WO2023225181A1 - Tumor infiltrating lymphocytes and anti-pd-1 antibody combination therapy for the treatment of cancer - Google Patents

Tumor infiltrating lymphocytes and anti-pd-1 antibody combination therapy for the treatment of cancer Download PDF

Info

Publication number
WO2023225181A1
WO2023225181A1 PCT/US2023/022712 US2023022712W WO2023225181A1 WO 2023225181 A1 WO2023225181 A1 WO 2023225181A1 US 2023022712 W US2023022712 W US 2023022712W WO 2023225181 A1 WO2023225181 A1 WO 2023225181A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
tils
antibody
cells
population
Prior art date
Application number
PCT/US2023/022712
Other languages
French (fr)
Inventor
Zachary Roberts
Original Assignee
Instil Bio, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Instil Bio, Inc. filed Critical Instil Bio, Inc.
Publication of WO2023225181A1 publication Critical patent/WO2023225181A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the subject matter disclosed herein relates to methods of enhancing an anti-tumor response by administering combinations comprising a population of tumor infiltrating lymphocyte (TILs) and an anti-PD-1 antibody to tumor cells in a patient with a cancer BACKGROUND
  • TILs tumor infiltrating lymphocyte
  • the immune system plays a role in the pathogenesis of a many cancers. When cancers progress, the immune system either fails to respond sufficiently or fails to respond appropriately, allowing cancer cells to grow. Improved strategies that combine specific manipulation of the immune response to cancer in combination with standard medical treatments may provide a means for enhanced efficacy and decreased toxicity.
  • antitumor immunity is often ineffective due to the tight regulation associated with the maintenance of immune homeostasis.
  • T-cell exhaustion results from chronic exposure to antigens and is characterized by the upregulation of inhibitory receptors. These inhibitory receptors serve as immune checkpoints in order to prevent uncontrolled immune reactions. Blocking of one or several of these immune checkpoints with monoclonal antibodies (mAbs) has been shown to rescue otherwise exhausted antitumor T cells, and most importantly, has been associated with objective clinical responses in cancer patients.
  • mAbs monoclonal antibodies
  • the subject matter described herein is directed to a method of treating a cancer comprising administering to a patient a therapeutically effective amount of tumor infiltrating lymphocytes (TILs) in combination with an antibody that specifically binds to human Programmed Death (PD)-1, also referred herein as an anti-PD-1 antibody.
  • TILs tumor infiltrating lymphocytes
  • PD programmed Death
  • the subject matter described herein is directed to a method of treating a subject having a cancerous tumor, the method comprising: a) administering an effective dose of an anti-PD-1 antibody; and b) administering an effective dose of a population of TILs to the subject.
  • the method further comprises administering a cytokine, such as IL-2, to the subject.
  • a cytokine such as IL-2
  • the cancer is advanced.
  • the cancer is a type of cancer selected from the group consisting of cervical cancer, head and neck squamous-cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC).
  • HNSCC head and neck squamous-cell carcinoma
  • NSCLC non-small cell lung cancer
  • the cancer has progressed after the subject has received standard therapy for the type of cancer.
  • the subject has relapsed after standard therapy for the type of cancer.
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, pidilizumab, durvalumab, atezolizumab, avelumab, cemiplimab, sintilimab, toripalimab, and camrelizumab.
  • administration of the combination results in enhanced therapeutic efficacy relative to administration of the TILs or the anti-PD-1 antibody alone.
  • the population of TILs may be administered prior to, at the same time as, or following administration of the anti-PD-1 antibody. [017]
  • FIG. 1 depicts a protocol for certain embodiments of the combination therapies described herein.
  • FIG. 2 shows graphed outgrowth (days 1-13) total viable count (TVC) for ITIL-168 full-scale cervical runs.
  • FIG.3 shows graphed REP (days 13-25) TVC for ITIL-168 full-scale cervical runs.
  • FIG.4 shows graphed CD3+ cell counts throughout ITIL-168 full-scale cervical runs.
  • FIG.5 shows CD2 phenotype results for ITIL-168 full-scale cervical runs.
  • FIG.6 shows CD4/CD8 phenotype results for ITIL-168 full-scale cervical runs.
  • FIG.7 shows leukocyte data for ITIL-168 full-scale cervical runs.
  • FIG.8 shows potency results for ITIL-168 full-scale cervical runs.
  • FIGS.9A and 9B show outgrowth cell growth and REP cell growth, respectively for cSCC and cervical cancer indications (NC-200).
  • FIGS. 10A and 10B show total CD3+ cells over time for cSCC and cervical cancer indications, respectively, as measured by flow cytometry.
  • FIG. 11 shows CD3+ purity for both cSCC and cervical indications as measured by flow cytometry.
  • FIG.12 shows final product leukocyte data (T cells, NK cells, and monocytes; cSCC and cervical cancer indications).
  • FIG. 13 shows final product phenotype data (CD2+; cSCC and cervical cancer indications).
  • FIG.14 shows final product phenotype data (CD4+/CD8+/DP/DN; cSCC and cervical cancer indications).
  • FIG.15 shows final product potency data (cSCC and cervical cancer indications).
  • FIGS. 16A and 16B show outgrowth fold expansion and total viable CD3+ yield, respectively, for melanoma, NSCLC, cervical, and HNSCC indications.
  • FIGS. 17A and 17B show REP fold expansion and total viable CD3+ yield, respectively, for melanoma, NSCLC, cervical, and HNSCC indications.
  • FIGS. 17A and 17B show REP fold expansion and total viable CD3+ yield, respectively, for melanoma, NSCLC, cervical, and HNSCC indications.
  • FIGS. 19A-19C show post-thaw drug product purity, viability, and potency for TILs from NSCLC and HNSCC indications.
  • FIG. 20 depicts a protocol for certain embodiments of the combination therapies described herein. HD, high-dose; IL-2, interleukin-2.
  • the subject matter is directed to a method of treating a cancer comprising administering to a patient a therapeutically effective amount of tumor infiltrating lymphocytes (TILs) in combination with an antibody that specifically binds to human Programmed Death (PD)-1, also referred herein as an anti-PD-1 antibody.
  • TILs tumor infiltrating lymphocytes
  • PD programmed Death
  • a clinical trial will evaluate the safety, feasibility, and preliminary efficacy of TILs in combination with pembrolizumab in participants with advanced cancer whose disease has progressed after standard therapy.
  • the population of TILs to be administered is a cell therapy derived from a subject’s own tumor-infiltrating immune cells (lymphocytes; TILs).
  • TILs in combination with pembrolizumab do not provide any improvement compared to the use of individual components alone, the presently described methods have the potential to improve the effectiveness of a combination therapy and may provide synergistic results.
  • the subject matter is also directed to treating subjects who although having undergone previous cancer therapy, the cancer subject’s cancer has progressed. [039] All references cited herein are incorporated by reference to the same extent as if each individual publication, patent application, or patent, was specifically and individually indicated to be incorporated by reference. [040] A “population of TILs” and “TIL product” and the like refer to an expanded population of tumor infiltrating lymphocytes derived from the subject’s tumor.
  • the TIL product can be manufactured by the methods described in WO 2021/123832 and WO 2018/130845, each of which is incorporated by reference in its entirety.
  • the term “population of cells” can mean a number of cells that share some common traits. In general, populations generally range from 1 x 10 6 to 1 x 10 12 in number, with different TIL populations comprising different numbers.
  • tumor infiltrating lymphocytes or TILs herein is meant a population of cells originally obtained as white blood cells that have left the bloodstream of a subject and migrated into a tumor.
  • TILs include, but are not limited to, CD8 + cytotoxic T cells (lymphocytes), Thi and Thi 7 CD4 + T cells, natural killer cells, dendritic cells, and Ml macrophages.
  • TILs include both primary and secondary TILs. “Primary TILs” are those that are obtained from patient tissue samples as outlined herein (sometimes referred to as “freshly harvested”), and “secondary TILs” are any TIL cell populations that have been expanded or proliferated as discussed herein, including, but not limited to bulk TILs and expanded TILs (“REP TILs” or “post-REP TILs”). TIL cell populations can include genetically modified TILs.
  • TILs can generally be defined either biochemically, using cell surface markers, or functionally, by their ability to infiltrate tumors and effect treatment.
  • TILs can be generally categorized by expressing one or more of the following biomarkers: CD4, CD8, TCR ab, CD27, CD28, CD56, CCR7, CD45Ra, CD62L, CD95, PD-1, and CD25. Additionally and alternatively, TILs can be functionally defined by their ability to infiltrate solid tumors upon reintroduction into a patient.
  • TILs may further be characterized by potency— for example, TILs may be considered potent or functional if in response to TCR engagement they produce, for example, interferon (IFN) release is greater than about 50 pg/mL, greater than about 100 pg/mL, greater than about 150 pg/mL, or greater than about 200 pg/mL, or individual cells can be considered potent through intracellular staining for CD 137, CD 107a, INF-g TNF-a, and IL-2 following TCR induced stimulation by flow cytometry.
  • IFN interferon
  • the TIL product will have been assayed for the presence of and will contain T-cells expressing a combination of markers, the combination comprising two or more of: CD107a; CD137; TNF- ⁇ ; or IFN- ⁇ .
  • the combination comprises: CD107a and CD137; or CD107a and TNF- ⁇ ; or CD107a and IFN- ⁇ ; or CD137 and TNF- ⁇ ; or CD137 and IFN- ⁇ ; or TNF- ⁇ and IFN- ⁇ .
  • the combination comprises three or more of: CD107a; CD137; TNF- ⁇ ; and IFN- ⁇ .
  • the combination comprises: CD107a, CD137, and TNF- ⁇ ; or CD107a, CD137, and IFN- ⁇ ; or CD107a, TNF- ⁇ , and IFN- ⁇ ; or CD137, TNF- ⁇ , and IFN- ⁇ . In certain aspects of these embodiments, the combination comprises each of CD107a, CD137, TNF- ⁇ , and IFN- ⁇ .
  • the TIL product will have been assayed for the presence of and will contain CD2+ T-cells expressing a combination of markers, the combination comprising two or more of: a T cell expressing CD107a; a T cell expressing CD137; a T cell expressing TNF ⁇ ; or a T-cell expressing IFN- ⁇ .
  • the TIL product will have been assayed for the presence of and will contain CD3+ T-cells expressing a combination of markers, the combination comprising two or more of: a T-cell expressing CD107a; a T-cell expressing CD137; a T-cell expressing TNF- ⁇ ; or a T-cell expressing IFN- ⁇ .
  • the combination comprises: CD107a and CD137, or CD107a and TNF- ⁇ , or CD107a and IFN- ⁇ , or CD137 and TNF- ⁇ , or CD137 and IFN- ⁇ , or TNF- ⁇ and IFN- ⁇ . In certain embodiments, the combination comprises three or more of: CD107a; CD137; TNF- ⁇ ; and IFN- ⁇ . In certain aspects of these embodiments, the combination comprises: CD107a, CD137 and TNF- ⁇ ; or CD107a, CD137 and IFN- ⁇ ; or CD107a, TNF- ⁇ and IFN- ⁇ ; or CD137, TNF- ⁇ and IFN- ⁇ . In certain embodiments, the combination comprises each of CD107a, CD137, TNF- ⁇ and IFN- ⁇ .
  • TILs in particular unmodified TILs (UTILs) are an autologous product; consequently, each batch manufactured provides a single dose for a specified patient. There are no sub-batches or pooling of batches.
  • the drug product is a small aseptically prepared batch of T cells (5x10 9 to 5x10 10 ) cryopreserved in a saline based solution with 8.5% human serum albumin and 10% DMSO of between 125-270 mL for a single intravenous infusion after thawing. The number of cells present is dependent on the ability of each individual’s TIL cells to be expanded in culture in conjunction with the culture conditions and the manufacturing reproducibility.
  • anti-PD-1 antibodies as described herein are part of a combination therapy for improving an individual's immune response to cancer (e.g., a target cancer antigen or antigens) by co-administering a TIL product, an antibody which specifically binds to human Programmed Death (PD)-1, and optionally a cytokine.
  • PD-1 is an immunoinhibitory receptor belonging to the CD28 family (Freeman et al., J. Exp. Med.192: 1027 (2000); Okazaki et al., Curr. Opin. Immunol.14: 779 (2002)) and binds to two ligands, PD-L1 and PD-L2.
  • PD-1 is induced on T-cells, B-cells and myeloid cells in vitro (Agata et al., Int. Immunol.8:765 (1996)), but is predominantly expressed on previously activated T-cells in vivo (Iwai et al., Immunol. Lett.83:215 (2002)).
  • Exemplary anti-PD-1 antibodies include, but are not limited to, one or more of nivolumab, pembrolizumab, pidilizumab, durvalumab, atezolizumab, avelumab, cemiplimab, sintilimab, toripalimab, and camrelizumab.
  • the antibodies for use in the present methods include, but are not limited to, monoclonal antibodies, synthetic antibodies, polyclonal antibodies, multispecific antibodies (including bi-specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain Fvs (scFv) (including bi-specific scFvs), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked Fvs (sdFv), and epitope-binding fragments of any of the above.
  • antibodies for use in the present methods include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain a PD- 1 binding site that immunospecifically binds to PD-1.
  • the immunoglobulin molecules for use in the present methods can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule.
  • the antibodies for use in the invention are IgG, such as IgG1.
  • the antibodies for use in the present methods may be from any animal origin including birds and mammals (e.g., human, murine, donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken).
  • the antibodies are human or humanized monoclonal antibodies.
  • human antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from mice or other animals that express antibodies from human genes.
  • the antibodies for use in the present methods include derivatives of any of the antibodies described herein. Standard techniques known to those of skill in the art can be used to introduce mutations in the nucleotide sequence encoding an antibody to be used with the methods for use in the invention, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis which result in amino acid substitutions.
  • the derivatives include less than 25 amino acid substitutions, less than 20 amino acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the original molecule.
  • the derivatives have conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues.
  • a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a side chain with a similar charge. Families of amino acid residues having side chains with similar charges have been defined in the art.
  • 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
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed and the activity of the protein can be determined.
  • the antibodies for use in the present methods include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the antibody.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, synthesis in the presence of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids. [050] The present methods also provide antibodies for use in the methods that comprise a framework region known to those of skill in the art.
  • one or more framework regions, for example all of the framework regions, of an antibody to be used in the compositions and methods for use are human.
  • the fragment region of an antibody for use in the methods is humanized.
  • the antibody to be used with the present methods is a synthetic antibody, a monoclonal antibody, an intrabody, a chimeric antibody, a human antibody, a humanized chimeric antibody, a humanized antibody, a glycosylated antibody, a multispecific antibody, a human antibody, a single-chain antibody, or a bispecific antibody.
  • an antibody for use in the invention has a high binding affinity for PD-1.
  • an antibody for use in the invention has an association rate constant or k on , rate of about 10 5 M -1 s -1 or more, about 5 ⁇ 10 5 M -1 s -1 or more, about 10 6 M -1 s -1 or more, about 5 10 M s or more, about 10 M s or more, about 5 10 M s or more, about 10 8 M -1 s -1 or more, about 5 ⁇ 10 8 M -1 s -1 or more, or about 1 ⁇ 10 9 M -1 s -1 or more.
  • an antibody for use in the invention has a koff rate for PD-1 of about 5 ⁇ 10 ⁇ 1 s -1 or less, about 10 ⁇ 1 s -1 or less, about 5 ⁇ 10 ⁇ 12 s -1 or less, about 10 ⁇ 2 s -1 or less, about 5 ⁇ 10 ⁇ 3 s -1 or less, about 10 ⁇ 3 s -1 or less, about 5 ⁇ 10 ⁇ 4 s -1 or less, about 10 ⁇ 4 s -1 or less, about 5 ⁇ 10 ⁇ 5 s -1 or less, about 10 ⁇ 5 s -1 or less, about 5 ⁇ 10 ⁇ 6 s -1 or less, about 10 ⁇ 6 s -1 or less, about 5 ⁇ 10 ⁇ 7 s -1 or less, about 10 ⁇ 7 s -1 or less, about 5 ⁇ 10 ⁇ 8 s -1 or less, about 10 ⁇ 8 s -1 or less, about 5 ⁇ 10 ⁇ 9 s -1 or less, about 10 ⁇ 9
  • an antibody for use in the methods has an affinity constant or K a (k on /k off ) for PD-1 of about 10 2 M -1 or more, about 5 ⁇ 10 2 M -1 or more, about 10 3 M -1 or more, about 5 ⁇ 10 3 M -1 or more, about 10 4 M -1 or more, about 5 ⁇ 10 4 M -1 or more, about 10 5 M -1 or more, about 5 ⁇ 10 5 M -1 or more, about 10 6 M -1 or more, about 5 ⁇ 10 6 M -1 or more, about 10 7 M -1 or more, about 5 ⁇ 10 7 M -1 or more, about 10 8 M -1 or more, about 5 ⁇ 10 8 M -1 or more, about 10 9 M -1 or more, about 5 ⁇ 10 9 M -1 or more, about 10 10 M -1 or more, about 5 ⁇ 10 10 M -1 or more, about 10 11 M -1 or more, about 5 ⁇ 10 11 M -1 or more, about 10 12 M -1 or more, about 5 ⁇ 10 12 M -1
  • an antibody for use in the methods has a low dissociation constant.
  • the antibody-binding domain of a carrier construct for use in the methods has a dissociation constant or K d (k off /k on ) for antibody about 5 ⁇ 10 ⁇ 1 M or less, about 10 ⁇ 1 M or less, about 5 ⁇ 10 ⁇ 2 M or less, about 10 ⁇ 2 M or less, about 5 ⁇ 10 ⁇ 3 M or less, about 10 ⁇ 3 M or less, about 5 ⁇ 10 ⁇ 4 M or less, about 10 ⁇ 4 M or less, about 5 ⁇ 10 ⁇ 5 M or less, about 10 ⁇ 5 M or less, about 5 ⁇ 10 ⁇ 6 M or less, about 10 ⁇ 6 M or less, about 5 ⁇ 10 ⁇ 7 M or less, about 10 ⁇ 7 M or less, about 5 ⁇ 10 ⁇ 8 M or less, about 10 ⁇ 8 M or less, about 5 ⁇ 10 ⁇ 9 M or less, about 10 ⁇ 9 M or less, about 5 ⁇ 10 ⁇ 10
  • an antibody for use in the present methods has a median effective concentration (EC 50 ) of less than 0.01 nM, less than 0.025 nM, less than 0.05 nM, less than 0.1 nM, less than 0.25 nM, less than 0.5 nM, less than 0.75 nM, less than 1 nM, less than 1.25 nM, less than 1.5 nM, less than 1.75 nM, or less than 2 nM, in an in vitro microneutralization assay.
  • the median effective concentration is the concentration of antibody that neutralizes 50% of PD-1 in an in vitro microneutralization assay.
  • cytokine and immunostimulatory cytokine includes cytokines that mediate or enhance the immune response to a foreign antigen, including viral, bacterial, or tumor antigens.
  • Innate immunostimulatory cytokines can include, e.g., TNF-a, IL-1, IL-10, IL-12, IL-15, type I interferons (IFN- ⁇ and IFN- ⁇ ), IFN- ⁇ , and chemokines.
  • Adaptive immunostimulatory cytokines include, e.g., IL-2, IL-4, IL-5, TGF- ⁇ , IL-10 and IFN- ⁇ .
  • Cytokines and combinations of cytokines have been shown to play an important role in the stimulation of the immune system.
  • the term “cytokine” is understood by those of skill in the art, as referring to any immunopotentiating protein (including a modified protein such as a glycoprotein) that enhances or modifies the immune response to a tumor present in the host.
  • the cytokine typically enhances or modifies the immune response by activating or enhancing the activity of cells of the immune system and is not itself immunogenic to the host.
  • a variety of cytokines will find use in the methods described herein.
  • Exemplary cytokines for use in practicing the invention include but are not limited to interferon- alpha (IFN- ⁇ ), interferon-beta (IFN- ⁇ ), and interferon-gamma (IFN- ⁇ ), interleukins (e.g., IL-1 to IL-29, in particular, IL-2, IL-7, IL-12, IL-15 and IL-18), tumor necrosis factors (e.g., TNF-alpha and TNF-beta), erythropoietin (EPO), MIP3a, intracellular adhesion molecule (ICAM), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF).
  • IFN- ⁇ interferon- alpha
  • IFN- ⁇ interferon-beta
  • IFN-gamma IFN- ⁇
  • interleukins e.g., IL-1 to
  • the cytokine may be from any source, however, optimally the cytokine is of murine or human origin (a native human or murine cytokine) or is a sequence variant of such a cytokine, so long as the cytokine has a sequence with substantial homology to the human form of the cytokine and exhibits a similar activity on the immune system.
  • cytokines with substantial homology to the human forms of IFN- alpha, IFN-beta, and IFN-gamma, IL-1 to IL-29, TNF-alpha, TNF-beta, EPO, MIP3a, ICAM, M- CSF, G-CSF and GM-CSF are useful in practicing the invention, so long as the homologous form exhibits the same or a similar effect on the immune system. Proteins that are substantially similar to any particular cytokine, but have relatively minor changes in protein sequence find use in the present invention.
