1, 1.75:1, 1.5:1, 1.25:1, 1:1, 1:1.25, 1:1.5, 1:1.75, or 1:2.
Disclosed herein are methods of treating a disease comprising administering to a subject a chimeric antigen receptor modified T cell (CAR T cell). The CAR T cells are administered to the subject to target and destroy cancer cells. In some embodiments, the CAR T cell is a second or third generation CAR T cell. In some embodiments, a CAR T cell is a Th/Tc17 CAR T cell or a 7/15 CAR T cell. In certain embodiments, the CAR T cells are Th/Tc17 CAR T cells. Th/Tc17 CAR T cells may exhibit superior persistence in a tumor as compared to other CAR T cells, such as IL7/15 CAR T cells. In some embodiments, the Th/Tc17 CAR T cells are engineered to include the costimulatory domain from CD28. In some embodiments, the CAR construct for Th/Tc17 CAR T cells includes a scFv (7.16.4), hinge and transmembrane domain from CD8, and intracellular domains from CD28 and CD3ζ. In some embodiments, the CAR T cells are either derived from T cells removed from a subject in need thereof, e.g., are autologous, or are derived from T cells removed from a healthy donor, e.g., are allogenic. In some aspects, T cells are engineered from stem cells.
In some embodiments, a CAR T cell is administered to a subject, e.g. in a subject in need thereof, such as a subject diagnosed with cancer, in combination with a STING agonist. A STING agonist activates the STING pathway, which, in some aspects, can cause a tumor to go from “cold” to “hot”. A “cold tumor” is used herein to refer to a tumor that does not provoke an immune response, while a “hot tumor” is used herein to refer to a tumor that is more prone to recognition by the immune system. In some embodiments, a STING agonist increases expression of CXCL9 and CXCL10 by myeloid cells located within the tumor microenvironment.
In some embodiments, a STING agonist is any viral or bacterial product that activates the STING pathway. In certain embodiments, a STING agonist is selected from the group consisting of DMXAA, cGAMP, CMA, c-Di-AMP, and c-Di-GMP. In certain embodiments, a STING agonist is DMXAA. A STING agonist may be administered prior to administration of a CAR T cell, concurrently with a CAR T cell, or after administration of a CAR T cell. In certain embodiments, a STING agonist is administered prior to administration of the CAR T cells. In some aspects, administration of a STING agonist in combination with a CAR T cell enhanced the persistence of the CAR T cells in the tumor microenvironment (TME). In certain aspects, administration of a STING agonist increased antitumor efficacy and/or overall survival of the CAR T cells.
In some embodiments, a myeloid-derived suppressor cell (MDSC) depleting agent is administered to the subject in combination with the CAR T cells, e.g., Th/Tc17 CAR T cells engineered to include a CD28 costimulatory domain, and the STING agonist, e.g., DMXAA. An MDSC depleting agent may be a MDSC targeting agent. In some aspects, an MDSC depleting agent is an antibody, such as an anti-Gr-1 antibody. In some aspects, MDSC is modified, e.g., depleted, by an agent that leads to differentiation or maturation of the MDSCs. Non-limiting examples of agents that lead to differentiation or maturation of the MDSCs include retinoids, chemotherapy drugs (e.g., gemcitabine), and antibodies that inhibit the M-CSF, g-CSF, or GM-CSF receptors. In other aspects, the migration of MDSCs to the tumor environment is diminished by inhibiting the function of CXCL2, CXCL1, or CXCL5. In still other aspects, the function of MDSCs is targeted by inhibiting the activity of proteins important for the function of these cells, such as Cox-2, PGE2, arginase I/II, or iNOS. In some embodiments, the MDSC depleting or targeting agent is administered concurrently with CAR T cells and/or STING agonist. In other embodiments, the MDSC depleting agent is administered sequentially with the CAR T cells and/or STING agonist. In some aspects, an MDSC depleting agent is administered concurrently with CAR T cells. In some aspects, an MDSC depleting agent is administered within the first two weeks following CAR T cell therapy. In some aspects, an MDSC depleting agent is administered concurrently with a STING agonist. In some aspects, a STING agonist, e.g., DMXAA, is administered to a subject, followed by administration of a population of CAR T cells, e.g., Th/Tc17 CAR T cells engineered to include a CD28 costimulatory domain, and further followed by administration of an MDSC depleting agent, e.g., anti-Gr-1 antibody. In certain aspects, administration of an MDSC depleting agent to a subject in combination with CAR T cells and a STING agonist improves overall survival of the subject and improves control of tumor growth.