  • IL-2 refers to the T cell growth factor known as interleukin-2, and includes all forms of IL-2 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof.
  • IL-2 is described, e.g., in Nelson, J. Immunol.2004, 172, 398388 and Malek, Annu. Rev. Immunol.2008, 26, 453-79, the disclosures of which are incorporated by reference herein.
  • IL-2 encompasses human, recombinant forms of IL-2 such as aldesleukin (PROLEUKIN, available commercially from multiple suppliers in 22 million IU per single use vials), as well as the form of recombinant IL-2 commercially supplied by CellGenix, Inc., Portsmouth, N.H., USA (CELLGRO GMP) or ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-209-b) and other commercial equivalents from other vendors.
  • aldesleukin PROLEUKIN, available commercially from multiple suppliers in 22 million IU per single use vials
  • CELLGRO GMP CellGenix, Inc.
  • ProSpec-Tany TechnoGene Ltd. East Brunswick, N.J., USA
  • Aldesleukin (des-alanyl-1, serine- 125 human IL-2) is a nonglycosylated human recombinant form of IL-2 with a molecular weight of approximately 15 kDa.
  • the term IL-2 also encompasses pegylated forms of IL-2, as described herein, including the pegylated IL2 prodrug NKTR-214, available from Nektar Therapeutics, South San Francisco, Calif., USA.
  • NKTR-214 and pegylated IL-2 suitable for use in the invention is described in U.S. Patent Application Publication No. US 2014/0328791 A1 and International Patent Application Publication No. WO 2012/065086 Al.
  • IL-2 suitable for use in the invention are described in U.S. Pat. Nos. 4,766,106, 5,206,344, 5,089,261 and 4902,502.
  • Formulations of IL-2 suitable for use in the invention are described in U.S. Pat. No. 6,706,289.
  • IL-2 (such as variants of IL-2) can encompasses modified IL-2.
  • modified IL-2 can mediate potent IL-2 R ⁇ stimulation of CD8 effector T and NK cells, yet reduce IL-2 R ⁇ binding to mitigate Treg-mediated suppression and off-target toxicity.
  • Modified IL-2 can exhibit, for example, similar or enhanced affinity and/or activation of the IL-2 R ⁇ and ⁇ subunits as compared to a non-modified IL-2. In some embodiments, modified IL-2 can exhibit similar or decreased affinity and/or engagement of the IL-2R ⁇ subunit, as compared to a non-modified IL-2.
  • IL-4 also referred to herein as “IL4” refers to the cytokine known as interleukin 4, which is produced by Th2 T cells and by eosinophils, basophils, and mast cells. IL- 4 regulates the differentiation of naive helper T cells (ThO cells) to Th2 T cells. Steinke and Borish, Respir.
  • IL-4 IL-4 also stimulates B cell proliferation and class II MHC expression, and induces class switching to IgE and IgGl expression from B cells.
  • Recombinant human IL-4 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-211) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL 15 recombinant protein, Cat. No. Gibco CTP0043).
  • IL-7 refers to a glycosylated tissue- derived cytokine known as interleukin 7, which may be obtained from stromal and epithelial cells, as well as from dendritic cells. Fry and Mackall, Blood 2002, 99, 3892-904. IL-7 can stimulate the development of T cells. IL-7 binds to the IL-7 receptor, a heterodimer consisting of IL-7 receptor alpha and common gamma chain receptor, which in a series of signals important for T cell development within the thymus and survival within the periphery.
  • IL-7 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-254) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No. Gibco PHC0071).
  • IL-12 also referred to herein as “IL12” refers to the T cell growth factor known as interleukin- 12.
  • Interleukin (IL)-12 is a secreted heterodimeric cytokine comprised of 2 disulfide- linked glycosylated protein subunits, designated p35 and p40 for their approximate molecular weights.
  • IL-12 is produced primarily by antigen-presenting cells and drives cell- mediated immunity by binding to a two-chain receptor complex that is expressed on the surface of T cells or natural killer (NK) cells.
  • the IL-12 receptor beta-1 (IL-12Rpi) chain binds to the p40 subunit of IL-12, providing the primary interaction between IL-12 and its receptor. However, it is IL-12p35 ligation of the second receptor chain, IL-12RP2, that confers intracellular signaling.
  • IL- 12 signaling concurrent with antigen presentation is thought to invoke T cell differentiation towards the T helper 1 (Thl) phenotype, characterized by interferon gamma (IFNy) production.
  • Thl cells are believed to promote immunity to some intracellular pathogens, generate complement fixing antibody isotypes, and contribute to tumor immunosurveillance.
  • IL-12 is thought to be a significant component to host defense immune mechanisms.
  • IL-12 is part of the IL-12 family of cytokines which also includes IL-23, IL-27, IL-35, IL-39.
  • IL-15 refers to the T cell growth factor known as interleukin- 15, and includes all forms of IL-15 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof.
  • IL-15 is described, e.g., in Fehniger and Caligiuri, Blood 2001, 97, 14-32, the disclosure of which is incorporated by reference herein.
  • IL-15 shares b and g signaling receptor subunits with IL-2.
  • Recombinant human IL 15 is a single, non glycosylated polypeptide chain containing 114 amino acids (and an N-terminal methionine) with a molecular mass of 12.8 kDa.
  • Recombinant human IL-15 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-230-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No.34-8159-82).
  • the term “IL-18” (also referred to herein as “IL18”) refers to the T cell growth factor known as interleukin- 15.
  • Interleukin- 18 is a proinflammatory cytokine that belongs to the IL-1 cytokine family, due to its structure, receptor family and signal transduction pathways. Related cytokines include IL-36, IL-37, IL-38.
  • IL-21 refers to the pleiotropic cytokine protein known as interleukin-21, and includes all forms of IL-21 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-21 is described, e.g., in Spolski and Leonard, Nat. Rev. Drug.
  • IL-21 is primarily produced by natural killer T cells and activated human CD4 + T cells.
  • Recombinant human IL-21 is a single, non-glycosylated polypeptide chain containing 132 amino acids with a molecular mass of 15.4 kDa.
  • Recombinant human IL-21 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-408-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-21 recombinant protein, Cat. No.14-8219-80).
  • the term “subject” refers to any animal, such as, but not limited to, a mammal such as a human. In certain embodiments, the term “subject” refers to a patient that has undergone prior cancer treatment. In certain embodiments, the term “subject” refers to a patient whose cancer has progressed after standard therapy. Among these groups of subjects are patients who have a cancer that is susceptible to TIL therapy. Among these groups of subjects are patients who have cervical cancer, HNSCC or NSCLC. The subject will have undergone a resection of one or tumors in the subject to obtain a sample from which the TIL product will be developed. In certain embodiments, the subject will have undergone a prior treatment for the cancer.
  • cancer includes a myriad of diseases generally characterized by inappropriate cellular proliferation, abnormal or excessive cellular proliferation.
  • diseases include but are not limited to, breast cancer, cervical cancer, colon cancer, head and neck squamous cell carcinoma (HNSCC), prostate cancer, pancreatic cancer, melanoma, lung cancer including non small cell lung cancer (NSCLC), ovarian cancer, kidney cancer, brain cancer, or sarcomas.
  • HNSCC head and neck squamous cell carcinoma
  • NSCLC non small cell lung cancer
  • ovarian cancer ovarian cancer
  • kidney cancer kidney cancer
  • brain cancer or sarcomas.
  • sarcomas may be caused by one or more factors, including but not limited to environmental factors, chromosomal abnormalities, degenerative growth and developmental disorders, mitogenic agents, ultraviolet radiation (UV), viral infections, inappropriate tissue expression of a gene, alterations in expression of a gene, or carcinogenic agents.
  • UV ultraviolet radiation
  • an anti-tumor effective amount “a tumor-inhibiting effective amount”, “therapeutically effective amount,” or “therapeutic amount”
  • 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). It can generally be stated that a pharmaceutical composition comprising the tumor infiltrating lymphocytes (e.g.
  • secondary TILs or genetically modified cytotoxic lymphocytes described herein may be administered at a dosage of 10 4 to 10 11 cells/kg body weight (e.g., 10 5 to 10 6 , 10 5 to 10 10 , 10 5 to 10 11 , 10 6 to 10 10 , 10 6 to 10 11 , 10 7 to 10 11 , 10 7 to 10 10 , 10 8 to 10 11 , 10 8 to 10 10 , 10 9 to 10 11 , or 10 9 to 10 10 cells/kg body weight), including all integer values within those ranges.
  • Tumor infiltrating lymphocytes (including in some cases, genetically modified cytotoxic lymphocytes) compositions may also be administered multiple times at these dosages.
  • the tumor infiltrating lymphocytes can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et ah, New Eng. J. of Med.319: 1676, 1988).
  • the optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly.
  • the term “therapeutically effective amount” or “therapeutically effective combination” as used herein can refer to an amount or dose of a TIL product together with the amount or dose of an anti-PD-1 antibody, and optionally a cytokine that is sufficient for treatment.
  • the amount of each agent in a given therapeutically effective combination may be different for different individuals and different tumor types, and will be dependent upon the one or more additional agents or treatments included in the combination.
  • the “therapeutically effective amount” is determined using procedures routinely employed by those of skill in the art such that an “improved therapeutic outcome” results.
  • a combination therapy disclosed herein can provide synergistic effects whereby the effective amount of any one agent can be lower in the combination than the effective amount of the agent if given alone to achieve the same effect.
  • the term “synergism,” “synergistic” or “synergistically” refers to the combined action of two or more agents wherein the combined action is greater than the sum of the actions of each of the agents used alone.
  • any suitable dose of TILs can be administered.
  • from about 2.3x10 10 to about 13.7x10 10 TILs are administered, with an average of around 7.8x10 10 TILs.
  • about 1.2x10 10 to about 4.3x10 10 of TILs are administered.
  • about 3x10 10 to about 12x10 10 TILs are administered.
  • about 4x10 10 to about 10x10 10 TILs are administered.
  • about 5x10 10 to about 8x10 10 TILs are administered.
  • about 6x10 10 to about 8x10 10 TILs are administered.
  • about 7x10 10 to about 8x10 10 TILs are administered.
  • the therapeutically effective dosage is about 2.3x10 10 to about 13.7x10 10 . In some embodiments, the therapeutically effective dosage is about 7.8x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 1.2x10 10 to about 4.3x10 10 of TILs. In some embodiments, the therapeutically effective dosage is about 3x10 10 to about 12x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 4x10 10 to about 10x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 5x10 10 to about 8x10 10 TILs. In some embodiments, the therapeutically effective dosage is about 6x10 10 to about 8x10 10 TILs.
  • the therapeutically effective dosage is about 7x10 10 to about 8x10 10 TILs.
  • the number of the TILs provided in the pharmaceutical compositions of the invention is about any one of: 1x10 6 , 2x10 6 , 3x10 6 , 4x10 6 , 5x10 6 , 6x10 6 , 7x10 6 8x10 6 , 9x10 6 , 1x10 7 , 2x10 7 , 3x10 7 , 4x10 7 , 5x10 7 , 6x10 7 , 7x10 7 , 8x10 7 , 9x10 7 , 1x10 8 , 2x10 8 , 3x10 8 , 4x10 8 , 5x10 8 , 6x10 8 , 7x10 8 , 8x10 8 , 9x10 8 , 1x10 9 , 2x10 9 , 3x10 9 , 4x10 9 , 5x10 9 , 6x10 9 , 6x10 9 , 7x10 9 , 7
  • the number of the TILs provided in the pharmaceutical compositions of the invention is in the range of 1x10 6 to 5x10 6 , 5x10 6 to 1x10 7 , 1x10 7 to 5x10 7 , 5x10 7 to 1x10 8 , 1x10 8 to 5x10 8 , 5x10 8 to 1x10 9 , 1x10 9 to 5x10 9 , 5x10 9 to 1x10 10 , 1x10 10 to 5x10 10 , 5x10 10 to 1x10 11 , 5x10 u to 1x10 12 , 1x10 12 to 5x10 12 , or 5x10 12 to 1x10 13 .
  • the number of TILs provided in a pharmaceutical composition is greater than 5x10 9 , and up to about 13.7x10 10 . In some embodiments, the number of TILs provided in a pharmaceutical composition is at least about 5x10. In some embodiments, the number of TILs provided in a pharmaceutical composition is up to about 13.7x10 10 .
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is less than, for example, 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002% or 0.0001% w/w, w/v or v/v of the pharmaceutical composition.
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.7
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.0001% to about 50%, about 0.001% to about 40%, about 0.01% to about 30%, about 0.02% to about 29%, about 0.03% to about 28%, about 0.04% to about 27%, about 0.05% to about 26%, about 0.06% to about 25%, about 0.07% to about 24%, about 0.08% to about 23%, about 0.09% to about 22%, about 0.1% to about 21%, about 0.2% to about 20%, about 0.3% to about 19%, about 0.4% to about 18%, about 0.5% to about 17%, about 0.6% to about 16%, about 0.7% to about 15%, about 0.8% to about 14%, about 0.9% to about 12% or about 1% to about 10% w/w, w/v or v/v of the pharmaceutical composition.
  • the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.001% to about 10%, about 0.01% to about 5%, about 0.02% to about 4.5%, about 0.03% to about 4%, about 0.04% to about 3.5%, about 0.05% to about 3%, about 0.06% to about 2.5%, about 0.07% to about 2%, about 0.08% to about 1.5%, about 0.09% to about 1%, about 0.1% to about 0.9% w/w, w/v or v/v of the pharmaceutical composition.
  • the amount of the TILs provided in the pharmaceutical compositions of the invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01
  • the amount of the TILs provided in the pharmaceutical compositions of the invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g
  • TILs can be administered to a patient as a pharmaceutical composition.
  • the pharmaceutical composition is a suspension of TILs in a sterile buffer.
  • TILs may be administered by any suitable route as known in the art.
  • the T-cells are administered as a single intra-arterial or intravenous infusion.
  • the T cell administration lasts approximately 30 to 60 minutes.
  • Other suitable routes of administration include intraperitoneal, intrathecal, and intralymphatic administration.
  • the TILs provided in the pharmaceutical compositions of the invention are effective over a wide dosage range.
  • TILs may be administered in a single dose. Such administration may be by injection, e.g., intravenous injection.
  • TILs may be administered in multiple doses. Dosing may be once, twice, three times, four times, five times, six times, or more than six times per year. Dosing may be once a month, once every two weeks, once a week, or once every other day. Administration of TILs may continue as long as necessary.
  • an effective dosage of TILs is about 1x10 6 , 2x10 6 , 3x10 6 , 4x10 6 , 5x10 6 , 6x10 6 , 7x10 6 , 8x10 6 , 9x10 6 , 1x10 7 , 2x10 7 , 3x10 7 , 4x10 7 , 5x10 7 , 6x10 7 , 7x10 7 , 8x10 7 , 9x10 7 , 1x10 8 , 2x10 8 , 3x10 8 , 4x10 8 , 5x10 8 , 6x10 8 , 7x10 8 , 8x10 8 , 9x10 8 , 1x10 9 , 2x10 9 , 3x10 9 , 4x10 9 , 5x10 9 , 6x10 9 , 7x10 9 , 8x10 9 , 9x10 9 , 1x10 10 , 2x10 9 , 3x10 9 , 4x10 9 , 5x10 9
  • an effective dosage of TILs is in the range of 1x10 6 to 5x10 6 , 5x10 6 to 1x10 7 , 1x10 7 to 5x10 7 , 5x10 7 to 1x10 8 , 1x10 8 to 5x10 8 , 5x10 8 to 1x10 9 , 1x10 9 to 5x10 9 , 5x10 9 to 1x10 10 , 1x10 10 to 5x10 10 , 5x10 10 to 1x10 11 , 5x10 11 to 1x10 12 , 1x10 12 to 5x10 12 , or 5x10 12 to 1x10 13 .
  • an effective dosage of TILs is in the range of about 0.01 mg/kg to about 4.3 mg/kg, about 0.15 mg/kg to about 3.6 mg/kg, about 0.3 mg/kg to about 3.2 mg/kg, about 0.35 mg/kg to about 2.85 mg/kg, about 0.15 mg/kg to about 2.85 mg/kg, about 0.3 mg to about 2.15 mg/kg, about 0.45 mg/kg to about 1.7 mg/kg, about 0.15 mg/kg to about 1.3 mg/kg, about 0.3 mg/kg to about 1.15 mg/kg, about 0.45 mg/kg to about 1 mg/kg, about 0.55 mg/kg to about 0.85 mg/kg, about 0.65 mg/kg to about 0.8 mg/kg, about 0.7 mg/kg to about 0.75 mg/kg, about 0.7 mg/kg to about 2.15 mg/kg, about 0.85 mg/kg to about 2 mg/kg, about 1 mg/kg to about 1.85 mg/kg, about 1.15 mg/kg to about 1.7 mg/kg, about
  • an effective dosage of TILs is in the range of about 1 mg to about 500 mg, about 10 mg to about 300 mg, about 20 mg to about 250 mg, about 25 mg to about 200 mg, about 1 mg to about 50 mg, about 5 mg to about 45 mg, about 10 mg to about 40 mg, about 15 mg to about 35 mg, about 20 mg to about 30 mg, about 23 mg to about 28 mg, about 50 mg to about 150 mg, about 60 mg to about 140 mg, about 70 mg to about 130 mg, about 80 mg to about 120 mg, about 90 mg to about 110 mg, or about 95 mg to about 105 mg, about 98 mg to about 102 mg, about 150 mg to about 250 mg, about 160 mg to about 240 mg, about 170 mg to about 230 mg, about 180 mg to about 220 mg, about 190 mg to about 210 mg, about 195 mg to about 205 mg, or about 198 to about 207 mg.
  • kits useful in performing diagnostic and prognostic assays using the TILs include buffers, cytokines, flasks, media, product containers, reagents and instructions.
  • An anti-PD-1 antibody may be administered in any effective dose, which can be determined based on all relevant factors by those of skill in this field.
  • an effective dose can be from 1 mg/kg to 20 mg/kg administered daily, once a week, once every two weeks, once every three weeks, once a month, and over longer periods of time.
  • a particular anti-PD-1 antibody is pembrolizumab (Merck & Co., NJ), which is a potent and highly selective humanized mAb of the IgG4/kappa isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. This antibody is supplied in a solution for injection at 100 mg/4mL.
  • a recommended dose is 2 mg/kg as an intravenous infusion over 30 minutes every three weeks.
  • Exemplary dosing regimens for pembrolizumab include: 200 mg every 3 weeks or 400 mg every 6 weeks, or 2 mg/kg (up to 200mg) for pediatrics.
  • the prescribed dose of pembrolizumab may be increased to 10 mg/kg every 21 days or 10 mg/kg every 14 days.
  • administration of pembrolizumab may be discontinued, or the dose reduced in the case of significant adverse effects. In certain embodiments, this recommended dosing can be reduced in an effective combination therapy.
  • nivolumab Another particular anti-PD-1 antibody is nivolumab (BMS).
  • BMS nivolumab
  • This antibody is supplied in a solution for injection at 40 mg/4 mL; 100 mg/10 mL; 120 mg/12 mL, or 240 mg/24 mL.
  • a recommended dose is between 1 mg/kg to 3 mg/kg every one to three weeks.
  • Exemplary dosing regimens for nivolumab include: 240 mg every 2 weeks, 360 mg every 3 weeks and 480 mg every 4 weeks. At the discretion of the clinician, depending upon individual tolerance, the prescribed dose of nivolumab may be increased or decreased.
  • IL-2 dosing may be administered in any effective dose, which can be determined based on all relevant factors by those of skill in this field.
  • Exemplary dosing regimens include: from about 50,000 IU/kg to about 1,000,000 IU/kg per dose; from about 100,000 IU/kg to about 800,000 IU/kg per dose; from about 550,000 IU/kg to about 750,000 IU/kg per dose; or from about 600,000 IU/kg to about 720,000 IU/kg per dose.
  • the current FDA-approved dose of high dose IL-2 is 600,000 IU/kg per dose administered intravenously every 8 hours for a maximum of 14 doses on days 1 to 5 (cycle 1) and days 15 to 19 (cycle 2), with a maximum of 28 doses for 1 course.
  • exemplary regimens include: in particular, 600,000 IU/kg per dose administered intravenously every 8 hours for a maximum of 8 doses for 4-5 days starting on day 0; a low dose regimen can be given, e.g., as low as about 50,000 IU/kg per dose, or as low as about 75,000 IU/kg per dose, or as low as about 100,000 IU/kg per dose, or as low as about 125,000 IU/kg per dose, or as low as about 150,000 IU/kg per dose, or as low as about 175,000 IU/kg per dose, or as low as about 200,000 IU/kg per dose, or as low as about 225,000 IU/kg per dose, or as low as about 250,000 IU/kg per dose, or as low as about 275,000 IU/kg per dose, or as low as about 300,000 IU/kg per dose, or as low as about 325,000 IU/kg per dose, or as low as about 350,000 IU/kg per dose, or as low as about 375,000 IU/kg per dose, or
  • IL-2 administration of doses of IL-2 can vary as appropriate and can be determined by those of skill in this field. Dosing can be as frequent as once about every 8 hours to once a day, or there can be longer periods of time between dosing.
  • Exemplary regimens include: low-dose subcutaneous IL-2 (125,000 IU/kg/day, maximum 9-10 doses over 2 weeks).
  • An exemplary regimen can be, day 0 bolus infusion of TIL followed by a continuous IL2 infusion administered in a decrescendo regimen (18 MIU/m 2 over 6, 12, and 24 hours followed by 4.5 MIU/m 2 over 24 hours for 3 days). IL2 infusion can start within about 12 hours after the TIL infusion.
  • maximum total dose of IL2 administered was limited to 135 MIU, corresponding to a body surface area of 2 m 2 .
  • maximum total dose of IL-2 administered was limited to 240 MIU, corresponding to a body surface area of 2 m 2 .
  • maximum total dose of IL2 administered was limited to 360 MIU, corresponding to a body surface area of 2 m 2 .
  • Treatment includes, but is not limited to, administration of a TIL product, an anti-PD-1 antibody, and optionally a cytokine, and may be performed either prophylactically or subsequent to diagnosis as part of a primary or follow-up therapeutic regimen.
  • Treatment includes, but is not limited to, inhibition or reduction of proliferation of cancer cells, destruction of cancer cells, prevention of proliferation of cancer cells or prevention of initiation of malignant cells or arrest or reversal of the progression of transformed premalignant cells to malignant disease or amelioration of the disease.
  • administering refers to the physical introduction of a TIL product, an anti-PD-1 antibody and/or a cytokine to a patient with cancer.
  • co-administering means a process whereby the combination of a TIL product, an anti-PD-1 antibody, and optionally a cytokine is administered to the same patient.
  • the TIL product, anti-PD-1 antibody and cytokine may be administered simultaneously, at essentially the same time, or sequentially. If administration takes place sequentially, the TIL product, anti PD 1 antibody and cytokine can be administered in any order. These agents need not be administered by means of the same vehicle.
  • the agents may be administered one or more times and the number of administrations of each component of the combination may be the same or different. In addition, the agents need not be administered at the same site. [094] As used herein, the terms “improved therapeutic outcome” and “enhanced therapeutic efficacy,” relative to cancer can refer to a slowing or diminution of the growth of cancer cells or a solid tumor, or a reduction in the total number of cancer cells or total tumor burden.
  • an “improved therapeutic outcome” or “enhanced therapeutic efficacy” therefore means there is an improvement in the condition of the patient according to any clinically acceptable criteria, including, for example, decreased tumor size, an increase in time to tumor progression, increased progression- free survival, increased overall survival time, an increase in life expectancy, or an improvement in quality of life.
  • “improved” or “enhanced” refers to an improvement or enhancement of 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of any clinically acceptable indicator of therapeutic outcome or efficacy.
  • a “targetable mutation” is a genomic alteration that occurs in a cancer cell that can be exploited for targeted therapy to that cancer cell.
  • Targetable genomic alterations include but are not limited to EGFR, KRAS, HER2, BRAF, ALK, RET, ROS1, NTRK fusions, and MET.
  • programmed death-1 is synonymous with one another, and include variants, isoforms, species homologs of human PD-1, and analogs having at least one common epitope with PD 1.
  • the full length human PD 1 cDNA is 2106 nucleotides long and encodes a protein of 288 amino acid residues.
  • the human PD-1 and murine PD-1 genes share 70% homology at the nucleotide level and 60% homology at the amino acid level.
  • the complete cDNA sequence of human PD-1 has the GenBank accession number U64863 (Shinohara et al., Genomics 23(3): 704-706 (1994).
  • the extracellular domain is encoded by amino acids 1-166; the transmembrane domain is encoded by amino acids 167-196; and the cytoplasmic domain is encoded by amino acids 197-288.
  • the extracellular domain contains an immunoglobulin superfamily domain, and the cytoplasmic domain includes an immunoreceptor tyrosine-based inhibitory motif (ITIM).
  • ITIM immunoreceptor tyrosine-based inhibitory motif
  • the complete cDNA sequence of murine PD-1 has the GenBank accession number X67914 (Ishida et al., EMBO J.11(11):3887-3895 (1992)).
  • the term “antibody” as used herein includes immunoglobulins, which are the product of B cells and variants thereof.
  • An immunoglobulin is a protein comprising one or more polypeptides substantially encoded by the immunoglobulin kappa and lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
  • subclasses of the heavy chain are known.
  • IgG heavy chains in humans can be any of IgG1, IgG2, IgG3 and IgG4 subclass.
  • Antibodies exist as full length intact antibodies or as a number of well-characterized fragments produced by digestion with various peptidases or chemicals. While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that any of a variety of antibody fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized de novo or antibodies and fragments obtained by using recombinant DNA methodologies. [0101] Recombinant antibodies may be conventional full length antibodies, antibody fragments known from proteolytic digestion, unique antibody fragments such as Fv or single chain Fv (scFv), domain deleted antibodies, and the like.
  • Fragments may include domains or polypeptides with as little as one or a few amino acid deleted or mutated while more extensive deletion is possible such as deletion of one or more domains.
  • Various adjuvants may be used to increase any immunological response, depending on the host species, including but not limited to Freund's adjuvant (complete and incomplete), mineral salts such as aluminum hydroxide or aluminum phosphate, various cytokines, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • the immune response could be enhanced by combination and or coupling with molecules such as keyhole limpet hemocyanin, tetanus toxoid, diphtheria toxoid, ovalbumin, cholera toxin or fragments thereof.
  • the present subject matter provides an immunotherapeutic approach for treating cancer, reducing the size of a tumor, inhibiting the growth of cancer cells in an individual, or reducing or inhibiting the development of metastatic cancer in an individual suffering from cancer. Therapy is achieved by treating a subject with a population of TILs, an anti-PD-1 antibody, and optionally a cytokine. In certain embodiments, therapy is achieved by treating a subject with a population of TILs, pembrolizumab and IL-2.
  • the TIL product, anti-PD-1 antibody and cytokine can be administered in any order.
  • the population of TILs may be administered before, during, or after administration of an anti-PD-1 antibody or cytokine.
  • the anti-PD-1 antibody may be administered before, during, or after administration of a population of TILs or cytokine.
  • the administration of one of these agents can be concurrent or within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or within a week to two weeks, or from weeks to months, of any other agent.
  • These therapies can be administered systemically at recommended dosage levels, which may be at lower doses compared to dosage levels required when administering the agents alone.
  • the anti-PD1 antibody is administered before, concurrent with, and/or after administration of a population of TILs. In some embodiments, the anti-PD1 antibody is administered before administration of the population of TILs. In some embodiments, the anti-PD1 antibody is administered before administration of the population of TILs. In some embodiments, the anti-PD1 antibody is administered concurrent with administration the population of TILs. In some embodiments, the anti-PD1 antibody is administered after administration of the population of TILs. In some embodiments, the method comprises administration of the anti-PD1 antibody one or more times, including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more times.
  • the anti PD1 antibody is administered after administration of the population of TILs until disease progression. In some embodiments, the anti-PD1 antibody is administered after administration of the population of TILs until intolerable toxicity occurs. In some embodiments, the anti-PD1 antibody is pembrolizumab. In some embodiments, the dose of pembrolizumab per administration is about any one of: 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg or more. In some embodiments, the dose of pembrolizumab per administration is from about 50 mg to about 150 mg, from about 150 mg to about 250 mg, from about 250 mg to about 350 mg, or from about 350 mg to about 450 mg.
  • the method comprises administering to the subject about 200 mg of pembrolizumab before administration of the population of TILs (such as during baseline imaging). In some embodiments, the method comprises administering pembrolizumab to the after administration of the population of TILs, such as administering to the subject about 200 mg of pembrolizumab 21 days after administration of the population of TILs. In some embodiments, the method comprises multiple administration of pembrolizumab to the subject after administration of the population of TILs.
  • the method comprises administering to the subject about 400 mg of pembrolizumab every 6 weeks after Day 21 post- administration of TILs, optionally wherein the administration of pembrolizumab every 6 week lasts for ⁇ 48 weeks in total, or until disease progression or until intolerable toxicity occurs.
  • the subject has undergone lymphodepletion (such as by lymphodepleting chemotherapy) prior to the administering of the combination.
  • the subject has undergone lymphodepletion prior to the administering of the population of TILs.
  • the lymphodepletion comprises administration of one or more lymphodepleting agents (such as lymphodepleting chemotherapeutics) on one or more days from about 1 day to about 14 days prior to administering of the population of TILs (e.g., from about day -1 to about day -14, such as one or more of days -1, -2, -3, -4, -5, -6, -7, -8, -9, -10, -11, -12, -13, and/or -14).
  • the lymphodepletion comprises one or more administrations of cyclophosphamide prior to administering of the population of TILs.
  • the lymphodepletion comprises administrations of cyclophosphamide at 60 mg/kg on day -7 and/or day -6. In some embodiments, the lymphodepletion comprises one or more administrations of fludarabine prior to administering of the population of TILs. In some embodiments, the lymphodepletion comprises administrations of fludarabine at 25 mg/m 2 on day 7, day 6, day 5, day 4 and/or day 3. In some embodiments, the lymphodepletion comprises one or more administrations of cyclophosphamide and fludarabine prior to administering of the population of TILs. [0107] In some embodiments according to any of the methods described herein, combination further comprises a cytokine.
  • the cytokine is IL-2.
  • the method comprises administering one or more doses of the cytokine to the subject (such as any one of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 doses).
  • the method comprises administering one or more doses of IL-2 to the subject concurrent and/or subsequent to administering of the population of TILs.
  • the method comprises administering up to 8 doses of IL-2 to the subject subsequent to administering of the population of TILs.
  • each dose of IL-2 administered is about 400,000 IU/kg to about 800,000 IU/kg.
  • each dose of IL-2 administered is about 600,000 IU/kg.
  • the subject matter described herein includes the following non-limiting embodiments: 1. A method of treating cancer in a subject, the method comprising administering to the subject a combination comprising: a) an effective amount of a population of TILs; and, b) an effective amount of an anti-PD-1 antibody. 2. The method of embodiment 1, wherein the combination further comprises an effective amount of a cytokine. 3. The method of embodiment 2, wherein the cytokine is IL-2. 4. The method of any one of embodiments 1-3, wherein the anti-PD-1 antibody is pembrolizumab. 5. The method of any one of embodiments 1-4, wherein the subject has undergone a previous cancer therapy.
  • the previous cancer therapy comprises a checkpoint inhibitor.
  • the cancer is HNSCC.
  • the method of embodiment 12, wherein the previous cancer therapy comprises a platinum-based chemotherapy.
  • the method of embodiment 12 or 13, wherein the previous cancer therapy comprises a checkpoint inhibitor.
  • the method of embodiment 8, wherein the cancer is NSCLC.
  • the method of embodiment 15, wherein the previous cancer therapy comprises a platinum-based chemotherapy.
  • the method of embodiment 15 or 16, wherein the previous cancer therapy comprises a checkpoint inhibitor. 18.
  • any one of embodiments 1-17 wherein: (I) the subject has undergone lymphodepletion prior to the administering of the combination, such as, cyclophosphamide 60 mg/kg IV on days -7 and -6 and fludarabine 25 mg/m 2 IV on days -7 to -3, optionally wherein the population of TILs is administered on day 0; or (II) the subject has undergone lymphodepletion prior to the administering of the population of TILs, such as, cyclophosphamide 60 mg/kg IV on days -7 and -6 and fludarabine 25 mg/m 2 IV on days -7 to -3, optionally wherein the population of TILs is administered on day 0. 19.
  • the subject has undergone lymphodepletion prior to the administering of the combination, such as, cyclophosphamide 60 mg/kg IV on days -7 and -6 and fludarabine 25 mg/m 2 IV on days -7 to -3, optionally wherein the population of TILs is administered on day
  • the effective amount of a population of TILs comprises a single infusion.
  • TILs tumor-infiltrating immune cells
  • the primary disease or conditions being studied in the trial, or focus of the study, are (1) cervical cancer, (2) head and neck squamous-cell carcinoma (HNSCC), and (3) non-small cell lung cancer (NSCLC).
  • HNSCC head and neck squamous-cell carcinoma
  • NSCLC non-small cell lung cancer
  • Secondary outcome measure means an outcome measure that is of lesser importance than a primary outcome measure, but is part of a pre-specified analysis plan for evaluating the effects of the intervention or interventions under investigation in a clinical study and is not specified as an exploratory or other measure.
  • a clinical study may have more than one secondary outcome measure.
  • Run A started with 1.33e+07 cells and reached a final TVC of 30.2E+09 cells
  • run B started with 3.33E+07 cells and reached a final TVC of 27.0E+09 cells
  • Run A started with 1.76E+06 CD3+ cells and reached a final count of 28.0E+09 CD3+ cells
  • run B started with 2.33E+06 CD3+ cells and reached a final count of 26.5E+09 CD3+ cells (FIG.4).
  • Phenotype assays were performed on both runs for CD2 and CD4/CD8 phenotype breakdown. Results for both runs show that most of the cells that are CD2+ are effector memory (EM) cells (FIG.5).
  • EM effector memory
  • TIL therapy has demonstrated efficacy in a variety of advanced solid tumors, including cervical cancer, non-small cell lung cancer (NSCLC), and head and neck squamous cell carcinoma (HNSCC).
  • NSCLC non-small cell lung cancer
  • HNSCC head and neck squamous cell carcinoma
  • TMP TIL manufacturing process
  • TIL production data included fold of expansion (total viable, CD3+) and characterization throughout day 1, 12, and 24 (viability, CD45%, CD3%, CD4%, and CD8%).
  • TIL phenotypic characterization throughout day 1, 12, and 24 included immune cell subsets (T ⁇ , T ⁇ , B, NK, monocytes, neutrophils, and dendritic cells), T cell memory subsets (Te, Tem, Tcm, and Tscm), and T cell activation and exhaustion status (41BB, OX40, PD1, TIM3, LAG3, CD39, CD103, CD69, CD25, CD27, CD28, and CD127).
  • TIL functional evaluation included cytokine secretion at 24 hours post-co-culture and activation markers regulation at 24 hours post-co-culture.
  • Two HNSCC tumors and two melanoma tumors were used in the TIL production workflow (tumor digest and cryopreservation, followed by TIL outgrowth starting at Day 1 (T-cell medium + 3000 IU/mL IL-2), REP starting at Day 12 (T-cell medium + 3000 IU/mL IL-2; 1 cell:200 feeder; 30 ng/mL OKT3), and TIL final product collection at Day 24).
  • TIL products were successfully produced from both HNSCC samples (data not shown).
  • HNSCC TIL products had good viability (higher than 80%) and good purity (higher than 90% of CD45+ as well as CD3+) (data not shown). There were little to no NK, B, monocyte, neutrophil, or dendritic cells in the TIL final products (data not shown). More effector T cells were observed in CD8 than CD4, and the majority of CD8 were effector memory and effector T cells, while the majority of CD4 were effector memory and central memory T cells (data not shown). The HNSCC TILs secreted IFN-gamma when co-cultured with autologous tumor digest, although less than melanoma TILs (data not shown). [0149] In summary, HNSCC TIL products were produced successfully.
  • HNSCC TIL products Besides the majority of T ⁇ cells, T ⁇ cells could expand to a significant portion in the final product from certain HNSCC tumors. Similar to melanoma TIL products, HNSCC TIL products expressed LAG3, TIM3, and CD39. The expression frequency of these 3 markers were higher on CD8 than CD4. Similar to melanoma TIL products, HNSCC CD8 mostly consists of effector memory and effector T cells, while CD4 mostly consists of effector memory and central memory T cells. Functional evaluation indicated that HNSCC TILs secrete IFN ⁇ when cocultured with autologous tumor digest, but to a lesser degree than melanoma TILs. Upregulation of 41BB, OX40, and CD69 were observed when TILs were stimulated with PMA/Ionomycin.
  • TIL production and phenotypic and functional characterization was also done on TILs generated from NSCLC tumors using the small scale research manufacturing process described above. TIL products were successfully produced from five NSCLC tumors tested. Four products had TIL products were successfully produced from 5 NSCLC tumors. Four products had CD3+TCR ⁇ + frequency above 80%, and one had 79.2% of CD3+TCR ⁇ +. The frequency of CD4 and CD8 vary among the 5 products (data not shown). [0151] Phenotypic characterization of the TIL products showed that the majority of products are T ⁇ cells. None to very little NK, B, DC, monocytes or neutrophils were detected in the final products.
  • CD4 and CD8 products express CD25, CD28, CD39, CD69, CD127, LAG3 and PD1.
  • a small subpopulation of CD4 express OX40.
  • the majority of products are effector memory T cells (CCR7- CD45RA-) (data not shown).
  • Co-culture with autologous tumor digest showed that the TILS secreted IFN ⁇ (less than 200 pg/mL) when cocultured with digest. No cytokines secretion was observed when cocultured with BA/F3. High IFN ⁇ , IL-2, and TNF ⁇ secretion was observed when cocultured with BA/F3 OKT3 or stimulated with PMA/Ionomycin.
  • TILs upregulated CD69 and PD1 on both CD4 and CD8 when cocultured with BA/F3 OKT3, but not BA/F3.
  • 41BB was upregulated on CD8 and OX40 was upregulated on CD4, when TILs were cocultured with BA/F3 OKT3 but not BA/F3.
  • TILs proliferated when cocultured with BA/F3 OKT3, but not with BA/F3 or autologous digest (data not shown).
  • Example 4 Clinical Study for Combination Therapy [0153] A clinical trial will evaluate the safety, feasibility, and preliminary efficacy of TILs in combination with pembrolizumab in participants with advanced cancer whose disease has progressed after standard therapy.
  • TILs tumor-infiltrating immune cells
  • HNSCC head and neck squamous-cell carcinoma
  • NSCLC non-small cell lung cancer
  • Intervention 1 Intervention Type Biological/Vaccine (select one) s n g a s r nce specified in the protocol, usually the one(s) used in the statistical power calculation. Most clinical studies have one primary outcome measure, but a clinical study may have more than one. [0158] “Secondary outcome measure” means an outcome measure that is of lesser importance than a primary outcome measure, but is part of a pre-specified analysis plan for evaluating the effects of the intervention or interventions under investigation in a clinical study and is not specified as an exploratory or other measure. A clinical study may have more than one secondary outcome measure.
  • Bridging therapy (at the discretion of the investigator) is allowed (FIG.20).
  • Baseline imaging occurs within 14 days prior to lymphodepleting chemotherapy (cyclophosphamide and fludarabine) followed by a single ITIL-168 infusion ( ⁇ 5 ⁇ 10 9 cells) on Day 0 and supportive short-course high-dose IL-2 (600,000 IU/kg, up to 8 doses).
  • Dosage and frequency of lymphodepleting chemotherapy will be adjusted based on cohort and patient comorbidities.
  • Pembrolizumab will be administered at baseline before ITIL-168 infusion (200 mg), at Day 21 post ITIL-168 infusion (200 mg), and then every 6 weeks for ⁇ 48 weeks (400 mg) or until disease progression or intolerable toxicity occur.
  • Primary analysis Will be conducted independently for each cohort after all patients in that cohort have been treated with ITIL-168 and have had the opportunity to be followed for ⁇ 24 weeks after the ITIL-168 infusion, are considered lost to follow-up, request a full withdrawal/are withdrawn from the study, or die (whichever occurs first). Study Populations [0168] Safety analysis set: All patients treated with ITIL-168; used for analysis of safety.
  • Full analysis set All enrolled patients; used for the analysis of manufacturing success rate, post-enrollment dropout rate prior to treatment, the summary of patient disposition, and patient listings of deaths.
  • Modified intent-to-treat analysis set All patients enrolled and treated with ITIL-168 products which met release specification, except for patients whose disease at the baseline visit appears to be responding since the time of enrollment; used for analyses of efficacy endpoints.
  • Study Analysis [0171] Primary analysis: Will be conducted independently for each cohort after all patients in that cohort have been treated with ITIL-168 and have had the opportunity to be followed for ⁇ 24 weeks after the ITIL-168 infusion, are considered lost to follow-up, request a full withdrawal/are withdrawn from the study, or die (whichever occurs first).