In some embodiments, a PD-1 inhibitor is administered to the subject in combination with the CAR T cells and the STING agonist. In certain embodiments, the PD-1 inhibitor is administered to the subject in combination with the MDSC depleting agent, e.g., anti-Gr-1 antibody, a population of CAR T cells, e.g., Th/Tc17 CAR T cells engineered to include a CD28 costimulatory domain, and the STING agonist, e.g., DMXAA. In some aspects, a PD-1 inhibitor is an anti-PD-1 antibody or a fusion protein targeting PD-1. In some embodiments, PD-1 is inactivated using gene editing in the CAR T cell. In certain embodiments, PD-1 is inactivated via CRISPR/Cas9, TALENs, or ZFN in the CAR T cell. In some embodiments, a PD-1 inhibitor, is administered concurrently with the MDSC depleting agent, CAR T cells and/or the STING agonist. In other embodiments, the PD-1 inhibitor is administered prior to the MDSC targeting agent, and in some aspects is administered concurrently with or, alternatively after, CAR T cell therapy. In other embodiments, the PD-1 inhibitor is administered sequentially with the MDSC depleting agent, the CAR T cells, and the STING agonist. In some aspects, the PD-1 inhibitor, e.g., anti-PD-1 antibody, is administered to a subject concurrently with an MDSC depleting agent, e.g., anti-Gri-1 antibody. In some aspects, a STING agonist, e.g., DMXAA, is administered to a subject, followed by administration of a population of CAR T cells, e.g., Th/Tc17 CAR T cells engineered to include a CD28 costimulatory domain, and further followed by administration of an MDSC depleting agent, e.g., anti-Gr-1 antibody, and a PD-1 inhibitor, e.g., anti-PD-1 antibody. In certain aspects, administration of a PD-1 inhibitor to a subject in combination with CAR T cells and a STING agonist improves overall survival of the subject and improves control of tumor growth.
In some embodiments, a subject is further administered an anti-IL-6 antibody, an anti-IL-6R antibody, or an antibody or protein targeting IL-1. In certain embodiments, the anti-IL-6 antibody is administered to the subject after administration of the STING agonist, the population of CAR T cells, the MDSC depleting agent, and the PD-1 inhibitor. In some aspects, the anti-IL-6 antibody is administered at the peak time of symptoms from CRS, or about 3 to 7 days, or about 5 days after administration of the STING agonist, the population of CAR T cells, the MDSC depleting agent, and the PD-1 inhibitor.
In some embodiments, a subject has a disease or disorder. In certain embodiments, the disease is cancer, which term is generally used interchangeably to refer to a disease characterized by one or more tumors, e.g., one or more malignant or potentially malignant tumors. The term “tumor” as used herein encompasses abnormal growths comprising aberrantly proliferating cells. As known in the art, tumors are typically characterized by excessive cell proliferation that is not appropriately regulated (e.g., that does not respond normally to physiological influences and signals that would ordinarily constrain proliferation) and may exhibit one or more of the following properties: dysplasia (e.g., lack of normal cell differentiation, resulting in an increased number or proportion of immature cells); anaplasia (e.g., greater loss of differentiation, more loss of structural organization, cellular pleomorphism, abnormalities such as large, hyperchromatic nuclei, high nuclearxytoplasmic ratio, atypical mitoses, etc.); invasion of adjacent tissues (e.g., breaching a basement membrane); and/or metastasis. The term “tumor” includes malignant solid tumors, e.g., carcinomas (cancers arising from epithelial cells), sarcomas (cancers arising from cells of mesenchymal origin), and malignant growths in which there may be no detectable solid tumor mass (e.g., certain hematologic malignancies). Cancer includes, but is not limited to: breast cancer; biliary tract cancer; bladder cancer; brain cancer (e.g., glioblastomas, medulloblastomas); cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms including acute lymphocytic leukemia and acute myelogenous leukemia; T-cell acute lymphoblastic leukemia/lymphoma; hairy cell leukemia; chronic lymphocytic leukemia, chronic myelogenous leukemia, multiple myeloma; adult T-cell leukemia/lymphoma; intraepithelial neoplasms including Bowen's disease and Paget's disease; liver cancer; lung cancer; lymphomas including Hodgkin's disease and lymphocytic lymphomas; neuroblastoma; melanoma, oral cancer including squamous cell carcinoma; ovarian cancer including ovarian cancer arising from epithelial cells, stromal cells, germ cells and mesenchymal cells; neuroblastoma, pancreatic cancer; prostate cancer; rectal cancer; sarcomas including angiosarcoma, gastrointestinal stromal tumors, leiomyosarcoma, rhabdomyosarcoma, liposarcoma, fibrosarcoma, and osteosarcoma; renal cancer including renal cell carcinoma and Wilms tumor; skin cancer including basal cell carcinoma and squamous cell cancer; testicular cancer including germinal tumors such as seminoma, non-seminoma (teratomas, choriocarcinomas), stromal tumors, and germ cell tumors; thyroid cancer including thyroid adenocarcinoma and medullary carcinoma. It will be appreciated that a variety of different tumor types can arise in certain organs, which may differ with regard to, e.g., clinical and/or pathological features and/or molecular markers. Tumors arising in a variety of different organs are discussed, e.g., the WHO Classification of Tumours series, 4th ed, or 3rd ed (Pathology and Genetics of Tumours series), by the International Agency for Research on Cancer (IARC), WHO Press, Geneva, Switzerland, all volumes of which are incorporated herein by reference.
In certain embodiments, a subject is administered a therapeutic regimen comprising a population of CAR T cells, e.g., Th/Tc17 CAR T cells engineered to include a CD28 costimulatory domain, a STING agonist, e.g., DMXAA, an MDSC depleting agent, e.g., anti-Gr-1 antibody, and a PD-1 inhibitor, e.g., anti-PD-1 antibody, to treat a cancer, e.g., a cancer characterized by one or more tumors. In some aspects, the administration of the therapeutic regimen to the subject results in the subject suffering from cytokine-release like syndrome (CRS). A subject suffering from CRS may further be administered an anti-IL-6 antibody.
The agents, e.g., the population of CAR T cells, STING agonist, MDSC depleting agent, PD-1 inhibitor, and/or anti-IL-6 antibody, may be administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients.
The agents may be formulated into preparations in solid, semi-solid, liquid or gaseous forms such as tablets, capsules, powders, granules, ointments, solutions, depositories, inhalants and injections, and usual ways for oral, parenteral or surgical administration. The invention also embraces pharmaceutical compositions which are formulated for local administration, such as by implants.
Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent. Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion.
In some embodiments, agents may be administered directly to a tissue. Direct tissue administration may be achieved by direct injection. The agents may be administered once, or alternatively they may be administered in a plurality of administrations. If administered multiple times, the peptides may be administered via different routes. For example, the first (or the first few) administrations may be made directly into the affected tissue while later administrations may be systemic.
For oral administration, compositions can be formulated readily by combining the agent with pharmaceutically acceptable carriers well known in the art. Such carriers enable the agents to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained as solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Optionally the oral formulations may also be formulated in saline or buffers for neutralizing internal acid conditions or may be administered without any carriers.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
Pharmaceutical preparations which can be used orally include push fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Microspheres formulated for oral administration may also be used. Such microspheres have been well defined in the art. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
The compounds, when it is desirable to deliver them systemically, may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like. Lower doses will result from other forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated in some embodiments to achieve appropriate systemic levels of compounds.
Specific examples of certain aspects of the inventions disclosed herein are set forth below in the Examples.
One skilled in the art readily appreciates that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The details of the description and the examples herein are representative of certain embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Modifications therein and other uses will occur to those skilled in the art. These modifications are encompassed within the spirit of the invention. It will be readily apparent to a person skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention.
The articles “a” and “an” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to include the plural referents. Claims or descriptions that include “or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention also includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process. Furthermore, it is to be understood that the invention provides all variations, combinations, and permutations in which one or more limitations, elements, clauses, descriptive terms, etc., from one or more of the listed claims is introduced into another claim dependent on the same base claim (or, as relevant, any other claim) unless otherwise indicated or unless it would be evident to one of ordinary skill in the art that a contradiction or inconsistency would arise. It is contemplated that all embodiments described herein are applicable to all different aspects of the invention where appropriate. It is also contemplated that any of the embodiments or aspects can be freely combined with one or more other such embodiments or aspects whenever appropriate. Where elements are presented as lists, e.g., in Markush group or similar format, it is to be understood that each subgroup of the elements is also disclosed, and any element(s) can be removed from the group. It should be understood that, in general, where the invention, or aspects of the invention, is/are referred to as comprising particular elements, features, etc., certain embodiments of the invention or aspects of the invention consist, or consist essentially of, such elements, features, etc. For purposes of simplicity those embodiments have not in every case been specifically set forth in so many words herein. It should also be understood that any embodiment or aspect of the invention can be explicitly excluded from the claims, regardless of whether the specific exclusion is recited in the specification. For example, any one or more nucleic acids, polypeptides, cells, species or types of organism, disorders, subjects, or combinations thereof, can be excluded.