Abstract

The present subject matter provides an immunotherapeutic approach for treating cancer, reducing the size of a tumor, inhibiting the growth of cancer cells in an individual, or reducing or inhibiting the development of metastatic cancer in an individual suffering from cancer. Therapy is achieved by treating a subject with a combination therapy comprising a population of TILs, an anti-PD-1 antibody, and optionally a cytokine. The subject matter is also directed to treating a subject with a population of TILs, pembrolizumab and IL-2. The subject matter is also directed to treating subjects who although having undergone previous cancer therapy, the cancer subject's cancer has progressed.

Description

TUMOR INFILTRATING LYMPHOCYTES AND ANTI PD 1 ANTIBODY COMBINATION THERAPY FOR THE TREATMENT OF CANCER CROSS-REFERENCE TO RELATED APPLICATIONS [001] This application claims benefit of and priority to U.S. Provisional Patent Application No. 63/382,587, filed November 7, 2022, U.S. Provisional Patent Application No. 63/390,577, filed July 19, 2022, U.S. Provisional Patent Application No.63/366,886, filed June 23, 2022, and U.S. Provisional Patent Application No. 63/344,327, filed May 20, 2022, each of which is incorporated by reference herein in its entirety. FIELD [002] The subject matter disclosed herein relates to methods of enhancing an anti-tumor response by administering combinations comprising a population of tumor infiltrating lymphocyte (TILs) and an anti-PD-1 antibody to tumor cells in a patient with a cancer BACKGROUND [003] The immune system plays a role in the pathogenesis of a many cancers. When cancers progress, the immune system either fails to respond sufficiently or fails to respond appropriately, allowing cancer cells to grow. Improved strategies that combine specific manipulation of the immune response to cancer in combination with standard medical treatments may provide a means for enhanced efficacy and decreased toxicity. [004] In humans with cancer, antitumor immunity is often ineffective due to the tight regulation associated with the maintenance of immune homeostasis. One of the major limitations is a process known as “T-cell exhaustion,” which results from chronic exposure to antigens and is characterized by the upregulation of inhibitory receptors. These inhibitory receptors serve as immune checkpoints in order to prevent uncontrolled immune reactions. Blocking of one or several of these immune checkpoints with monoclonal antibodies (mAbs) has been shown to rescue otherwise exhausted antitumor T cells, and most importantly, has been associated with objective clinical responses in cancer patients. [005] The use of autologous cancer cells as immunotherapies to augment anti-tumor immunity has been explored. However, due to the weak immunogenicity of many cancers, down regulation of MHC molecules, the lack of adequate costimulatory molecule expression and secretion of immunoinhibitory cytokines by cancer cells, the response to such immunotherapies has been too often limited or short-term. [006] Targeted therapy of multiple non-redundant molecular pathways regulating immune responses may enhance antitumor immunotherapy. However, not all combinations have acceptable therapies. There remains a need for combination therapies with an acceptable safety profile and high efficacy that enhance antitumor immune responses compared to monotherapy and other immunotherapy combinations. SUMMARY [007] In certain embodiments, the subject matter described herein is directed to a method of treating a cancer comprising administering to a patient a therapeutically effective amount of tumor infiltrating lymphocytes (TILs) in combination with an antibody that specifically binds to human Programmed Death (PD)-1, also referred herein as an anti-PD-1 antibody. [008] In certain embodiments, the subject matter described herein is directed to a method of treating a subject having a cancerous tumor, the method comprising: a) administering an effective dose of an anti-PD-1 antibody; and b) administering an effective dose of a population of TILs to the subject. [009] In certain embodiments, the method further comprises administering a cytokine, such as IL-2, to the subject. [010] In certain embodiments, the cancer is advanced. [011] In certain embodiments, the cancer is a type of cancer selected from the group consisting of cervical cancer, head and neck squamous-cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC). [012] In certain embodiments, the cancer has progressed after the subject has received standard therapy for the type of cancer. [013] In certain embodiments, the subject has relapsed after standard therapy for the type of cancer. [014] In certain embodiments, the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, pidilizumab, durvalumab, atezolizumab, avelumab, cemiplimab, sintilimab, toripalimab, and camrelizumab. [015] In certain embodiments, administration of the combination results in enhanced therapeutic efficacy relative to administration of the TILs or the anti-PD-1 antibody alone. [016] In certain embodiments, the population of TILs may be administered prior to, at the same time as, or following administration of the anti-PD-1 antibody. [017] These and other embodiments are described fully below. BRIEF DESCRIPTION OF THE FIGURES [018] FIG. 1 depicts a protocol for certain embodiments of the combination therapies described herein. [019] FIG. 2 shows graphed outgrowth (days 1-13) total viable count (TVC) for ITIL-168 full-scale cervical runs. [020] FIG.3 shows graphed REP (days 13-25) TVC for ITIL-168 full-scale cervical runs. [021] FIG.4 shows graphed CD3+ cell counts throughout ITIL-168 full-scale cervical runs. [022] FIG.5 shows CD2 phenotype results for ITIL-168 full-scale cervical runs. [023] FIG.6 shows CD4/CD8 phenotype results for ITIL-168 full-scale cervical runs. [024] FIG.7 shows leukocyte data for ITIL-168 full-scale cervical runs. [025] FIG.8 shows potency results for ITIL-168 full-scale cervical runs. [026] FIGS.9A and 9B show outgrowth cell growth and REP cell growth, respectively for cSCC and cervical cancer indications (NC-200). [027] FIGS. 10A and 10B show total CD3+ cells over time for cSCC and cervical cancer indications, respectively, as measured by flow cytometry. [028] FIG. 11 shows CD3+ purity for both cSCC and cervical indications as measured by flow cytometry. [029] FIG.12 shows final product leukocyte data (T cells, NK cells, and monocytes; cSCC and cervical cancer indications). [030] FIG. 13 shows final product phenotype data (CD2+; cSCC and cervical cancer indications). [031] FIG.14 shows final product phenotype data (CD4+/CD8+/DP/DN; cSCC and cervical cancer indications). [032] FIG.15 shows final product potency data (cSCC and cervical cancer indications). [033] FIGS. 16A and 16B show outgrowth fold expansion and total viable CD3+ yield, respectively, for melanoma, NSCLC, cervical, and HNSCC indications. [034] FIGS. 17A and 17B show REP fold expansion and total viable CD3+ yield, respectively, for melanoma, NSCLC, cervical, and HNSCC indications. [035] FIGS. 18A and 18B show % viable of CD3+ cells and purity %CD3+ of CD45+), respectively, for melanoma, NSCLC, cervical, and HNSCC indications. [036] FIGS. 19A-19C show post-thaw drug product purity, viability, and potency for TILs from NSCLC and HNSCC indications. [037] FIG. 20 depicts a protocol for certain embodiments of the combination therapies described herein. HD, high-dose; IL-2, interleukin-2. DETAILED DESCRIPTION [038] As described fully herein, in certain embodiments, the subject matter is directed to a method of treating a cancer comprising administering to a patient a therapeutically effective amount of tumor infiltrating lymphocytes (TILs) in combination with an antibody that specifically binds to human Programmed Death (PD)-1, also referred herein as an anti-PD-1 antibody. As described herein, a clinical trial will evaluate the safety, feasibility, and preliminary efficacy of TILs in combination with pembrolizumab in participants with advanced cancer whose disease has progressed after standard therapy. The population of TILs to be administered is a cell therapy derived from a subject’s own tumor-infiltrating immune cells (lymphocytes; TILs). Despite some reports that certain TILs in combination with pembrolizumab do not provide any improvement compared to the use of individual components alone, the presently described methods have the potential to improve the effectiveness of a combination therapy and may provide synergistic results. The subject matter is also directed to treating subjects who although having undergone previous cancer therapy, the cancer subject’s cancer has progressed. [039] All references cited herein are incorporated by reference to the same extent as if each individual publication, patent application, or patent, was specifically and individually indicated to be incorporated by reference. [040] A “population of TILs” and “TIL product” and the like refer to an expanded population of tumor infiltrating lymphocytes derived from the subject’s tumor. The TIL product can be manufactured by the methods described in WO 2021/123832 and WO 2018/130845, each of which is incorporated by reference in its entirety. In some instances, the term “population of cells” (including TILs) herein can mean a number of cells that share some common traits. In general, populations generally range from 1 x 106 to 1 x 1012 in number, with different TIL populations comprising different numbers. [041] By tumor infiltrating lymphocytes or TILs herein is meant a population of cells originally obtained as white blood cells that have left the bloodstream of a subject and migrated into a tumor. TILs include, but are not limited to, CD8+ cytotoxic T cells (lymphocytes), Thi and Thi 7 CD4+ T cells, natural killer cells, dendritic cells, and Ml macrophages. TILs include both primary and secondary TILs. “Primary TILs” are those that are obtained from patient tissue samples as outlined herein (sometimes referred to as “freshly harvested”), and “secondary TILs” are any TIL cell populations that have been expanded or proliferated as discussed herein, including, but not limited to bulk TILs and expanded TILs (“REP TILs” or “post-REP TILs”). TIL cell populations can include genetically modified TILs. TILs can generally be defined either biochemically, using cell surface markers, or functionally, by their ability to infiltrate tumors and effect treatment. TILs can be generally categorized by expressing one or more of the following biomarkers: CD4, CD8, TCR ab, CD27, CD28, CD56, CCR7, CD45Ra, CD62L, CD95, PD-1, and CD25. Additionally and alternatively, TILs can be functionally defined by their ability to infiltrate solid tumors upon reintroduction into a patient. TILs may further be characterized by potency— for example, TILs may be considered potent or functional if in response to TCR engagement they produce, for example, interferon (IFN) release is greater than about 50 pg/mL, greater than about 100 pg/mL, greater than about 150 pg/mL, or greater than about 200 pg/mL, or individual cells can be considered potent through intracellular staining for CD 137, CD 107a, INF-g TNF-a, and IL-2 following TCR induced stimulation by flow cytometry. [042] In certain embodiments, the TIL product will have been assayed for the presence of and will contain T-cells expressing a combination of markers, the combination comprising two or more of: CD107a; CD137; TNF-α; or IFN-γ. In certain aspects of these embodiments, the combination comprises: CD107a and CD137; or CD107a and TNF-α; or CD107a and IFN-γ; or CD137 and TNF-α; or CD137 and IFN-γ; or TNF-α and IFN-γ. In certain aspects of these embodiments, the combination comprises three or more of: CD107a; CD137; TNF-α; and IFN-γ. In certain aspects of these embodiments, the combination comprises: CD107a, CD137, and TNF- α; or CD107a, CD137, and IFN-γ; or CD107a, TNF-α, and IFN-γ; or CD137, TNF-α, and IFN-γ. In certain aspects of these embodiments, the combination comprises each of CD107a, CD137, TNF-α, and IFN-γ. [043] In certain embodiments, the TIL product will have been assayed for the presence of and will contain CD2+ T-cells expressing a combination of markers, the combination comprising two or more of: a T cell expressing CD107a; a T cell expressing CD137; a T cell expressing TNF α; or a T-cell expressing IFN-γ. In some embodiments, the TIL product will have been assayed for the presence of and will contain CD3+ T-cells expressing a combination of markers, the combination comprising two or more of: a T-cell expressing CD107a; a T-cell expressing CD137; a T-cell expressing TNF-α; or a T-cell expressing IFN-γ. In certain aspects of these embodiments, the combination comprises: CD107a and CD137, or CD107a and TNF-α, or CD107a and IFN-γ, or CD137 and TNF-α, or CD137 and IFN-γ, or TNF-α and IFN-γ. In certain embodiments, the combination comprises three or more of: CD107a; CD137; TNF-α; and IFN-γ. In certain aspects of these embodiments, the combination comprises: CD107a, CD137 and TNF-α; or CD107a, CD137 and IFN-γ; or CD107a, TNF-α and IFN-γ; or CD137, TNF-α and IFN-γ. In certain embodiments, the combination comprises each of CD107a, CD137, TNF-α and IFN-γ. [044] TILs, in particular unmodified TILs (UTILs), are an autologous product; consequently, each batch manufactured provides a single dose for a specified patient. There are no sub-batches or pooling of batches. The drug product is a small aseptically prepared batch of T cells (5x109 to 5x1010) cryopreserved in a saline based solution with 8.5% human serum albumin and 10% DMSO of between 125-270 mL for a single intravenous infusion after thawing. The number of cells present is dependent on the ability of each individual’s TIL cells to be expanded in culture in conjunction with the culture conditions and the manufacturing reproducibility. [045] In some embodiments, anti-PD-1 antibodies as described herein are part of a combination therapy for improving an individual's immune response to cancer (e.g., a target cancer antigen or antigens) by co-administering a TIL product, an antibody which specifically binds to human Programmed Death (PD)-1, and optionally a cytokine. PD-1 is an immunoinhibitory receptor belonging to the CD28 family (Freeman et al., J. Exp. Med.192: 1027 (2000); Okazaki et al., Curr. Opin. Immunol.14: 779 (2002)) and binds to two ligands, PD-L1 and PD-L2. PD-1 is induced on T-cells, B-cells and myeloid cells in vitro (Agata et al., Int. Immunol.8:765 (1996)), but is predominantly expressed on previously activated T-cells in vivo (Iwai et al., Immunol. Lett.83:215 (2002)). [046] Exemplary anti-PD-1 antibodies include, but are not limited to, one or more of nivolumab, pembrolizumab, pidilizumab, durvalumab, atezolizumab, avelumab, cemiplimab, sintilimab, toripalimab, and camrelizumab. Certain anti-PD-1 antibodies and methods for their use are described by Goldberg et al., Blood 110(1):186-192 (2007), Thompson et al., Clin. Cancer Res.13(6):17571761 (2007), and Korman et al., International Application No. PCT/JP2006/309606 (publication no. WO 2006/121168 A1), each of which is incorporated by reference in its entirety. The antibodies for use in the present methods include, but are not limited to, monoclonal antibodies, synthetic antibodies, polyclonal antibodies, multispecific antibodies (including bi-specific antibodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain Fvs (scFv) (including bi-specific scFvs), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked Fvs (sdFv), and epitope-binding fragments of any of the above. In particular, antibodies for use in the present methods include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain a PD- 1 binding site that immunospecifically binds to PD-1. The immunoglobulin molecules for use in the present methods can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule. In some embodiments, the antibodies for use in the invention are IgG, such as IgG1. [047] The antibodies for use in the present methods may be from any animal origin including birds and mammals (e.g., human, murine, donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken). For example, the antibodies are human or humanized monoclonal antibodies. As used herein, “human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from mice or other animals that express antibodies from human genes. [048] The antibodies for use in the present methods include derivatives of any of the antibodies described herein. Standard techniques known to those of skill in the art can be used to introduce mutations in the nucleotide sequence encoding an antibody to be used with the methods for use in the invention, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis which result in amino acid substitutions. In some embodiments, the derivatives include less than 25 amino acid substitutions, less than 20 amino acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the original molecule. In a preferred embodiment, the derivatives have conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a side chain with a similar charge. Families of amino acid residues having side chains with similar charges 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), 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). Alternatively, mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed and the activity of the protein can be determined. [049] The antibodies for use in the present methods include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the antibody. For example, but not by way of limitation, the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, synthesis in the presence of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids. [050] The present methods also provide antibodies for use in the methods that comprise a framework region known to those of skill in the art. In certain embodiments, one or more framework regions, for example all of the framework regions, of an antibody to be used in the compositions and methods for use are human. In certain other embodiments for use in the methods, the fragment region of an antibody for use in the methods is humanized. In certain embodiments, the antibody to be used with the present methods is a synthetic antibody, a monoclonal antibody, an intrabody, a chimeric antibody, a human antibody, a humanized chimeric antibody, a humanized antibody, a glycosylated antibody, a multispecific antibody, a human antibody, a single-chain antibody, or a bispecific antibody. [051] In certain embodiments, an antibody for use in the invention has a high binding affinity for PD-1. In specific embodiments, an antibody for use in the invention has an association rate constant or kon, rate of about 105 M-1s-1 or more, about 5×105 M-1s-1 or more, about 106 M-1s-1 or more, about 5 10 M s or more, about 10 M s or more, about 5 10 M s or more, about 108 M-1s-1 or more, about 5×108 M-1s-1 or more, or about 1×109 M-1s-1 or more. [052] In other embodiments, an antibody for use in the invention has a koff rate for PD-1 of about 5×10−1 s-1 or less, about 10−1 s-1 or less, about 5×10−12 s-1 or less, about 10−2 s-1 or less, about 5×10−3 s-1 or less, about 10−3 s-1 or less, about 5×10−4 s-1 or less, about 10−4 s-1 or less, about 5×10−5 s-1 or less, about 10−5 s-1 or less, about 5×10−6 s-1 or less, about 10−6 s-1 or less, about 5×10−7 s-1 or less, about 10−7 s-1 or less, about 5×10−8 s-1 or less, about 10−8 s-1 or less, about 5×10−9 s-1 or less, about 10−9 s-1 or less, about 5×10−10 s-1 or less, or about 10−10 s-1 or less. [053] In certain embodiments, an antibody for use in the methods has an affinity constant or Ka (kon/koff) for PD-1 of about 102 M-1 or more, about 5×102 M-1 or more, about 103 M-1 or more, about 5×103 M-1 or more, about 104 M-1 or more, about 5×104 M-1 or more, about 105 M-1 or more, about 5×105 M-1 or more, about 106 M-1 or more, about 5×106 M-1 or more, about 107 M-1 or more, about 5×107 M-1 or more, about 108 M-1 or more, about 5×108 M-1 or more, about 109 M-1 or more, about 5×109 M-1 or more, about 1010 M-1 or more, about 5×1010 M-1 or more, about 1011 M-1 or more, about 5×1011 M-1 or more, about 1012 M-1 or more, about 5×1012 M-1 or more, about 1013 M- 1 or more, about 5×1013 M-1 or more, about 1014 M-1 or more, about 5×1014 M-1 or more, about 1015 M-1 or more, or about 5×1015 M-1 or more. [054] In certain embodiments, an antibody for use in the methods has a low dissociation constant. In specific embodiments, the antibody-binding domain of a carrier construct for use in the methods has a dissociation constant or Kd (koff/kon) for antibody about 5×10−1 M or less, about 10−1 M or less, about 5×10−2 M or less, about 10−2 M or less, about 5×10−3 M or less, about 10−3 M or less, about 5×10−4 M or less, about 10−4 M or less, about 5×10−5 M or less, about 10−5 M or less, about 5×10−6 M or less, about 10−6 M or less, about 5×10−7 M or less, about 10−7 M or less, about 5×10−8 M or less, about 10−8 M or less, about 5×10−9 M or less, about 10−9 M or less, about 5×10−10 M or less, or about 10−10 M or less. [055] In certain embodiments, an antibody for use in the present methods has a median effective concentration (EC50) of less than 0.01 nM, less than 0.025 nM, less than 0.05 nM, less than 0.1 nM, less than 0.25 nM, less than 0.5 nM, less than 0.75 nM, less than 1 nM, less than 1.25 nM, less than 1.5 nM, less than 1.75 nM, or less than 2 nM, in an in vitro microneutralization assay. The median effective concentration is the concentration of antibody that neutralizes 50% of PD-1 in an in vitro microneutralization assay. [056] The term cytokine and immunostimulatory cytokine includes cytokines that mediate or enhance the immune response to a foreign antigen, including viral, bacterial, or tumor antigens. Innate immunostimulatory cytokines can include, e.g., TNF-a, IL-1, IL-10, IL-12, IL-15, type I interferons (IFN-α and IFN-β), IFN-γ, and chemokines. Adaptive immunostimulatory cytokines