Where the claims or description relate to a composition of matter, e.g., a nucleic acid, polypeptide, cell, or non-human transgenic animal, it is to be understood that methods of making or using the composition of matter according to any of the methods disclosed herein, and methods of using the composition of matter for any of the purposes disclosed herein are aspects of the invention, unless otherwise indicated or unless it would be evident to one of ordinary skill in the art that a contradiction or inconsistency would arise. Where the claims or description relate to a method, e.g., it is to be understood that methods of making compositions useful for performing the method, and products produced according to the method, are aspects of the invention, unless otherwise indicated or unless it would be evident to one of ordinary skill in the art that a contradiction or inconsistency would arise.
Where ranges are given herein, the invention includes embodiments in which the endpoints are included, embodiments in which both endpoints are excluded, and embodiments in which one endpoint is included and the other is excluded. It should be assumed that both endpoints are included unless indicated otherwise. Furthermore, it is to be understood that unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in different embodiments of the invention, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise. It is also understood that where a series of numerical values is stated herein, the invention includes embodiments that relate analogously to any intervening value or range defined by any two values in the series, and that the lowest value may be taken as a minimum and the greatest value may be taken as a maximum. Numerical values, as used herein, include values expressed as percentages. For any embodiment of the invention in which a numerical value is prefaced by “about” or “approximately”, the invention includes an embodiment in which the exact value is recited. For any embodiment of the invention in which a numerical value is not prefaced by “about” or “approximately”, the invention includes an embodiment in which the value is prefaced by “about” or “approximately”. “Approximately” or “about” generally includes numbers that fall within a range of 1% or in some embodiments within a range of 5% of a number or in some embodiments within a range of 10% of a number in either direction (greater than or less than the number) unless otherwise stated or otherwise evident from the context (except where such number would impermissibly exceed 100% of a possible value). It should be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one act, the order of the acts of the method is not necessarily limited to the order in which the acts of the method are recited, but the invention includes embodiments in which the order is so limited. It should also be understood that unless otherwise indicated or evident from the context, any product or composition described herein may be considered “isolated”.
Development of an Orthotopic Immune Competent Model to Evaluate CAR T Cell Therapy Given the need to understand mechanistically how to enhance CAR T cell function in solid tumors, second and third generation murine CAR constructs were developed targeting the proto-oncogene Neu (
The antitumor effects of CAR T cells were evaluated in an orthotopic murine breast tumor model of locally advanced disease in immunocompetent mice (
Th/Tc 17 CAR T Cells have Enhanced In Vivo Persistence
Th17 cells and their counterpart Tc17 cells (CD8+ T cells that express IL-17A) have longer persistence in the TME, in part due to their better survival in vivo5,6. To evaluate if Th/Tc17 CAR T cells had better migration and/or persistence, LH28z Th/Tc17 CAR T cells were generated (termed Th/Tc17 CAR T cells). These cells were compared with 7/15 CAR T cells. Transduction efficacy (
Next, the in vivo antitumor effects of Th/Tc17 CAR T cells were compared to those of 7/15 CAR T cells. Th/Tc17 CAR T cells were superior to 7/15 CAR T cells in controlling tumor growth up to day 5 post-infusion (
Given the short-term efficacy of Th/Tc17 CAR T cells in vivo, an approach that would further enhance the migration and persistence of CAR T cells in the TME was sought. Recent studies have shown a critical role for the STING pathway in generating “warm” tumors in the setting of checkpoint inhibitor therapy9. Thus, the activity of the STING agonist, DMXAA, was evaluated in combination with CAR-T cells. Mice were injected with NT2 tumors, and when tumors reached approximately 50 mm2, mice were given 7/15 or Th/Tc17 CAR T cells with DMXAA, which was given at a site remote from the tumor. Interestingly, DMXAA enhanced the persistence of 7/15 CAR T cells in the TME (
Single cell transcriptome sequencing (scRNA-seq) was performed on CD45+ cells isolated from the TME of mice that received Th/Tc17 CAR-T with or without DMXAA. These evaluations were performed on day 7 post CAR T cell therapy, a time point where the greatest tumor control was observed, and at day 10 when tumors began to regrow (
The most noticeable difference in the TME in mice receiving DMXAA was a substantial increase in CAR T cells compared to animals given Th/Tc17 CAR T cells without DMXAA (