include, e.g., IL-2, IL-4, IL-5, TGF-β, IL-10 and IFN-γ. Cytokines and combinations of cytokines have been shown to play an important role in the stimulation of the immune system. The term “cytokine” is understood by those of skill in the art, as referring to any immunopotentiating protein (including a modified protein such as a glycoprotein) that enhances or modifies the immune response to a tumor present in the host. The cytokine typically enhances or modifies the immune response by activating or enhancing the activity of cells of the immune system and is not itself immunogenic to the host. A variety of cytokines will find use in the methods described herein. Exemplary cytokines for use in practicing the invention include but are not limited to interferon- alpha (IFN-α), interferon-beta (IFN-β), and interferon-gamma (IFN-γ), interleukins (e.g., IL-1 to IL-29, in particular, IL-2, IL-7, IL-12, IL-15 and IL-18), tumor necrosis factors (e.g., TNF-alpha and TNF-beta), erythropoietin (EPO), MIP3a, intracellular adhesion molecule (ICAM), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF). The cytokine may be from any source, however, optimally the cytokine is of murine or human origin (a native human or murine cytokine) or is a sequence variant of such a cytokine, so long as the cytokine has a sequence with substantial homology to the human form of the cytokine and exhibits a similar activity on the immune system. It follows that cytokines with substantial homology to the human forms of IFN- alpha, IFN-beta, and IFN-gamma, IL-1 to IL-29, TNF-alpha, TNF-beta, EPO, MIP3a, ICAM, M- CSF, G-CSF and GM-CSF are useful in practicing the invention, so long as the homologous form exhibits the same or a similar effect on the immune system. Proteins that are substantially similar to any particular cytokine, but have relatively minor changes in protein sequence find use in the present invention. It is well known that small alterations in protein sequence may not disturb the functional activity of a protein molecule, and thus proteins can be made that function as cytokines in the present invention but differ slightly from current known or native sequences. [057] The term “IL-2” (also referred to herein as “IL2”) refers to the T cell growth factor known as interleukin-2, and includes all forms of IL-2 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-2 is described, e.g., in Nelson, J. Immunol.2004, 172, 398388 and Malek, Annu. Rev. Immunol.2008, 26, 453-79, the disclosures of which are incorporated by reference herein. For example, the term IL-2 encompasses human, recombinant forms of IL-2 such as aldesleukin (PROLEUKIN, available commercially from multiple suppliers in 22 million IU per single use vials), as well as the form of recombinant IL-2 commercially supplied by CellGenix, Inc., Portsmouth, N.H., USA (CELLGRO GMP) or ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-209-b) and other commercial equivalents from other vendors. Aldesleukin (des-alanyl-1, serine- 125 human IL-2) is a nonglycosylated human recombinant form of IL-2 with a molecular weight of approximately 15 kDa. The term IL-2 also encompasses pegylated forms of IL-2, as described herein, including the pegylated IL2 prodrug NKTR-214, available from Nektar Therapeutics, South San Francisco, Calif., USA. NKTR-214 and pegylated IL-2 suitable for use in the invention is described in U.S. Patent Application Publication No. US 2014/0328791 A1 and International Patent Application Publication No. WO 2012/065086 Al. Alternative forms of conjugated IL-2 suitable for use in the invention are described in U.S. Pat. Nos. 4,766,106, 5,206,344, 5,089,261 and 4902,502. Formulations of IL-2 suitable for use in the invention are described in U.S. Pat. No. 6,706,289. IL-2 (such as variants of IL-2) can encompasses modified IL-2. In some instances, modified IL-2 can mediate potent IL-2 Rβγ stimulation of CD8 effector T and NK cells, yet reduce IL-2 Rα binding to mitigate Treg-mediated suppression and off-target toxicity. Modified IL-2 can exhibit, for example, similar or enhanced affinity and/or activation of the IL-2 Rβ and γ subunits as compared to a non-modified IL-2. In some embodiments, modified IL-2 can exhibit similar or decreased affinity and/or engagement of the IL-2Rα subunit, as compared to a non-modified IL-2. [058] The term “IL-4” (also referred to herein as “IL4”) refers to the cytokine known as interleukin 4, which is produced by Th2 T cells and by eosinophils, basophils, and mast cells. IL- 4 regulates the differentiation of naive helper T cells (ThO cells) to Th2 T cells. Steinke and Borish, Respir. Res.2001, 2, 66-70. Upon activation by IL-4, Th2 T cells subsequently produce additional IL-4 in a positive feedback loop. IL-4 also stimulates B cell proliferation and class II MHC expression, and induces class switching to IgE and IgGl expression from B cells. Recombinant human IL-4 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-211) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL 15 recombinant protein, Cat. No. Gibco CTP0043). [059] The term “IL-7” (also referred to herein as “IL7”) refers to a glycosylated tissue- derived cytokine known as interleukin 7, which may be obtained from stromal and epithelial cells, as well as from dendritic cells. Fry and Mackall, Blood 2002, 99, 3892-904. IL-7 can stimulate the development of T cells. IL-7 binds to the IL-7 receptor, a heterodimer consisting of IL-7 receptor alpha and common gamma chain receptor, which in a series of signals important for T cell development within the thymus and survival within the periphery. Recombinant human IL-7 suitable for use in the invention is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-254) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No. Gibco PHC0071). [060] The term “IL-12” (also referred to herein as “IL12”) refers to the T cell growth factor known as interleukin- 12. Interleukin (IL)-12 is a secreted heterodimeric cytokine comprised of 2 disulfide- linked glycosylated protein subunits, designated p35 and p40 for their approximate molecular weights. IL-12 is produced primarily by antigen-presenting cells and drives cell- mediated immunity by binding to a two-chain receptor complex that is expressed on the surface of T cells or natural killer (NK) cells. The IL-12 receptor beta-1 (IL-12Rpi) chain binds to the p40 subunit of IL-12, providing the primary interaction between IL-12 and its receptor. However, it is IL-12p35 ligation of the second receptor chain, IL-12RP2, that confers intracellular signaling. IL- 12 signaling concurrent with antigen presentation is thought to invoke T cell differentiation towards the T helper 1 (Thl) phenotype, characterized by interferon gamma (IFNy) production. Thl cells are believed to promote immunity to some intracellular pathogens, generate complement fixing antibody isotypes, and contribute to tumor immunosurveillance. Thus, IL-12 is thought to be a significant component to host defense immune mechanisms. IL-12 is part of the IL-12 family of cytokines which also includes IL-23, IL-27, IL-35, IL-39. [061] The term “IL-15” (also referred to herein as “IL15”) refers to the T cell growth factor known as interleukin- 15, and includes all forms of IL-15 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-15 is described, e.g., in Fehniger and Caligiuri, Blood 2001, 97, 14-32, the disclosure of which is incorporated by reference herein. IL-15 shares b and g signaling receptor subunits with IL-2. Recombinant human IL 15 is a single, non glycosylated polypeptide chain containing 114 amino acids (and an N-terminal methionine) with a molecular mass of 12.8 kDa. Recombinant human IL-15 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-230-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-15 recombinant protein, Cat. No.34-8159-82). [062] The term “IL-18” (also referred to herein as “IL18”) refers to the T cell growth factor known as interleukin- 15. Interleukin- 18 (IL-18) is a proinflammatory cytokine that belongs to the IL-1 cytokine family, due to its structure, receptor family and signal transduction pathways. Related cytokines include IL-36, IL-37, IL-38. [063] The term “IL-21” (also referred to herein as “IL21”) refers to the pleiotropic cytokine protein known as interleukin-21, and includes all forms of IL-21 including human and mammalian forms, conservative amino acid substitutions, glycoforms, biosimilars, and variants thereof. IL-21 is described, e.g., in Spolski and Leonard, Nat. Rev. Drug. Disc.2014, 13, 379-95, the disclosure of which is incorporated by reference herein. IL-21 is primarily produced by natural killer T cells and activated human CD4+ T cells. Recombinant human IL-21 is a single, non-glycosylated polypeptide chain containing 132 amino acids with a molecular mass of 15.4 kDa. Recombinant human IL-21 is commercially available from multiple suppliers, including ProSpec-Tany TechnoGene Ltd., East Brunswick, N.J., USA (Cat. No. CYT-408-b) and ThermoFisher Scientific, Inc., Waltham, Mass., USA (human IL-21 recombinant protein, Cat. No.14-8219-80). [064] The term “subject” refers to any animal, such as, but not limited to, a mammal such as a human. In certain embodiments, the term “subject” refers to a patient that has undergone prior cancer treatment. In certain embodiments, the term “subject” refers to a patient whose cancer has progressed after standard therapy. Among these groups of subjects are patients who have a cancer that is susceptible to TIL therapy. Among these groups of subjects are patients who have cervical cancer, HNSCC or NSCLC. The subject will have undergone a resection of one or tumors in the subject to obtain a sample from which the TIL product will be developed. In certain embodiments, the subject will have undergone a prior treatment for the cancer. [065] The term “cancer” includes a myriad of diseases generally characterized by inappropriate cellular proliferation, abnormal or excessive cellular proliferation. Examples of cancer include but are not limited to, breast cancer, cervical cancer, colon cancer, head and neck squamous cell carcinoma (HNSCC), prostate cancer, pancreatic cancer, melanoma, lung cancer including non small cell lung cancer (NSCLC), ovarian cancer, kidney cancer, brain cancer, or sarcomas. Such cancers may be caused by one or more factors, including but not limited to environmental factors, chromosomal abnormalities, degenerative growth and developmental disorders, mitogenic agents, ultraviolet radiation (UV), viral infections, inappropriate tissue expression of a gene, alterations in expression of a gene, or carcinogenic agents. [066] When “an anti-tumor effective amount”, “a tumor-inhibiting effective amount”, “therapeutically 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). It can generally be stated that a pharmaceutical composition comprising the tumor infiltrating lymphocytes (e.g. secondary TILs or genetically modified cytotoxic lymphocytes) described herein may be administered at a dosage of 104 to 1011 cells/kg body weight (e.g., 105 to 106, 105 to 1010, 105 to 1011, 106 to 1010, 106 to 1011, 107 to 1011, 107 to 1010, 108 to 1011, 108 to 1010, 109 to 1011, or 109 to 1010 cells/kg body weight), including all integer values within those ranges. Tumor infiltrating lymphocytes (including in some cases, genetically modified cytotoxic lymphocytes) compositions may also be administered multiple times at these dosages. The tumor infiltrating lymphocytes (including in some cases, genetically) can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et ah, New Eng. J. of Med.319: 1676, 1988). The optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly. [067] Also, the term “therapeutically effective amount” or “therapeutically effective combination” as used herein can refer to an amount or dose of a TIL product together with the amount or dose of an anti-PD-1 antibody, and optionally a cytokine that is sufficient for treatment. The amount of each agent in a given therapeutically effective combination may be different for different individuals and different tumor types, and will be dependent upon the one or more additional agents or treatments included in the combination. The “therapeutically effective amount” is determined using procedures routinely employed by those of skill in the art such that an “improved therapeutic outcome” results. [068] Additionally, a combination therapy disclosed herein can provide synergistic effects whereby the effective amount of any one agent can be lower in the combination than the effective amount of the agent if given alone to achieve the same effect. As used herein, the term “synergism,” “synergistic” or “synergistically” refers to the combined action of two or more agents wherein the combined action is greater than the sum of the actions of each of the agents used alone. [069] Any suitable dose of TILs can be administered. In some embodiments, from about 2.3x1010 to about 13.7x1010 TILs are administered, with an average of around 7.8x1010 TILs. In an embodiment, about 1.2x1010 to about 4.3x1010 of TILs are administered. In some embodiments, about 3x1010 to about 12x1010 TILs are administered. In some embodiments, about 4x1010 to about 10x1010 TILs are administered. In some embodiments, about 5x1010 to about 8x1010 TILs are administered. In some embodiments, about 6x1010 to about 8x1010 TILs are administered. In some embodiments, about 7x1010 to about 8x1010 TILs are administered. In some embodiments, the therapeutically effective dosage is about 2.3x1010 to about 13.7x1010. In some embodiments, the therapeutically effective dosage is about 7.8x1010 TILs. In some embodiments, the therapeutically effective dosage is about 1.2x1010 to about 4.3x1010 of TILs. In some embodiments, the therapeutically effective dosage is about 3x1010 to about 12x1010 TILs. In some embodiments, the therapeutically effective dosage is about 4x1010 to about 10x1010 TILs. In some embodiments, the therapeutically effective dosage is about 5x1010 to about 8x1010 TILs. In some embodiments, the therapeutically effective dosage is about 6x1010 to about 8x1010 TILs. In some embodiments, the therapeutically effective dosage is about 7x1010 to about 8x1010 TILs. [070] In some embodiments, the number of the TILs provided in the pharmaceutical compositions of the invention is about any one of: 1x106, 2x106, 3x106, 4x106, 5x106, 6x106, 7x1068x106, 9x106, 1x107, 2x107, 3x107, 4x107, 5x107, 6x107, 7x107, 8x107, 9x107, 1x108, 2x108, 3x108, 4x108, 5x108, 6x108, 7x108, 8x108, 9x108, 1x109, 2x109, 3x109, 4x109, 5x109, 6x109, 7x109, 8x109, 9x109, 1x1010, 2x1010, 3x1010, 4x1010, 5x1010, 6x1010, 7x1010, 8x1010, 9x1010, 1x1011, 2x1011, 3x1011, 4x1011, 5x1011, 6x1011, 7x1011, 8x1011, 9x1011, 1x1012, 2x1012, 3x1012, 4x1012, 5x1012, 6x1012, 7x1012, 8x1012, 9x1012, 1x1013, 2x1013, 3x1013, 4x1013, 5x1013, 6x1013, 7x1013, 8x1013, or 9x1013. In an embodiment, the number of the TILs provided in the pharmaceutical compositions of the invention is in the range of 1x106 to 5x106, 5x106 to 1x107, 1x107 to 5x107, 5x107 to 1x108, 1x108 to 5x108, 5x108 to 1x109, 1x109 to 5x109, 5x109to 1x1010, 1x1010 to 5x1010, 5x1010 to 1x1011, 5x10u to 1x1012, 1x1012 to 5x1012, or 5x1012 to 1x1013. In a particular embodiment, the number of TILs provided in a pharmaceutical composition is greater than 5x109, and up to about 13.7x1010. In some embodiments, the number of TILs provided in a pharmaceutical composition is at least about 5x10. In some embodiments, the number of TILs provided in a pharmaceutical composition is up to about 13.7x1010. [071] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is less than, for example, 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002% or 0.0001% w/w, w/v or v/v of the pharmaceutical composition. [072] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%, 4.25%, 4%, 3.75%, 3.50%, 3.25%, 3%, 2.75%, 2.50%, 2.25%, 2%, 1.75%, 1.50%, 125%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002% or 0.0001% w/w, w/v, or v/v of the pharmaceutical composition. [073] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.0001% to about 50%, about 0.001% to about 40%, about 0.01% to about 30%, about 0.02% to about 29%, about 0.03% to about 28%, about 0.04% to about 27%, about 0.05% to about 26%, about 0.06% to about 25%, about 0.07% to about 24%, about 0.08% to about 23%, about 0.09% to about 22%, about 0.1% to about 21%, about 0.2% to about 20%, about 0.3% to about 19%, about 0.4% to about 18%, about 0.5% to about 17%, about 0.6% to about 16%, about 0.7% to about 15%, about 0.8% to about 14%, about 0.9% to about 12% or about 1% to about 10% w/w, w/v or v/v of the pharmaceutical composition. [074] In some embodiments, the concentration of the TILs provided in the pharmaceutical compositions of the invention is in the range from about 0.001% to about 10%, about 0.01% to about 5%, about 0.02% to about 4.5%, about 0.03% to about 4%, about 0.04% to about 3.5%, about 0.05% to about 3%, about 0.06% to about 2.5%, about 0.07% to about 2%, about 0.08% to about 1.5%, about 0.09% to about 1%, about 0.1% to about 0.9% w/w, w/v or v/v of the pharmaceutical composition. [075] In some embodiments, the amount of the TILs provided in the pharmaceutical compositions of the invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.008 g, 0.007 g, 0.006 g, 0.005 g, 0.004 g, 0.003 g, 0.002 g, 0.001 g, 0.0009 g, 0.0008 g, 0.0007 g, 0.0006 g, 0.0005 g, 0.0004 g, 0.0003 g, 0.0002 g, or 0.0001 g. [076] In some embodiments, the amount of the TILs provided in the pharmaceutical compositions of the invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075 g, 0.08 g, 0.085 g, 0.09 g, 0.095 g, 0.1 g, 0.15 g, 0.2 g, 0.25 g, 0.3 g, 0.35 g, 0.4 g, 0.45 g, 0.5 g, 0.55 g, 0.6 g, 0.65 g, 0.7 g, 0.75 g, 0.8 g, 0.85 g, 0.9 g, 0.95 g, 1 g, 1.5 g, 2 g, 2.5, 3 g, 3.5, 4 g, 4.5 g, 5 g, 5.5 g, 6 g, 6.5 g, 7 g, 7.5 g, 8 g, 8.5 g, 9 g, 9.5 g, or 10 g. [077] TILs can be administered to a patient as a pharmaceutical composition. In an embodiment, the pharmaceutical composition is a suspension of TILs in a sterile buffer. TILs may be administered by any suitable route as known in the art. In some embodiments, the T-cells are administered as a single intra-arterial or intravenous infusion. In some embodiments, the T cell administration lasts approximately 30 to 60 minutes. Other suitable routes of administration include intraperitoneal, intrathecal, and intralymphatic administration. [078] The TILs provided in the pharmaceutical compositions of the invention are effective over a wide dosage range. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the gender and age of the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician. The clinically-established dosages of the TILs may also be used if appropriate. The amounts of the pharmaceutical compositions administered using the methods herein, such as the dosages of TILs, will be dependent on the human or mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the active pharmaceutical ingredients and the discretion of the prescribing physician. [079] In some embodiments, TILs may be administered in a single dose. Such administration may be by injection, e.g., intravenous injection. In some embodiments, TILs may be administered in multiple doses. Dosing may be once, twice, three times, four times, five times, six times, or more than six times per year. Dosing may be once a month, once every two weeks, once a week, or once every other day. Administration of TILs may continue as long as necessary. [080] In some embodiments, an effective dosage of TILs is about 1x106, 2x106, 3x106, 4x106, 5x106, 6x106, 7x106, 8x106, 9x106, 1x107, 2x107, 3x107, 4x107, 5x107, 6x107, 7x107, 8x107, 9x107, 1x108, 2x108, 3x108, 4x108, 5x108, 6x108, 7x108, 8x108, 9x108, 1x109, 2x109, 3x109, 4x109, 5x109, 6x109, 7x109, 8x109, 9x109, 1x1010, 2x1010, 3x1010, 4x1010, 5x1010, 6x1010, 7x10108x1010, 9x1010, 1x1011, 2x1011, 3x1011, 4x1011, 5x1011, 6x1011, 7x1011, 8x1011, 9x1011, 1x1012, 2x1012, 3x1012, 4x1012, 5x1012, 6x1012, 7x1012, 8x1012, 9x1012, 1x1013, 2x1013, 3x1013, 4x1013, 5x1013, 6x1013, 7x1013, 8x1013, and 9x1013. [081] In some embodiments, an effective dosage of TILs is in the range of 1x106 to 5x106, 5x106 to 1x107, 1x107 to 5x107, 5x107 to 1x108, 1x108 to 5x108, 5x108to 1x109, 1x109 to 5x109, 5x109 to 1x1010, 1x1010 to 5x1010, 5x1010to 1x1011, 5x1011 to 1x1012, 1x1012 to 5x1012, or 5x1012 to 1x1013. [082] In some embodiments, an effective dosage of TILs is in the range of about 0.01 mg/kg to about 4.3 mg/kg, about 0.15 mg/kg to about 3.6 mg/kg, about 0.3 mg/kg to about 3.2 mg/kg, about 0.35 mg/kg to about 2.85 mg/kg, about 0.15 mg/kg to about 2.85 mg/kg, about 0.3 mg to about 2.15 mg/kg, about 0.45 mg/kg to about 1.7 mg/kg, about 0.15 mg/kg to about 1.3 mg/kg, about 0.3 mg/kg to about 1.15 mg/kg, about 0.45 mg/kg to about 1 mg/kg, about 0.55 mg/kg to about 0.85 mg/kg, about 0.65 mg/kg to about 0.8 mg/kg, about 0.7 mg/kg to about 0.75 mg/kg, about 0.7 mg/kg to about 2.15 mg/kg, about 0.85 mg/kg to about 2 mg/kg, about 1 mg/kg to about 1.85 mg/kg, about 1.15 mg/kg to about 1.7 mg/kg, about 1.3 mg/kg mg to about 1.6 mg/kg, about 1.35 mg/kg to about 1.5 mg/kg, about 2.15 mg/kg to about 3.6 mg/kg, about 2.3 mg/kg to about 3.4 mg/kg, about 2.4 mg/kg to about 3.3 mg/kg, about 2.6 mg/kg to about 3.15 mg/kg, about 2.7 mg/kg to about 3 mg/kg, about 2.8 mg/kg to about 3 mg/kg, or about 2.85 mg/kg to about 2.95 mg/kg. [083] In some embodiments, an effective dosage of TILs is in the range of about 1 mg to about 500 mg, about 10 mg to about 300 mg, about 20 mg to about 250 mg, about 25 mg to about 200 mg, about 1 mg to about 50 mg, about 5 mg to about 45 mg, about 10 mg to about 40 mg, about 15 mg to about 35 mg, about 20 mg to about 30 mg, about 23 mg to about 28 mg, about 50 mg to about 150 mg, about 60 mg to about 140 mg, about 70 mg to about 130 mg, about 80 mg to about 120 mg, about 90 mg to about 110 mg, or about 95 mg to about 105 mg, about 98 mg to about 102 mg, about 150 mg to about 250 mg, about 160 mg to about 240 mg, about 170 mg to about 230 mg, about 180 mg to about 220 mg, about 190 mg to about 210 mg, about 195 mg to about 205 mg, or about 198 to about 207 mg. [084] An effective amount of the TILs may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, topically, by transplantation, or by inhalation. [085] The present invention also includes kits useful in performing diagnostic and prognostic assays using the TILs, in particular unmodified TILs (UTILs), of the present invention. Kits of the invention include buffers, cytokines, flasks, media, product containers, reagents and instructions. [086] An anti-PD-1 antibody may be administered in any effective dose, which can be determined based on all relevant factors by those of skill in this field. In certain embodiments, an effective dose can be from 1 mg/kg to 20 mg/kg administered daily, once a week, once every two weeks, once every three weeks, once a month, and over longer periods of time. [087] A particular anti-PD-1 antibody is pembrolizumab (Merck & Co., NJ), which is a potent and highly selective humanized mAb of the IgG4/kappa isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. This antibody is supplied in a solution for injection at 100 mg/4mL. A recommended dose is 2 mg/kg as an intravenous infusion over 30 minutes every three weeks. Exemplary dosing regimens for pembrolizumab include: 200 mg every 3 weeks or 400 mg every 6 weeks, or 2 mg/kg (up to 200mg) for pediatrics. In the discretion of the clinician, depending upon individual tolerance, the prescribed dose of pembrolizumab may be increased to 10 mg/kg every 21 days or 10 mg/kg every 14 days. In the discretion of the clinician, together with the warnings provided with prescribing information, administration of pembrolizumab may be discontinued, or the dose reduced in the case of significant adverse effects. In certain embodiments, this recommended dosing can be reduced in an effective combination therapy. [088] Another particular anti-PD-1 antibody is nivolumab (BMS). This antibody is supplied in a solution for injection at 40 mg/4 mL; 100 mg/10 mL; 120 mg/12 mL, or 240 mg/24 mL. A recommended dose is between 1 mg/kg to 3 mg/kg every one to three weeks. Exemplary dosing regimens for nivolumab include: 240 mg every 2 weeks, 360 mg every 3 weeks and 480 mg every 4 weeks. At the discretion of the clinician, depending upon individual tolerance, the prescribed dose of nivolumab may be increased or decreased. In the discretion of the clinician, together with the warnings provided with prescribing information, administration of nivolumab may be discontinued, or the dose reduced in the case of significant adverse effects. In certain embodiments, this recommended dosing can be reduced in an effective combination therapy. [089] IL-2 dosing may be administered in any effective dose, which can be determined based on all relevant factors by those of skill in this field. Exemplary dosing regimens include: from about 50,000 IU/kg to about 1,000,000 IU/kg per dose; from about 100,000 IU/kg to about 800,000 IU/kg per dose; from about 550,000 IU/kg to about 750,000 IU/kg per dose; or from about 600,000 IU/kg to about 720,000 IU/kg per dose. The current FDA-approved dose of high dose IL-2 is 600,000 IU/kg per dose administered intravenously every 8 hours for a maximum of 14 doses on days 1 to 5 (cycle 1) and days 15 to 19 (cycle 2), with a maximum of 28 doses for 1 course. Other exemplary regimens include: in particular, 600,000 IU/kg per dose administered intravenously every 8 hours for a maximum of 8 doses for 4-5 days starting on day 0; a low dose regimen can be given, e.g., as low as about 50,000 IU/kg per dose, or as low as about 75,000 IU/kg per dose, or as low as about 100,000 IU/kg per dose, or as low as about 125,000 IU/kg per dose, or as low as about 150,000 IU/kg per dose, or as low as about 175,000 IU/kg per dose, or as low as about 200,000 IU/kg per dose, or as low as about 225,000 IU/kg per dose, or as low as about 250,000 IU/kg per dose, or as low as about 275,000 IU/kg per dose, or as low as about 300,000 IU/kg per dose, or as low as about 325,000 IU/kg per dose, or as low as about 350,000 IU/kg per dose, or as low as about 375,000 IU/kg per dose, or as low as about 400,000 IU/kg per dose, or as low as about 425,000 IU/kg per dose, or as low as about 450,000 IU/kg per dose, or as low as about 475,000 IU/kg per dose, or as low as about 500,000 IU/kg per dose, or as low as about 525,000 IU/kg per dose, or as low as about 550,000 IU/kg per dose, or as low as about 575,000 IU/kg per dose, or as low as about 600,000 IU/kg per dose, or as low as about 625,000 IU/kg per dose, or as low as about 650,000 IU/kg per dose, or as low as about 675,000 IU/kg per dose. [090] Administration of doses of IL-2 can vary as appropriate and can be determined by those of skill in this field. Dosing can be as frequent as once about every 8 hours to once a day, or there can be longer periods of time between dosing. Exemplary regimens include: low-dose subcutaneous IL-2 (125,000 IU/kg/day, maximum 9-10 doses over 2 weeks). An exemplary regimen can be, day 0 bolus infusion of TIL followed by a continuous IL2 infusion administered in a decrescendo regimen (18 MIU/m2 over 6, 12, and 24 hours followed by 4.5 MIU/m2 over 24 hours for 3 days). IL2 infusion can start within about 12 hours after the TIL infusion. In some embodiments, maximum total dose of IL2 administered was limited to 135 MIU, corresponding to a body surface area of 2 m2. In some embodiments, maximum total dose of IL-2 administered was limited to 240 MIU, corresponding to a body surface area of 2 m2. In some embodiments, maximum total dose of IL2 administered was limited to 360 MIU, corresponding to a body surface area of 2 m2. [091] As used herein “treatment” of an individual or a cell is any type of intervention used in an attempt to alter the natural course of the individual or cell. Treatment includes, but is not limited to, administration of a TIL product, an anti-PD-1 antibody, and optionally a cytokine, and may be performed either prophylactically or subsequent to diagnosis as part of a primary or follow-up therapeutic regimen. “Treatment” includes, but is not limited to, inhibition or reduction of proliferation of cancer cells, destruction of cancer cells, prevention of proliferation of cancer cells or prevention of initiation of malignant cells or arrest or reversal of the progression of transformed premalignant cells to malignant disease or amelioration of the disease. [092] The term “administering” as used herein refers to the physical introduction of a TIL product, an anti-PD-1 antibody and/or a cytokine to a patient with cancer. Any and all means of introduction may be contemplated according to the methods described herein; the method is not dependent on any particular means of introduction. Means of introduction are well-known to those skilled in the art, examples of which are provided herein. [093] The term “co-administering” as used herein means a process whereby the combination of a TIL product, an anti-PD-1 antibody, and optionally a cytokine is administered to the same patient. The TIL product, anti-PD-1 antibody and cytokine may be administered simultaneously, at essentially the same time, or sequentially. If administration takes place sequentially, the TIL product, anti PD 1 antibody and cytokine can be administered in any order. These agents need not be administered by means of the same vehicle. The agents may be administered one or more times and the number of administrations of each component of the combination may be the same or different. In addition, the agents need not be administered at the same site. [094] As used herein, the terms “improved therapeutic outcome” and “enhanced therapeutic efficacy,” relative to cancer can refer to a slowing or diminution of the growth of cancer cells or a solid tumor, or a reduction in the total number of cancer cells or total tumor burden. An “improved therapeutic outcome” or “enhanced therapeutic efficacy” therefore means there is an improvement in the condition of the patient according to any clinically acceptable criteria, including, for example, decreased tumor size, an increase in time to tumor progression, increased progression- free survival, increased overall survival time, an increase in life expectancy, or an improvement in quality of life. In particular, “improved” or “enhanced” refers to an improvement or enhancement of 1%, 5%, 10%, 25% 50%, 75%, 100%, or greater than 100% of any clinically acceptable indicator of therapeutic outcome or efficacy. [095] The term “relative to” or “compared to,” when used in the context of comparing the activity and/or efficacy of a combination composition comprising a TIL product plus an anti-PD- 1 antibody (anti-PD-1) and optionally a cytokine, refers to a comparison using amounts known to be comparable according to one of skill in the art. Comparable amounts of a TIL product, when comparing the combination therapy to a TIL product alone, may be based on cell number or any other relevant metric. Comparable amounts of anti-PD-1 antibody, when comparing the combination therapy to anti-PD-1 antibody alone, may be based on equimolar amounts, weight- to-weight equivalents, or units of PD-1 binding activity. [096] The term “reversal of an established tumor” as used herein means the suppression, regression, partial or complete disappearance of a pre-existing tumor. The definition is meant to include any diminution, for example, in the size, growth rate, appearance or cellular compositions of a preexisting tumor. [097] A “targetable mutation” is a genomic alteration that occurs in a cancer cell that can be exploited for targeted therapy to that cancer cell. Targetable genomic alterations include but are not limited to EGFR, KRAS, HER2, BRAF, ALK, RET, ROS1, NTRK fusions, and MET. [098] The terms “programmed death-1,” “programmed death receptor-1” and “PD-1” are synonymous with one another, and include variants, isoforms, species homologs of human PD-1, and analogs having at least one common epitope with PD 1. The full length human PD 1 cDNA is 2106 nucleotides long and encodes a protein of 288 amino acid residues. The human PD-1 and murine PD-1 genes share 70% homology at the nucleotide level and 60% homology at the amino acid level. The complete cDNA sequence of human PD-1 has the GenBank accession number U64863 (Shinohara et al., Genomics 23(3): 704-706 (1994). The extracellular domain is encoded by amino acids 1-166; the transmembrane domain is encoded by amino acids 167-196; and the cytoplasmic domain is encoded by amino acids 197-288. The extracellular domain contains an immunoglobulin superfamily domain, and the cytoplasmic domain includes an immunoreceptor tyrosine-based inhibitory motif (ITIM). The complete cDNA sequence of murine PD-1 has the GenBank accession number X67914 (Ishida et al., EMBO J.11(11):3887-3895 (1992)). [099] The term “antibody” as used herein includes immunoglobulins, which are the product of B cells and variants thereof. An immunoglobulin is a protein comprising one or more polypeptides substantially encoded by the immunoglobulin kappa and lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. Also subclasses of the heavy chain are known. For example, IgG heavy chains in humans can be any of IgG1, IgG2, IgG3 and IgG4 subclass. [0100] Antibodies exist as full length intact antibodies or as a number of well-characterized fragments produced by digestion with various peptidases or chemicals. While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that any of a variety of antibody fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized de novo or antibodies and fragments obtained by using recombinant DNA methodologies. [0101] Recombinant antibodies may be conventional full length antibodies, antibody fragments known from proteolytic digestion, unique antibody fragments such as Fv or single chain Fv (scFv), domain deleted antibodies, and the like. Fragments may include domains or polypeptides with as little as one or a few amino acid deleted or mutated while more extensive deletion is possible such as deletion of one or more domains. [0102] Various adjuvants may be used to increase any immunological response, depending on the host species, including but not limited to Freund's adjuvant (complete and incomplete), mineral salts such as aluminum hydroxide or aluminum phosphate, various cytokines, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum. Alternatively, the immune response could be enhanced by combination and or coupling with molecules such as keyhole limpet hemocyanin, tetanus toxoid, diphtheria toxoid, ovalbumin, cholera toxin or fragments thereof. [0103] The present subject matter provides an immunotherapeutic approach for treating cancer, reducing the size of a tumor, inhibiting the growth of cancer cells in an individual, or reducing or inhibiting the development of metastatic cancer in an individual suffering from cancer. Therapy is achieved by treating a subject with a population of TILs, an anti-PD-1 antibody, and optionally a cytokine. In certain embodiments, therapy is achieved by treating a subject with a population of TILs, pembrolizumab and IL-2. [0104] The TIL product, anti-PD-1 antibody and cytokine can be administered in any order. The population of TILs may be administered before, during, or after administration of an anti-PD-1 antibody or cytokine. The anti-PD-1 antibody may be administered before, during, or after administration of a population of TILs or cytokine. The administration of one of these agents can be concurrent or within 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or within a week to two weeks, or from weeks to months, of any other agent. These therapies can be administered systemically at recommended dosage levels, which may be at lower doses compared to dosage levels required when administering the agents alone. [0105] In some embodiments according to any of the methods described herein, the anti-PD1 antibody is administered before, concurrent with, and/or after administration of a population of TILs. In some embodiments, the anti-PD1 antibody is administered before administration of the population of TILs. In some embodiments, the anti-PD1 antibody is administered before administration of the population of TILs. In some embodiments, the anti-PD1 antibody is administered concurrent with administration the population of TILs. In some embodiments, the anti-PD1 antibody is administered after administration of the population of TILs. In some embodiments, the method comprises administration of the anti-PD1 antibody one or more times, including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more times. In some embodiments, the anti PD1 antibody is administered after administration of the population of TILs until disease progression. In some embodiments, the anti-PD1 antibody is administered after administration of the population of TILs until intolerable toxicity occurs. In some embodiments, the anti-PD1 antibody is pembrolizumab. In some embodiments, the dose of pembrolizumab per administration is about any one of: 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg or more. In some embodiments, the dose of pembrolizumab per administration is from about 50 mg to about 150 mg, from about 150 mg to about 250 mg, from about 250 mg to about 350 mg, or from about 350 mg to about 450 mg. In some embodiments, the method comprises administering to the subject about 200 mg of pembrolizumab before administration of the population of TILs (such as during baseline imaging). In some embodiments, the method comprises administering pembrolizumab to the after administration of the population of TILs, such as administering to the subject about 200 mg of pembrolizumab 21 days after administration of the population of TILs. In some embodiments, the method comprises multiple administration of pembrolizumab to the subject after administration of the population of TILs. In some embodiments, the method comprises administering to the subject about 400 mg of pembrolizumab every 6 weeks after Day 21 post- administration of TILs, optionally wherein the administration of pembrolizumab every 6 week lasts for ≤48 weeks in total, or until disease progression or until intolerable toxicity occurs. [0106] In some embodiments according to any of the methods described herein, the subject has undergone lymphodepletion (such as by lymphodepleting chemotherapy) prior to the administering of the combination. In some embodiments according to any of the methods described herein, the subject has undergone lymphodepletion prior to the administering of the population of TILs. In some embodiments, the lymphodepletion comprises administration of one or more lymphodepleting agents (such as lymphodepleting chemotherapeutics) on one or more days from about 1 day to about 14 days prior to administering of the population of TILs (e.g., from about day -1 to about day -14, such as one or more of days -1, -2, -3, -4, -5, -6, -7, -8, -9, -10, -11, -12, -13, and/or -14). In some embodiments, the lymphodepletion comprises one or more administrations of cyclophosphamide prior to administering of the population of TILs. In some embodiments, the lymphodepletion comprises administrations of cyclophosphamide at 60 mg/kg on day -7 and/or day -6. In some embodiments, the lymphodepletion comprises one or more administrations of fludarabine prior to administering of the population of TILs. In some embodiments, the lymphodepletion comprises administrations of fludarabine at 25 mg/m2 on day 7, day 6, day 5, day 4 and/or day 3. In some embodiments, the lymphodepletion comprises one or more administrations of cyclophosphamide and fludarabine prior to administering of the population of TILs. [0107] In some embodiments according to any of the methods described herein, combination further comprises a cytokine. In some embodiments, the cytokine is IL-2. In some embodiments, the method comprises administering one or more doses of the cytokine to the subject (such as any one of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 doses). In some embodiments, the method comprises administering one or more doses of IL-2 to the subject concurrent and/or subsequent to administering of the population of TILs. In some embodiments, the method comprises administering up to 8 doses of IL-2 to the subject subsequent to administering of the population of TILs. In some embodiments, each dose of IL-2 administered is about 400,000 IU/kg to about 800,000 IU/kg. In some embodiments, each dose of IL-2 administered is about 600,000 IU/kg. [0108] The subject matter described herein includes the following non-limiting embodiments: 1. A method of treating cancer in a subject, the method comprising administering to the subject a combination comprising: a) an effective amount of a population of TILs; and, b) an effective amount of an anti-PD-1 antibody. 2. The method of embodiment 1, wherein the combination further comprises an effective amount of a cytokine. 3. The method of embodiment 2, wherein the cytokine is IL-2. 4. The method of any one of embodiments 1-3, wherein the anti-PD-1 antibody is pembrolizumab. 5. The method of any one of embodiments 1-4, wherein the subject has undergone a previous cancer therapy. 6. The method of any one of embodiments 1-5, wherein the cancer has progressed after previous cancer therapy. 7. The method of any one of embodiments 1-6, wherein the cancer is malignant. 8. The method of any one of embodiments 1-7, wherein the cancer is selected from the group consisting of breast cancer, cervical cancer, colon cancer, head and neck squamous cell carcinoma (HNSCC), prostate cancer, pancreatic cancer, melanoma, lung cancer including non-small cell lung cancer (NSCLC), ovarian cancer, kidney cancer, brain cancer, and sarcoma. 9. The method of embodiment 8, wherein the cancer is cervical cancer. 10. The method of embodiment 9, wherein the previous cancer therapy comprises a platinum-based chemotherapy. 11. The method of embodiment 9 or 10, wherein the previous cancer therapy comprises a checkpoint inhibitor. 12. The method of embodiment 8, wherein the cancer is HNSCC. 13. The method of embodiment 12, wherein the previous cancer therapy comprises a platinum-based chemotherapy. 14. The method of embodiment 12 or 13, wherein the previous cancer therapy comprises a checkpoint inhibitor. 15. The method of embodiment 8, wherein the cancer is NSCLC. 16. The method of embodiment 15, wherein the previous cancer therapy comprises a platinum-based chemotherapy. 17. The method of embodiment 15 or 16, wherein the previous cancer therapy comprises a checkpoint inhibitor. 18. The method of any one of embodiments 1-17, wherein: (I) the subject has undergone lymphodepletion prior to the administering of the combination, such as, cyclophosphamide 60 mg/kg IV on days -7 and -6 and fludarabine 25 mg/m2 IV on days -7 to -3, optionally wherein the population of TILs is administered on day 0; or (II) the subject has undergone lymphodepletion prior to the administering of the population of TILs, such as, cyclophosphamide 60 mg/kg IV on days -7 and -6 and fludarabine 25 mg/m2 IV on days -7 to -3, optionally wherein the population of TILs is administered on day 0. 19. The method of any one of embodiments 1-18, wherein the effective amount of a population of TILs comprises a single infusion. 20. The method of any one of embodiments 1-19, wherein the effective amount of a population of TILs is a single infusion from about 2.3x1010 to about 13.7x1010 TILs. 21. The method of any one of embodiments 120, wherein the effective amount of an anti-PD-1 antibody comprises one dose prior to the administering of the effective amount of a population of TILs, and one or more additional doses after the administering of the effective amount of a population of TILs. 22. The method of embodiment 21, wherein each of the dose of the anti-PD-1 antibody is from about 1 mg/kg to about 10 mg/kg. 23. The method of any one of embodiments 3-22, wherein the effective amount of IL-2 comprises up to 8 doses. 24. The method of any one of embodiments 1-23, wherein the cancer has one or more targetable mutations. 25. The method of embodiment 24, wherein the mutation is selected from the group consisting of EGFR and ALK. [0109] The disclosed subject matter is further described in the following non-limiting Examples. It should be understood that these Examples, while indicating preferred embodiments of the invention, are given by way of illustration only. EXAMPLES Example 1. Clinical Study for Combination Therapy [0110] A clinical trial will evaluate the safety, feasibility, and preliminary efficacy of TILs in combination with pembrolizumab in participants with advanced cancer whose disease has progressed after standard therapy. The population of TILs is a cell therapy derived from a subject’s own tumor-infiltrating immune cells (lymphocytes; TILs). [0111] The primary disease or conditions being studied in the trial, or focus of the study, are (1) cervical cancer, (2) head and neck squamous-cell carcinoma (HNSCC), and (3) non-small cell lung cancer (NSCLC).