A combination of Tc17 and Th17 cells were necessary for anti-tumor efficacy using DMXAA as Tc17 cells alone were not effective in tumor control (
CAR T cell Exhaustion Limits In Vivo Efficacy
LH28z Th/Tc17 CAR T cell therapy given with DMXAA controlled tumor growth for approximately 10 days but did not eradicate the tumor. To investigate the cause of loss of activity of LH28z Th/Tc17 CAR T cells with DMXAA, scRNA-seq of the TME was compared at days 7 and 10 post CAR T cell therapy. As early as day 7 post LH28z Tc/Th17 CAR T cell infusion, increased expression was found of PDCD1 (PD-1), LAG3, CD160 and TOX by CAR T cells with decreased expression of TCF7 (
Alteration of Myeloid Cells in the TME is Associated with the Function of DMXAA
DMXAA can modify the function of innate immune cells12,13. Thus, the effects of the STING agonist on myeloid cells in the TME was evaluated. Significant differences were found in the presence of M1-like and M2-like macrophages, inflammatory-like myeloid cells (iMCs) and myeloid derived suppressor-like cells (MDSC) in mice treated with and without DMXAA (
A second cluster of myeloid cells was found after treatment with DMXAA with greatly enhanced expression of the transcription factor BATF2, the chemokine ligands CXCL10 and CCL5, and downstream genes related to inflammatory function15, including SOCS1, NOS2 and IL12A (
To characterize the TME immediately prior to tumor growth, myeloid cell subsets were evaluated at day 10 post Th/Tc17 CAR T cell infusion with DMXAA. As shown in
Given the data regarding the expression of PD-1 on CAR T cells and the marked changes in the immunosuppressive nature of the TME, targeting these pathways was evaluated as to whether they could enhance the long-term efficacy of LH28z Th/Tc17 CAR T cells plus DMXAA. To this end, mice were treated with LH28z Th/Tc17 CAR T cells plus DMXAA along with anti-PD-1 and anti-GR-1 mAb therapy (
To evaluate whether combination therapy with DMXAA, anti-PD-1 and anti-GR-1 mAbs could also enhance the anti-tumor activity of 7/15 CAR T cells, tumor growth was compared in mice receiving anti-GR-1 and anti-PD-1 mAb with DMXAA and given either 7/15 or Th/Tc17 CAR T cells. Much greater anti-tumor activity was found with the administration of Th/Tc17 CAR T cells compared to 7/15 CAR T cells when combined with DMXX and anti-PD-1 and anti-GR-1 mAbs (
In conclusion, sustained tumor regression caused by Th/Tc17 CAR-T cells was associated with enhanced proliferation and expansion of central memory T cells.
Efficacy Associated with Cytokine-Like Release Syndrome
Treatment of mice with Th/Tc17 CAR T cells, DMXAA, and anti-PD-1 and anti-GR-1 mAbs led to significant tumor control with the majority of mice tumor-free 30 days post treatment. However, a small subset of mice that received this combination therapy was found to exhibit significant weight loss and hunching consistent with a cytokine-release like syndrome (
Although CAR T cells targeting CD19 have been successful in treating patients with B cell malignancies1,2, the clinical activity of CAR T cells in patients with solid tumors has been modest5,6. Using a rational and sequential approach, a combinatorial immunotherapy strategy was implemented based on CAR T cells polarized to a Th/Tc17 phenotype, anti-PD1 mAb, STING agonist DMXAA and anti-GR-1 mAb that leads to tumor eradication in an immunocompetent syngeneic model of breast cancer. The most compelling finding was the ability of DMXAA, given at a site remote of the tumor, to promote CAR T cell recruitment and persistence at the tumor site. It is believed that these data are the first to demonstrate the function of a STING agonist in altering the trafficking properties of adoptive T cellular products. Furthermore, this activity did not require intratumoral injection of the STING agonist.
A critical aspect of effective CAR T cell therapy is the ability of the transferred cells to traffic to tumor sites. CAR T cells expanded ex vivo and infused intravenously in patients home physiologically to secondary lymphoid organs such as the bone marrow and lymph nodes, which explains in part the robust antitumor effects of CAR T cells observed in patients with leukemia, lymphoma and multiple myeloma18. In contrast, T cells do not constitutively traffic to breast tissue. Th/Tc17 cells have enhanced biodistribution to peripheral tissues as compared to Th1 cells, and this may explain in part our observation that Th/Th17 CAR T cells showed superior persistence in the tumor compared to IL7/15 CAR T cells (
The immunocompetent mouse model developed allowed for the assessment of how the myeloid tumor compartment affected the antitumor activity of CAR T cells. It was demonstrated that the persistence of Th/Tc17 CAR T cells in the TME was associated with enhanced expression of genes associated with M1-like macrophages and a marked loss of genes associated with M2-like macrophages and MDSC-like cells. The decrease in BATF2-expressing iMCs was associated with the inability of CAR T cells to control tumor growth. Thus, these data suggest that enhancing CAR T cell trafficking and persistence without altering the immunosuppressive TME is unlikely to lead to significant sustained anti-tumor effects in solid tumors.
Th/Tc17 CAR T cell persistence within the TME, which is promoted by the pro-inflammatory myeloid switch caused by a STING agonist, does not prevent exhaustion of CAR T cells, which requires checkpoint blockade to sustain CAR T cell mediated tumor control. Moreover, a rapid influx of immunosuppressive monocytic MDSCs within the TME was also observed and associated with diminished control of tumor growth. Previous work had shown that CAR T cells specific for CEA were inhibited by liver myeloid derived suppressor cells when given via the portal vein to mice with liver metastasis26. Here, it was found that depletion of MDSC-like cells in combination with CAR T cells, STING agonist and PD-1 blockade allowed sustained regression of the tumor. However, it was observed that the optimal combination also led to the development of rapid cachexia consistent with a cytokine release syndrome. Recent work has highlighted the critical role of myeloid cells in the pathogenesis of cytokine release syndrome27-29. The data suggests that MDSCs, and perhaps other immunosuppressive myeloid cells, limit the anti-tumor efficacy of CAR T cells but also attenuate the severity of the inflammatory reaction. Fully licensing CAR T cells by depleting MDSCs exacerbated a cytokine release syndrome, which responded to IL-6 blockade as observed in patients receiving CAR T cells targeting CD19.
In summary (
3T3 cell lines were obtained from the ATCC and cultured in DMEM (Gibco) supplemented with 10% fetal bovine serum (FBS; Gemini), and 1% Pen/Strep (Invitrogen). 3T3-Neu and NT2 tumor have been previously described30. 3T3-Neu cultures were made as described and cultured with the addition of 0.3 μM methotrexate. NT2 (Passage 6) were cultured in RPMI-1640 (Gibco) supplemented with 20% FBS, 2 mM L-glutamine (Invitrogen), 12 mM HEPES (Invitrogen), 0.1 mM NEAA (Invitrogen), 1 mM sodium pyruvate (Invitrogen), 1% Pen/Strep, 50 μM 2-mercaptoethanol (Invitrogen) and 0.2 U/ml Novolin R-insulin (Novo Nordisk). The virus encoding the LH28z CAR was cultured in PLAT-E cells from Cell Biolabs Inc. in DMEM supplemented with 10% FBS and 1% Pen/Strep. All cells used for the generation of the CAR constructs were used within 3 passages in vitro and 0.05% trypsin (Invitrogen) was used for digestion between passages.
7/15 and Th/Tc17 CARs contain the scFv of LH28z construct generated from the 7.16.4 antibody. Whole RNA from the 7.16.4 hybridoma was isolated (RNEasy Kit, Qiagen) and made into cDNA (M-MLV Reverse Transcriptase kit, Invitrogen). Antibody heavy and light chains were cloned with IgH or IgL primer mix (iRepertoire Inc.) and sequenced by Sanger sequencing (Genewiz). CAR cDNAs were cloned into a construct containing murine CD8 transmembrane domain and intracellular domain from murine CD28 or 4-1BB and CD3ξ (LH28z, LHBBz). The CAR construct was then cloned into MSGV plasmid and driven by 5′ LTR. The plasmid construct have been previously described32.
Plasmids encoding CAR (LH28z) and pCL749 Eco retrovirus packaging vector (Novus) were co-transfected into PLAT-E cells using lipofectamine 2000 (Invitrogen). Supernatant from LH28z-transfected PLAT-E cells was collected after 48 hours and stored at −80° C. For T cell transduction, LH28z or LHBBz or LH28z/LHBBz virus were plated onto a retronectin-coated plate (Takara) and incubated at 32° C. for 2 h followed by the additional of activated T cells. The co-culture was spun at 1200×g for 90 minutes and then stored at 370 C overnight.
For 7/15 CAR T cells, splenocytes from 8-14 w old female 757 FVB/NJ mice (Jackson Labs) were harvested and activated using plate bound anti-CD3 (145-2C11, Invitrogen) and anti-CD28 mAb (37.51, Invitrogen) and cultured in RPMI-1640 supplemented with 10% FBS, 2 mM L-glutamine, 12 mM HEPES, 0.1 mM NEAA, 1 mM sodium pyruvate, 1% Pen/Strep, 50 μM 2-ME, along with the addition of 1 μg/ml IL-7 and IL-15 (Peprotech). 48 hours later T cells were transduced with retrovirus and cultured for an additional 3 days with IL-7 and IL-15. For Th/Tc17 CAR T cells, naïve CD4+ and CD8+ T cells were isolated from the spleen of 8-14 week old female FVB/NJ mice (Jackson Labs) using murine naive CD4/CD8 T Cell Isolation Kit (Miltenyi) according to the manufacturer's instructions. T cells were activated using plate bound anti-CD3 and anti-CD28 mAb (Invitrogen). For the first week, the culture was maintained as described above with the addition of TGF-β1 (2 ng/ml), IL-1β (10 ng/ml), TNF (20 ng/ml), IL-6 (30 ng/ml) (Peprotech), anti-IFNγ (R4-6A2, 10 μg/ml, Bioxcell) and anti-IL-2 (JES6-5H4, 10 μg/ml, Bioxcell) (Th17 cytokine cocktail). Two days post-activation, T cells were transduced with retrovirus and kept in culture with Th17 cytokine cocktail for the rest of the week. During the second week, cultured T cells were re-activated using plate bound anti-CD3 and anti-CD28 mAb and maintained in Th17 cytokine cocktail plus IL-23 (15 ng/ml) (Peprotech).