[0112] Table 1. Study Design. Study Type Interventional Primary Purpose (checkbox) Treatment
Figure imgf000030_0001
[0113] Table 2. Arms, Groups, and Interventions. Arm 1 Arm Type (select one): Experimental r -
Figure imgf000030_0002
Intervention 1: Intervention Type Biological/Vaccine (select one) s n g a s r
Figure imgf000031_0001
[0114] “Primary outcome measure” means the outcome measure(s) of greatest importance specified in the protocol, usually the one(s) used in the statistical power calculation. Most clinical studies have one primary outcome measure, but a clinical study may have more than one. [0115] “Secondary outcome measure” means an outcome measure that is of lesser importance than a primary outcome measure, but is part of a pre-specified analysis plan for evaluating the effects of the intervention or interventions under investigation in a clinical study and is not specified as an exploratory or other measure. A clinical study may have more than one secondary outcome measure.
[0116] Table 3. Outcome Measures. Outcome 1: Primary Outcome Measure Title: Frequency and severity of ITIL-168 treatment- emergent adverse even
Figure imgf000032_0001
Aes of special interest Time Frame: Up to 24 months Description:
Figure imgf000032_0002
econ ary Outcome Measure
Figure imgf000032_0003
, incidence of a complete response (CR) or a partial response (PR) per a modified Response .
Figure imgf000032_0004
[0117] A limited list of criteria for selection of participants in the clinical study, provided in terms of inclusion and exclusion criteria and suitable for assisting potential participants in identifying clinical studies of interest. Use a bulleted list for each criterion below the headers “Inclusion Criteria” and “Exclusion Criteria.” [0118] Table 4. Eligibility. Sex/Gender (checkbox) All Gender-Based (if any) No
Figure imgf000033_0001
Key Inclusion Criteria Histologically documented advanced (metastatic and/or unresectable) cervical cancer, HNSCC, or of e o n
Figure imgf000034_0001
based chemotherapy and a CPI. Participants with targetable mutations (e.g. EGFR/ALK) are required - a e,
Figure imgf000035_0001
Key Exclusion Criteria History of another primary malignancy within the previous 3 years nt s y e,
Figure imgf000036_0001
r mary ana ys s or eac co ort: 9- 5 pat ents treated and o owed or wee s Endpoints: ^ Primary – Safety, including treatment-emergent Aes; serious Aes and Aes of special interest ^ Secondary – Overall Response Rate, Overall Survival, Manufacturing Example 2. ITIL-168 Delta 2 Full-Scale Cervical Runs [0119] The purpose of these two studies was to expand the ITIL-168 process to cervical tumors. The ITIL-168 process is currently used for melanoma. These studies prove its success with other indications. [0120] The full-scale ITIL-168 process was assessed with two cervical donors’ tumor digests (Table 5) to gauge the feasibility of isolating doses from their final products. Both full-scale processes yielded high purity TILs that met final product specifications for the ITIL-168 program (Table 6). The full-scale process demonstrated the ability to enrich the T-cell population by the end of the ITIL-168 process using cervical tumors to give viable TILs that could be used to dose patients in the clinic. [0121] Table 5. Day 1 Tumor Digest Information. Tumor Study/ Tumor NC-200 Test 10 Test 10 Test 10 Tumor ID Indication Used in Source TVC CD45 TVC CD3 TVC %CD3 Purity R n
Figure imgf000037_0001
Day 25 CD3 TVC %CD3 Purity (Study arm- donor) TVC (NC-200) (NC-200*Accelix) (Accelix) %Viability
Figure imgf000037_0002
[ ] o u -scae cervca runs were compee usng e o owng suy pan: [0124] Table 7. Study plan for ITIL-168 full-scale cervical runs. Arm Process Scale Indication Tumor Replicates A Full (PL70) Cervical 9665 n = 1
Figure imgf000037_0003
[0125] Both full-scale cervical runs were completed following the ITIL-168 process (TMP 2.1) detailed in Table 8: [0126] Table 8. ITIL-168 TMP 2.1 Process. Day Process 1 Tumor Wash
Figure imgf000037_0004
[0127] Table 9. Equipment. Equipment Manufacturer Model or Catalog # Xuri Cytiva NA N l nt r Ch m m t NC200
Figure imgf000038_0001
. . Materials and Reagents Manufacturer Catalog # Lot # Expiry PL30 bags Origen PL30-2G NR NR
Figure imgf000038_0002
[0129] Both runs (A – 9665 and B – 9569) met all final product criteria. Run A started with 1.33e+07 cells and reached a final TVC of 30.2E+09 cells, and run B started with 3.33E+07 cells and reached a final TVC of 27.0E+09 cells (FIGS.2 and 3). Run A started with 1.76E+06 CD3+ cells and reached a final count of 28.0E+09 CD3+ cells, and run B started with 2.33E+06 CD3+ cells and reached a final count of 26.5E+09 CD3+ cells (FIG.4). Phenotype assays were performed on both runs for CD2 and CD4/CD8 phenotype breakdown. Results for both runs show that most of the cells that are CD2+ are effector memory (EM) cells (FIG.5). For run A, the population of cells is skewed mostly towards CD8+ cells (70% CD8+/22% CD4+), and for run B, the population of cells is more evenly split between CD8+ and CD4+ cells (37% CD8+/54% CD4%) (FIG. 6). The leukocyte flow panel was run and demonstrated the high T cell (CD3+CD19-) purity in both runs (FIG. 7). The potency assay showed run A to have 59% potency and run B to have 41% potency (FIG. 8). Both runs met criteria at harvest (day 25) for CD3+ TVC, CD3+ purity, and viability (Table 11). [0130] Table 11. Harvest results for ITIL-168 full scale cervical runs. Day 25 CD3 TVC %CD3 Purity (Study arm- donor) TVC (NC-200) (NC-200*Accelix) (Accelix) %Viability
Figure imgf000039_0001
e - u -sca e p ocess ca e success u y co p e e w so ce v cal tumors. The process was successfully run with two cervical tumors, demonstrating the ability of the ITIL-168 process to enrich T-cell population using cervical tumors to give viable, final product TILs that could be used to dose patients in the clinic.
[0132] Table 12. Data summary. CD3+ Data US23A – Day CC16 US23B – US30A – 9665 US30B – 9569 CC17 SCC i l i l M
Figure imgf000040_0001
Example 3. Additional ITIL-168 Delta 2 Runs [0133] TIL therapy has demonstrated efficacy in a variety of advanced solid tumors, including cervical cancer, non-small cell lung cancer (NSCLC), and head and neck squamous cell carcinoma (HNSCC). Using TIL manufacturing process (TMP) 2.1, studies were done on TILs from cervical cancer and cutaneous squamous cell carcinoma (cSCC) samples. Day 1 tumor characteristics are set forth below. [0134] Table 13. Day 1 tumor characteristics. Total Tumor Test 10 Test 10 Tumor NC-200 Test 10 Day 1 Indication Used in CD45 %CD3
Figure imgf000041_0001
[0135] Outgrowth cell growth and REP growth are shown in FIGS 9A and 9B, respectively. Outgrowth cell growth did not correlate with starting % CD3 purity, and REP growth did not correlate with outgrowth expansion or number of CD3+ cells seeded. Total CD3+ cells over time is shown in FIGS.10A and 10B. [0136] Table 14. %CD3 purity and CD3+ seeded on Day 1 and CD3+ TILs seeded on Day 13. Arm %CD3 Purity on Day 1 CD3+ Seeded Day 1 CD3+ TILs seeded Day 13
Figure imgf000041_0002
[0137] The percentage of CD3+ cells of the viable CD45+ cells increased for all donors during REP to > 90% by harvest (see FIG.11). Cervical arms grew comparatively better during REP than cSCC, but both indications show successful growth. US30A (13E+06 CD3 cells) showed best REP growth although it was seeded less than US30B (20E+06 CD3 cells). US23B showed the best outgrowth even though it had lowest %CD3 purity on Day 1. [0138] Table 15. CD3 fold expansion. Process Step Study Arm- Donor (Indication) CD3 Fold Exp by T10 and T15 US23A – CC16 (cSCC) 4.5
Figure imgf000042_0001
[0139] Final product leukocyte data are shown in FIG. 12 and show mostly pure T cell populations for all arms (both indications). Final product phenotype data (CD2+) are shown in FIG. 13 and show all arms are mostly CD2 EM. Final product phenotype data (CD2+/CD8+/DP/DN) are shown in FIG. 14 and show that US23A and US30B were heavily skewed toward CD8 or CD4, respectively. US23B and US30A were more evenly split between CD4 and CD8. Each arm had about the same DP, and US30A had a large DN population as compared to other arms. Final product potency data (2-analyte using CD107a and INF-gamma markers) are shown in FIG.15. US23A (cSCC) was activated the most in the presence of OKT3 at 65%, whereas US30B was activated the least at 41%. Final product attributes are shown in the table below (US23A was prematurely harvested two days early and should have continued to day 27). Even with the premature harvesting, three of four full-scale runs for two separate indications passed final release specifications, including high CD3+ purity and high cell viability for all four runs.
[0140] Table 16. Final product attributes. Day 25 CD3 TVC %CD3 Purity (Study arm- donor) TVC (NC-200) (NC-200*Accelix) (Accelix) %Viability
Figure imgf000043_0001
g g p . , ILs from melanoma, NSCLC, cervical cancer, and HNSCC. [0142] Outgrowth fold expansion and yield are shown in FIGS. 16A and 16B, respectively, and in the table below. [0143] Table 17. Outgrowth fold expansion and yield. Tumour ID Parameter C009118 9663 9688 9660 W007294 W007347 9664 eck 6 6 7
Figure imgf000043_0002
[0144] REP fold expansion and yield are shown in FIGS. 17A and 17B, respectively, and in the table below. Only 9660 and W007924 (plated into mid-scale REP) failed to reach a dose of 5E+09 viable CD3+ cells. [0145] Table 18. REP fold expansion and yield. Tumour ID Parameter C009118 9663 9688 9660 W007294 W007347 9664 ed
Figure imgf000043_0003
[0146] Percent viable of CD3+ cells and purity (%CD3+ of CD45+) are shown in FIGS.18A and 18B, respectively. Stability bags for two lung (9663 and 9688) and two HNSCC (W007347 and 9664) full scales were assessed for post-thaw purity (FIG. 19A), viability (FIG. 19B), and potency (FIG.19C). Pre-thaw and post-thaw at all time points (Day 25 of manufacturing process, and post-thaw at 0 weeks, 2 weeks, 1 month, 2 months, 3 months, 6 months, and 9 months post- thaw), all products had > 80% purity and between 90% and 95% viability. Likewise, all products at all time points met current (> 12%) and historic (> 40%) potency release criteria. [0147] TIL production and phenotypic and functional characterization was also done on TILs generated from melanoma and HNSCC tumors using a small scale research manufacturing process. TIL production data included fold of expansion (total viable, CD3+) and characterization throughout day 1, 12, and 24 (viability, CD45%, CD3%, CD4%, and CD8%). TIL phenotypic characterization throughout day 1, 12, and 24 included immune cell subsets (Tαβ, Tγδ, B, NK, monocytes, neutrophils, and dendritic cells), T cell memory subsets (Te, Tem, Tcm, and Tscm), and T cell activation and exhaustion status (41BB, OX40, PD1, TIM3, LAG3, CD39, CD103, CD69, CD25, CD27, CD28, and CD127). TIL functional evaluation (co-culture with autologous digest) included cytokine secretion at 24 hours post-co-culture and activation markers regulation at 24 hours post-co-culture. Two HNSCC tumors and two melanoma tumors were used in the TIL production workflow (tumor digest and cryopreservation, followed by TIL outgrowth starting at Day 1 (T-cell medium + 3000 IU/mL IL-2), REP starting at Day 12 (T-cell medium + 3000 IU/mL IL-2; 1 cell:200 feeder; 30 ng/mL OKT3), and TIL final product collection at Day 24). [0148] TIL products were successfully produced from both HNSCC samples (data not shown). Both HNSCC TIL products had good viability (higher than 80%) and good purity (higher than 90% of CD45+ as well as CD3+) (data not shown). There were little to no NK, B, monocyte, neutrophil, or dendritic cells in the TIL final products (data not shown). More effector T cells were observed in CD8 than CD4, and the majority of CD8 were effector memory and effector T cells, while the majority of CD4 were effector memory and central memory T cells (data not shown). The HNSCC TILs secreted IFN-gamma when co-cultured with autologous tumor digest, although less than melanoma TILs (data not shown). [0149] In summary, HNSCC TIL products were produced successfully. Besides the majority of Tαβ cells, Tγδ cells could expand to a significant portion in the final product from certain HNSCC tumors. Similar to melanoma TIL products, HNSCC TIL products expressed LAG3, TIM3, and CD39. The expression frequency of these 3 markers were higher on CD8 than CD4. Similar to melanoma TIL products, HNSCC CD8 mostly consists of effector memory and effector T cells, while CD4 mostly consists of effector memory and central memory T cells. Functional evaluation indicated that HNSCC TILs secrete IFNγ when cocultured with autologous tumor digest, but to a lesser degree than melanoma TILs. Upregulation of 41BB, OX40, and CD69 were observed when TILs were stimulated with PMA/Ionomycin. [0150] TIL production and phenotypic and functional characterization was also done on TILs generated from NSCLC tumors using the small scale research manufacturing process described above. TIL products were successfully produced from five NSCLC tumors tested. Four products had TIL products were successfully produced from 5 NSCLC tumors. Four products had CD3+TCRαβ+ frequency above 80%, and one had 79.2% of CD3+TCRαβ+. The frequency of CD4 and CD8 vary among the 5 products (data not shown). [0151] Phenotypic characterization of the TIL products showed that the majority of products are T αβ cells. None to very little NK, B, DC, monocytes or neutrophils were detected in the final products. Various frequency of both CD4 and CD8 products express CD25, CD28, CD39, CD69, CD127, LAG3 and PD1. A small subpopulation of CD4 express OX40. The majority of products are effector memory T cells (CCR7- CD45RA-) (data not shown). [0152] Co-culture with autologous tumor digest showed that the TILS secreted IFNγ (less than 200 pg/mL) when cocultured with digest. No cytokines secretion was observed when cocultured with BA/F3. High IFNγ , IL-2, and TNFα secretion was observed when cocultured with BA/F3 OKT3 or stimulated with PMA/Ionomycin. TILs upregulated CD69 and PD1 on both CD4 and CD8 when cocultured with BA/F3 OKT3, but not BA/F3. 41BB was upregulated on CD8 and OX40 was upregulated on CD4, when TILs were cocultured with BA/F3 OKT3 but not BA/F3. TILs proliferated when cocultured with BA/F3 OKT3, but not with BA/F3 or autologous digest (data not shown). Example 4. Clinical Study for Combination Therapy [0153] A clinical trial will evaluate the safety, feasibility, and preliminary efficacy of TILs in combination with pembrolizumab in participants with advanced cancer whose disease has progressed after standard therapy. The population of TILs is a cell therapy derived from a subject’s own tumor-infiltrating immune cells (lymphocytes; TILs). [0154] The primary disease or conditions being studied in the trial, or focus of the study, are (1) cervical cancer, (2) head and neck squamous-cell carcinoma (HNSCC), and (3) non-small cell lung cancer (NSCLC). [0155] Table 1. Study Design. Study Type Interventional Primary Purpose (checkbox) Treatment
Figure imgf000046_0001
[0156] Table 2. Arms, Groups, and Interventions. Arm 1 Arm Type (select one): Experimental e h or
Figure imgf000047_0001
Intervention 1: Intervention Type Biological/Vaccine (select one) s n g a s r
Figure imgf000048_0001
nce specified in the protocol, usually the one(s) used in the statistical power calculation. Most clinical studies have one primary outcome measure, but a clinical study may have more than one. [0158] “Secondary outcome measure” means an outcome measure that is of lesser importance than a primary outcome measure, but is part of a pre-specified analysis plan for evaluating the effects of the intervention or interventions under investigation in a clinical study and is not specified as an exploratory or other measure. A clinical study may have more than one secondary outcome measure.
[0159] Table 3. Outcome Measures. Outcome 1: Primary Outcome Measure Title: Frequency and severity of ITIL-168 treatment- emergent adverse even
Figure imgf000049_0001
aEs of special interest Time Frame: Up to 24 months Description:
Figure imgf000049_0002
econ ary Outcome Measure
Figure imgf000049_0003
, incidence of a complete response (CR) or a partial response (PR) per a modified Response .
Figure imgf000049_0004
[0160] A limited list of criteria for selection of participants in the clinical study, provided in terms of inclusion and exclusion criteria and suitable for assisting potential participants in identifying clinical studies of interest. Use a bulleted list for each criterion below the headers “Inclusion Criteria” and “Exclusion Criteria.” [0161] Table 4. Eligibility. Sex/Gender (checkbox) All Gender-Based (if any) No
Figure imgf000050_0001
Key Inclusion Criteria Histologically documented advanced (metastatic and/or unresectable) cervical cancer, HNSCC, or of e o n
Figure imgf000051_0001
based chemotherapy and a CPI. Participants with targetable mutations (e.g. EGFR/ALK) are required - a e,
Figure imgf000052_0001
Key Exclusion Criteria History of another primary malignancy within the previous 3 years nt d s y e,
Figure imgf000053_0002
nterm ana ys s or eac co ort: pat ents treate an o owe or wee s ^ Primary analysis for each cohort: 9-15 patients treated and followed for ≥24 weeks Endpoints: ^ Primary – Safety, including treatment-emergent aEs; serious aEs and aEs of special interest ^ Secondary – Overall Response Rate (Complete Response or Partial Response), Disease Control Rate (Complete Response, Partial Response or Stable Disease), Best Overall Response (Complete Response, Partial Response, Stable Disease, Partial Disease or Not Evaluable), Time to Response (to the first Complete Response or Partial Response), Duration Response Rate, Progression-Free Survival, Overall Survival, Manufacturing Success Rate, Pretreatment Dropout Rate
Figure imgf000053_0001
Treatment Schema [0162] Described here is an exemplary treatment schema: [0163] Tumor resection occurs within 30 days after patient consent. Bridging therapy (at the discretion of the investigator) is allowed (FIG.20). [0164] Baseline imaging occurs within 14 days prior to lymphodepleting chemotherapy (cyclophosphamide and fludarabine) followed by a single ITIL-168 infusion (≥5×109 cells) on Day 0 and supportive short-course high-dose IL-2 (600,000 IU/kg, up to 8 doses). [0165] Dosage and frequency of lymphodepleting chemotherapy will be adjusted based on cohort and patient comorbidities. [0166] Pembrolizumab will be administered at baseline before ITIL-168 infusion (200 mg), at Day 21 post ITIL-168 infusion (200 mg), and then every 6 weeks for ≤48 weeks (400 mg) or until disease progression or intolerable toxicity occur. [0167] Primary analysis: Will be conducted independently for each cohort after all patients in that cohort have been treated with ITIL-168 and have had the opportunity to be followed for ≥24 weeks after the ITIL-168 infusion, are considered lost to follow-up, request a full withdrawal/are withdrawn from the study, or die (whichever occurs first). Study Populations [0168] Safety analysis set: All patients treated with ITIL-168; used for analysis of safety. [0169] Full analysis set: All enrolled patients; used for the analysis of manufacturing success rate, post-enrollment dropout rate prior to treatment, the summary of patient disposition, and patient listings of deaths. [0170] Modified intent-to-treat analysis set: All patients enrolled and treated with ITIL-168 products which met release specification, except for patients whose disease at the baseline visit appears to be responding since the time of enrollment; used for analyses of efficacy endpoints. Study Analysis [0171] Primary analysis: Will be conducted independently for each cohort after all patients in that cohort have been treated with ITIL-168 and have had the opportunity to be followed for ≥24 weeks after the ITIL-168 infusion, are considered lost to follow-up, request a full withdrawal/are withdrawn from the study, or die (whichever occurs first). [0172] Designation of a range of values includes all integers within or defining the range, and all subranges defined by integers within the range. [0173] Unless otherwise apparent from the context, the term “about” encompasses values within a standard margin of error of measurement (e.g., SEM) of a stated value or variations ± 0.5%, 1%, 5%, or 10% from a specified value. [0174] Many modifications and other embodiments of the subject matter set forth herein will come to mind to one skilled in the art to which the inventions pertain having the benefit of the teachings presented in the foregoing descriptions and the associated drawings. Therefore, it is to be understood that the inventions are not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.