8-12 week old female FVB-Neu mice (Jackson Labs) were used under a protocol approved by University of North Carolina Institutional Animal Care and Use Committee. For tumor inoculation, mice were intradermally injected with 5×104 NT2 tumor cells into an axillary mammary gland. 21 days later (day 0), 1.5×106 CD4+ and 1.5×106 CD8+ CAR T cells were injected intravenously. Tumors were measured twice weekly with calipers and the tumor area calculated as length×width. The change of tumor area was determined by subtracting the tumor area before CAR T injection with the value on the day of the measurement. Per institutional guidelines, tumor-bearing mice were sacrificed when the tumor area reached 200 mm2 or loss of more than 20% of total body weight. For tumor-bearing animals receiving combination therapy, animals underwent lymphodepletion with 5 Gy of whole body irradiation (X-RAD 320, Precision X-Ray) one day prior to CAR T injection. In the morning of day 0 mice were injected with 500 μg of DMXAA (Tocris) distal to the site of tumor growth with CAR T cells injected in the afternoon. Starting 1 day after CAR T infusion, anti-PD-1 (J43, 200 μg/mouse), anti-Gr-1 (RB6-8C5, 300 ag/mouse), and anti-IFN-7 (R4-6A2, 250 μg/mouse) mAb were injected intraperitoneally into respective recipients.
At indicated time points, tumors were bluntly resected and cut into small pieces and processed into a single-cell suspension using gentleMACS™ Dissociator (Miltenyi). Lymphocyte enrichment was achieved by isolating cells at the interface of 44% Percoll (Sigma-Aldrich) in media and Lympholyte-M (Cedarlane). Isolated immune cells were stained as described below. The lymphocyte fraction was not enriched in experiments where absolute cell number was determined.
At day 7 or day 10 after CAR-T therapy, tumors were bluntly resected and cut into small pieces and digested using Tumor Dissociation Kit, mouse (Miltenyi) in gentleMACS™ Dissociator (Miltenyi) according to manufacturer's instruction. After digestion, dead and dying cells were magnetically separated using a Dead Cell Removal Kit (Miltenyi), after which immune cells were magnetically enriched using CD45 (TIL) MicroBeads (Miltenyi). Single cells were partitioned using a Chromium Controller (10× Genomics), and gene expression sequencing libraries were generated using Chromium Single Cell 3′ Library & Gel Bead Kit v3 (10× Genomics). The libraries were pooled and sequenced using an Illumina NovaSeq 6000 (Illumina). Raw base call files were demultiplexed with Cell Ranger 3.1.0 (10× Genomics) and bcl2fastq Conversion Software v2.20.0 (Illumina).
Single cell results from Cell Ranger output was analyzed using Partek flow workstation (available at partek.com/application-page/single-cell-gene-expression/). In detail, single cell result from each sample of Cell Ranger output was filtered using single cell (QA/QC) filter to exclude doublets/multiplets, cells expressing genes <30 genes per cell and dead cells. Processed results were normalized using recommended method, including log normalization of gene expression and scaled to count per million. Following results were filtered out by feature (gene) that were not expressed in 99% of samples. Then the replicate samples were pooled and clustered with an unsupervised method based on PCA using the first 16 principle component. Clustered plots were visualized by t-SNE plot, where cells were clustered using shared nearest neighbor (SNN). Populations was determined by expression of key markers, including Cd3e (T cells), Adgre1 (macrophage) and Itgam and/or Itgax (myeloid cells), and T cells were further clustered by expression of Cd8 and Cd44 (activated CD8 T cells), lack of Cd44 (naive T cells), Cd4 Cd44 (activated CD4 T cells) and Cd4 Foxp3 (Treg). For comparison, samples from each treatment group was pooled by the mean expression to simulate bulk RNA-seq, and compared using GSA analysis directly. Gene hits that reached P<0.05 with more than 2 or less than −2 fold expression difference was reported and shown as a heat map for pooled samples (macrophage and myeloid cells) or Q<0.05 with 2 or −2 fold expression difference for non-pooled samples (T cells). Further detailed analysis steps can be found on Partek flow page (available at documentation.partek.com/pages/viewpage.action?pageId=12943422) and parameter setting was set as default unless indicated.
Anti-Neu CAR T cells were detected using recombinant Neu/Her2.Fc fusion protein conjugated with PE (Neu-PE) (Creative Biomart). Antibodies used in in vitro assays included anti-CD45-FITC/e450 (30-F11, Invitrogen), anti-IFN-γ-APC (XMG1.2, Invitrogen), anti-TNF-PE-Cy7 (MP6-XT22, Invitrogen) and anti-IL-17A-PE (eBio17B7, Invitrogen). Tumor-infiltrating immune cells were detected and analyzed with anti-CD45-FITC, anti-CD3-e450 (17A2, Invitrogen), anti-CD4-BV510 (GKL.5, Biolegend), anti-CD8-APC-Cy7 (53-6.7 BD Biosciences), anti-CD44-PerCP5.5 (IM7, Invitrogen), anti-CD62L-APC (MEL-14, Invitrogen), anti-CD11b-BV711 (M1/70, Invitrogen), anti-F4/80-BV605 (BM8, Invitrogen), anti-Gr-1-Percp5.5 (RB6-8C5, Invitrogen), anti-PD-1-PE-Cy7 (J43, Invitrogen), anti-CD127-BV421 (A7R34, Biolegend), anti-KLGR1-BV605 (2F1, BD Bioscience). For in vivo intracellular protein staining, anti-Foxp3-PerCP5.5 (FJK-16S. Invitrogen), anti-Ki67-APC (SOLA15, Invitrogen) were used. Dead cells were excluded with Fixable Viability Stain 700 (BD Biosciences) and Fc receptors were blocked with Fc block (BD Biosciences). Cell number was determined using Count Bright Beads (Invitrogen). Data were collected using a BD LSR-Fortessa or BD Canto cytometer (BD Biosciences). Data were analyzed with FlowJo software (Treestar).
For co-culture killing assays, 106 NT2 tumor cells were labeled with 5 μM CFSE (Invitrogen) and plated in culture media for 24 hours. CAR-T, mock transfected T cells or no T cells were added the next day at the indicated ratios and co-cultured in T cell media without cytokines for three days. Supernatant and trypsinized cells were collected and stained with 7-AAD (BD Biosciences) and anti-CD45 (30-F11, Invitrogen). Killing was measured by flow cytometry after gating on CFSE+, 7-AAD− and CD45− tumor cells and normalized to groups without T cell addition. For killing curve, NT2 tumor cells were labeled with CFSE and plated at 105 cells/well in a 48 well plate. CAR T cells, mock T cells or no T cells at the indicated ratios were added 12 hours later. Co-cultures were maintained in T cell media without cytokine for 18 hours and cell killing was measured as described above. Percent tumor lysis was calculated by subtracting the number of live tumor cells from 100% and dividing by the total number of live tumor cells in the group with no T cells added.
5×105 3T3 or 3T3-Neu cells were treated with trypsin and processed into a single cell suspension prior to culture with CAR T or mock T cells at indicated ratios in T cell media without additional cytokine. 5 μg/ml of Brefeldin A (BD Biosciences) was added after the first hour co-culture. Cells were stimulated for 5 hours and then pelleted for extracellular staining with anti-CD45, followed by fixation and permeabilization using Fix/Perm Kit (BD Biosciences) according to the manufacturer's instructions. Anti-IFN-7, anti-TNF and anti-IL-17A were used for intracellular cytokine staining and detected by flow cytometry.
Whole tumor RNA was isolated using the RNEasy Kit (Qiagen) and reverse transcribed using RT2 First Strand Kit (Qiagen). RT-qPCR was performed according to the manufacturer's instruction with the RT2 Profiler™ PCR Array Mouse Th17 Response (PAMM-073ZE-4, Qiagen) and performed on the QuantStudio 6K (Applied Biosystems Inc.). Ct values were determined by ABI software and data analysis was performed using the web-based RT2 Profiler PCR Array Data Analysis version 3.5 (Qiagen). Fold change was normalized to a housekeeping gene (Gusb).
This application claims the benefit of U.S. Provisional Application No. 63/069,649, filed on Aug. 24, 2020, and U.S. Provisional Application No. 62/932,913, filed on Nov. 8, 2019. The entire teachings of the above applications are incorporated herein by reference.
This invention was made with government support under Grant Nos. CA058223 and BC010763 awarded by the National Institutes of Health. The government has certain rights in the invention.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2020/059751 | 11/9/2020 | WO |
Number | Date | Country | |
---|---|---|---|
63069649 | Aug 2020 | US | |
62932913 | Nov 2019 | US |