Claims

WHAT IS CLAIMED IS: 1. A method of treating cancer in a subject, the method comprising administering to the subject a combination comprising: (a) an effective amount of a population of TILs; and (b) an effective amount of an anti-PD-1 antibody.
2. The method of claim 1, wherein the combination further comprises an effective amount of a cytokine.
3. The method of claim 2, wherein the cytokine is IL-2.
4. The method of claim 1, wherein the anti-PD-1 antibody is pembrolizumab.
5. The method of claim 1, wherein the subject has undergone a previous cancer therapy.
6. The method of claim 1, wherein the cancer has progressed after previous cancer therapy.
7. The method of claim 1, wherein the cancer is malignant.
8. The method of claim 1, wherein the cancer is selected from the group consisting of breast cancer, cervical cancer, colon cancer, head and neck squamous cell carcinoma (HNSCC), prostate cancer, pancreatic cancer, melanoma, lung cancer including non-small cell lung cancer (NSCLC), ovarian cancer, kidney cancer, brain cancer, and sarcoma.
9. The method of claim 8, wherein the cancer is cervical cancer.
10. The method of claim 9, wherein the previous cancer therapy comprises a platinum-based chemotherapy.
11. The method of claim 9, wherein the previous cancer therapy comprises a checkpoint inhibitor.
12. The method of claim 8, wherein the cancer is HNSCC.
13. The method of claim 12, wherein the previous cancer therapy comprises a platinum-based chemotherapy.
14. The method of claim 12, wherein the previous cancer therapy comprises a checkpoint inhibitor.
15. The method of claim 8, wherein the cancer is NSCLC.
16. The method of claim 15, wherein the previous cancer therapy comprises a platinum-based chemotherapy.
17. The method of claim 15, wherein the previous cancer therapy comprises a checkpoint inhibitor.
18. The method of any one of claim 1, wherein the subject has undergone lymphodepletion prior to the administering of the combination or wherein the subject has undergone lymphodepletion prior to the administering of the population of TILs.
19. The method of any one of claim 1, wherein the effective amount of a population of TILs comprises a single infusion.
20. The method of any one of claim 1, wherein the effective amount of a population of TILs is a single infusion from about 2.3x1010 to about 13.7x1010 TILs.
21. The method of any one of claim 1, wherein the effective amount of an anti-PD-1 antibody comprises one dose prior to the administering of the effective amount of a population of TILs, and one or more additional doses after the administering of the effective amount of a population of TILs.
22. The method of claim 21, wherein each of the dose of the anti-PD-1 antibody is from about 1 mg/kg to about 10 mg/kg.
23. The method of claim 3, wherein the effective amount of IL-2 comprises up to 8 doses.
24. The method of claim 1, wherein the cancer has one or more targetable mutations.
25. The method of claim 24, wherein the mutation is selected from the group consisting of EGFR and ALK.
PCT/US2023/022712 2022-05-20 2023-05-18 Tumor infiltrating lymphocytes and anti-pd-1 antibody combination therapy for the treatment of cancer WO2023225181A1 (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US202263344327P 2022-05-20 2022-05-20
US63/344,327 2022-05-20
US202263366886P 2022-06-23 2022-06-23
US63/366,886 2022-06-23
US202263390577P 2022-07-19 2022-07-19
US63/390,577 2022-07-19
US202263382587P 2022-11-07 2022-11-07
US63/382,587 2022-11-07

Publications (1)

Publication Number Publication Date
WO2023225181A1 true WO2023225181A1 (en) 2023-11-23

Family

ID=88836162

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/022712 WO2023225181A1 (en) 2022-05-20 2023-05-18 Tumor infiltrating lymphocytes and anti-pd-1 antibody combination therapy for the treatment of cancer

Country Status (1)

Country Link
WO (1) WO2023225181A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022076952A1 (en) * 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022076952A1 (en) * 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
MARITAZ CHRISTOPHE, BROUTIN SOPHIE, CHAPUT NATHALIE, MARABELLE AURÉLIEN, PACI ANGELO: "Immune checkpoint-targeted antibodies: a room for dose and schedule optimization?", JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 15, no. 1, 1 December 2022 (2022-12-01), London UK , pages 1 - 16, XP093113975, ISSN: 1756-8722, DOI: 10.1186/s13045-021-01182-3 *
O’LEARY CONNOR, GASPER HARRY, SAHIN KATHERINE B., TANG MING, KULASINGHE ARUTHA, ADAMS MARK N., RICHARD DEREK J., O’BYRNE KEN J.: "Epidermal Growth Factor Receptor (EGFR)-Mutated Non-Small-Cell Lung Cancer (NSCLC)", PHARMACEUTICALS, vol. 13, no. 10, 1 October 2022 (2022-10-01), CH , pages 1 - 15, XP093113980, ISSN: 1424-8247, DOI: 10.3390/ph13100273 *
ZHANG CHUNYU, XU CHAO, GAO XUEYUN, YAO QINGQIANG: "Platinum-based drugs for cancer therapy and anti-tumor strategies", THERANOSTICS, vol. 12, no. 5, 1 January 2022 (2022-01-01), AU , pages 2115 - 2132, XP093113972, ISSN: 1838-7640, DOI: 10.7150/thno.69424 *

Similar Documents

Publication Publication Date Title
Qiao et al. Targeting tumors with IL-10 prevents dendritic cell-mediated CD8+ T cell apoptosis
CN111164100A (en) Interleukin-21 muteins and methods of treatment
US20040058445A1 (en) Activation of tumor-reactive lymphocytes via antibodies or genes recognizing CD3 or 4-1BB
US20180147257A1 (en) Btn3a ectodomain proteins and methods of use
TW201643184A (en) TNFRSF-binding agents and uses thereof
CA3031846A1 (en) Chimeric antigen receptor
Zhang et al. Biological effects of IL-15 on immune cells and its potential for the treatment of cancer
US20150157710A1 (en) Dual ox40 agonist/il-2 cancer therapy methods
JP2022513778A (en) Chimeric antigen receptor and T cell receptor and how to use
KR20080112232A (en) Methods for improving immune function and methods for prevention or treatment of disease in a mammalian subject
Ruby et al. IL-12 is required for anti-OX40-mediated CD4 T cell survival
WO2003083069A2 (en) Activation of tumor-reactive lymphocytes via antibodies or genes recognizing cd3 or 4-1bb
JP7032396B2 (en) Methods for facilitating T cell responses
JP2020510050A (en) Use of an oncolytic virus alone or in combination with a checkpoint inhibitor to treat cancer
US20220403006A1 (en) A Precision Medicine Method for Immunotherapy
KR102440962B1 (en) Pharmaceutical composition for treating cancer comprising fusion protein comprising il-2 protein and cd80 protein and anticancer drug
US20200371103A1 (en) Precision Medicine Method for Cancer Immunotherapy
US20230340052A1 (en) Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof
US20230134725A1 (en) Fusion Protein Extensions
WO2023225181A1 (en) Tumor infiltrating lymphocytes and anti-pd-1 antibody combination therapy for the treatment of cancer
Sivinski et al. Molecular requirements for CD8-mediated rejection of a MUC1-expressing pancreatic carcinoma: implications for tumor vaccines
CN117203328A (en) Recombinant antigen presenting cells
Hnízdilová Antitumor activity of IL-2 and IL-7 immunocomplexes in combination with αCTLA-4 and αPD-1 mAbs
WO2022098756A1 (en) Chimeric antigen receptor cell therapy
Garo et al. Role of Cytokines in Tumor Immunity and Immune Tolerance to Cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23808304

Country of ref document: EP

Kind code of ref document: A1