US20170044496A1 - Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy - Google Patents

Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy Download PDF

Info

Publication number
US20170044496A1
US20170044496A1 US15/303,201 US201515303201A US2017044496A1 US 20170044496 A1 US20170044496 A1 US 20170044496A1 US 201515303201 A US201515303201 A US 201515303201A US 2017044496 A1 US2017044496 A1 US 2017044496A1
Authority
US
United States
Prior art keywords
tumor
til
cells
antibody
cancer
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US15/303,201
Other languages
English (en)
Inventor
Amod A. Sarnaik
Jeffrey S. Weber
Shari Pilon-Thomas
Laszlo G. Radvanyi
Jessica Ann Chacon
James J. Mule'
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
H Lee Moffitt Cancer Center and Research Institute Inc
University of Texas System
Original Assignee
H Lee Moffitt Cancer Center and Research Institute Inc
University of Texas System
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 H Lee Moffitt Cancer Center and Research Institute Inc, University of Texas System filed Critical H Lee Moffitt Cancer Center and Research Institute Inc
Priority to US15/303,201 priority Critical patent/US20170044496A1/en
Publication of US20170044496A1 publication Critical patent/US20170044496A1/en
Assigned to H. LEE MOFFITT CANCER CENTER AND RESEARCH INSTITUTE, INC. reassignment H. LEE MOFFITT CANCER CENTER AND RESEARCH INSTITUTE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HALL, MACLEAN S., WEBER, JEFFREY S., MULE', JAMES J., PILON-THOMAS, Shari, SARNAIK, Amod A.
Assigned to BOARD OF REGENTS, THE UNIVERSITY OF TEXAS SYSTEM reassignment BOARD OF REGENTS, THE UNIVERSITY OF TEXAS SYSTEM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RADVANYI, LASZLO, CHACON, JESSICA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4636Immune checkpoint inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464417Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/46449Melanoma antigens
    • A61K39/464491Melan-A/MART
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/49Breast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/57Skin; melanoma
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/25Tumour necrosing factors [TNF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/599Cell markers; Cell surface determinants with CD designations not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere

Definitions

  • T-cell-based therapies have emerged to be powerful mediators of anti-tumor responses in both liquid and solid tumors [Dudley, M E (2011) J Cancer 2:360-2; Rosenberg, S A, et al (2008) Nat Rev Cancer 8(4):299-308].
  • TILs tumor-infiltrating lymphocytes
  • Adoptive cell therapy (ACT) with TILs for melanoma is predicated on the enriched tumor antigen-specificity of T cells infiltrating tumors which can be expanded to high numbers and re-infused.
  • the protocol used in most centers involves the initial outgrowth of TILs from 4-6 mm 2 cut fragments from metastatic melanoma surgical resections or biopsies using IL-2 as a growth factor [Dudley, M E (2011) J Cancer 2:360-2; Rosenberg, S A, et al (2008) Nat Rev Cancer 8(4):299-308; Radyanyi, L G, et al (2012) Clin Cancer Res 18(24):6758-70].
  • CD8 + TILs enriched in tumor specificity have emerged to be critical in mediating tumor regression in a number of Phase II TIL trials, and current efforts are now aimed to not only increase the expansion of CD8 + T cells from tumor tissue, but also increase the anti-tumor activity and effector-memory phenotype of the final TIL infusion product to improve persistence after adoptive transfer [Dudley, M E (2011) J Cancer 2:360-2; Rosenberg, S A, et al (2008) Nat Rev Cancer 8(4):299-308; Radvanyi, L G, et al (2012) Clin Cancer Res 18(24):6758-70; Besser, M J, et al (2010) Clin Cancer Res 16(9):2646-55; Valkenburg, S A, et al (2010) PLoS Pathog 6(8)].
  • Metastatic melanomas contain a population of CD8 + T cells expressing activation markers, such as PD-1 and 4-1BB/CD137, indicating a recent history of antigenic stimulation in the tumor microenvironment in vivo [Ye, Q, et al (2013) Clin Cancer Res 20(1):44-55].
  • CD8 + TIL expressing 4-1BB especially, represent the most highly enriched tumor-specific sub-population of T cells in melanoma [Ye, Q, et al (2013) Clin Cancer Res 20(1):44-55]. Protocols are now being developed to purify 4-1BB + CD8 + T cells from melanoma tissues and expand these selected cells for infusion.
  • a method for ex vivo expanding tumor-infiltrating lymphocytes for use in adoptive cell therapy involves culturing tumor fragments from the subject in a culture medium containing IL-2 and a 41BB agonist in an amount effective to expand tumor-infiltrating lymphocytes with enriched tumor-reactivity and specificity.
  • the method can enrich for tumor-specific CD8 + TILs compared to IL-2 alone. This increased tumor specificity can in some cases be maintained after further secondary rapid expansion (REP) with, for example, anti-CD3, irradiated PBMC feeder cells, and IL-2.
  • a method for treating a tumor in a subject that involves treating the subject with nonmyeloablative lymphodepleting chemotherapy, and administering tumor-infiltrating lymphocytes expanded by the disclosed methods.
  • FIGS. 1A and 1B are plots showing detection of 4-1BB on CD8 + T cells within melanoma tumor metastases and after 1 week of tumor fragment culture. Using flow cytometry, the expression of 4-1BB and OX40 was measured on CD8 + TIL single cell suspensions from 18 freshly-isolated melanoma metastasis ( FIG. 1A ) and tumor fragment cultures after 1 week from 7 patients ( FIG. 1B ).
  • FIGS. 2A to 2C show that anti-4-1BB agonistic antibody increases TIL expansion in vitro.
  • Melanoma tumors were surgically resected from patients. The tumors were then cut up into multiple fragments and placed in culture with IL-2 ⁇ anti-4-1BB antibody. Cell counts were conducted from days 6-14 using a hemocytometer and Trypan Blue. At these early time points, the 4-1BB activated TIL grew faster compared to the IL-2 control ( FIG. 2A ).
  • an increased expression of Ki67 proliferation marker was found in the 4-1BB activated TIL as compared to the control as demonstrated using immunocytochemistry ( FIG. 2B top) and further demonstrated in 4 independent TIL lines in the quantification data ( FIG. 2B bottom).
  • FIGS. 3A to 3D show increased NF ⁇ B translocation in TIL expanded with the anti-4-1BB antibody.
  • Melanoma tumors were cut up into multiple fragments and placed in culture with IL-2 and with or without an anti-4-1BB antibody. After 1 week, the cells were collected and cytospin and Immunocytochemistry were done. Staining for NF ⁇ B (p65) revealed more translocation in the nucleus of TIL grown with IL-2 and the anti-4-1BB antibody compared to TIL grown with IL-2 alone.
  • Vectra Intelligents slide analysis system (Nuance software) ( FIG. 3A ) was used to observe the translocation of NF ⁇ B (red is nucleus; green is NF ⁇ B; and yellow is overlay (translocation)).
  • the arrows indicate areas where translocation occurred. Quantification of NF ⁇ B translocation in 10 different areas per sample is demonstrated in 3 independent TIL samples ( FIG. 3B ). The percentage of NF ⁇ B and I ⁇ B ⁇ expression was also measured in the TIL within the fragment after 1 week in culture using flow cytometry ( FIG. 3C, 3D ). The fragments were set up with or without the addition of anti-4-1BB antibody with IL-2 or with IL-2 alone. After 1 week, the fragment was collected and mechanically disaggregated using glass slides. The cells were filtered and stained using flow cytometry. The TIL were gated on live, CD3 + , CD8 + TIL and the percentage of NF ⁇ B ( FIG. 3C ) and I ⁇ B ⁇ ( FIG. 3D ) was measured. The TIL cultured with IL-2 plus anti-4-1BB antibody exhibited an increase NF ⁇ B ( FIG. 3C ) with a paralleled decrease in I ⁇ B ⁇ ( FIG. 3D ).
  • FIGS. 4A to 4D show that CD8 + TIL percentage is increased with the addition of anti-4-1BB antibody to TIL cultures.
  • Experiments were conducted to determine whether activating the 4-1BB pathway could augment the percentage of CD8 + TIL in the cultures. 1 week after the fragments were set up, immunocytochemistry was conducted, revealing that the TIL expanded with IL-2 and anti-4-1BB antibody exhibited an increase in CD8 + cells, as shown in one representative TIL line ( FIG. 4A ). After being in culture for 3 weeks, the TILs were stained for the expression of CD3, CD8, and CD4 using flow cytometry.
  • FIGS. 5A to 5G show phenotypic characterization of effector-memory markers in TIL isolated from tumor fragment cultures treated with or without-anti-4-1BB. Fragments from melanoma tumors were set up in a 24 well plate with or without anti-4-1BB antibody. 3 weeks after the initial set-up, the TILs were harvested and flow cytometry was done to analyze effector-memory markers and cytolytic granule markers. When the expression of CD28 was measured in the CD3 + CD8 + subset in 48 independent TIL lines, no statistical significance was found in the expression of CD28 in the CD3 + CD8 + subset in the different conditions ( FIG. 5A ).
  • FIGS. 6A to 6E show increased tumor specificity of TIL from tumor fragment cultures treated with anti-4-1BB.
  • the fragments were set up with IL-2 alone or IL-2 plus anti-4-1BB antibody and expanded over a 3 week period (pre-REP)
  • the TIL were harvested and set up at a 1:1 ratio with autologous ( FIG. 6A , left and middle) or HLA-A matched ( FIG. 6A , right) tumor cells.
  • Supernatants were collected after 24 hours and IFN-gamma secretion was measured.
  • the TIL were set up at a 1:1 ratio with HLA-A matched tumor lines.
  • the amount of IFN-gamma+ cells was measured in the CD3 + CD8 + subset ( FIG. 6B ).
  • the TIL expanded with anti-4-1BB antibody exhibited an increase in IFN-gamma+ cells ( FIG. 6B ).
  • flow cytometry was used to determine the amount of degranulation after setting up the TIL at a 1:1 ratio with HLA-A matched tumor lines ( FIG. 6C ).
  • the TIL grown with the anti-4-1BB antibody exhibited an increase in CD107a + cells in the CD3 + CD8 + subset ( FIG. 6C ).
  • the percentage of MART-1 + specific T cells was measured on the CD3 + CD8 + TIL initially expanded with IL-2 alone or IL-2 with anti-4-1BB antibody.
  • the TIL expanded with the anti-4-1BB antibody exhibited an increase in antigen specific population, compared to the TIL expanded with IL-2 alone ( FIG. 6D ).
  • the pre-REP TILs that were initially expanded with or without the anti-4-1BB antibody then underwent the secondary expansion (REP).
  • the post-REP TIL were harvested and set up at a 1:1 ratio with the autologous tumor ( FIG. 6E , left) or HLA-matched ( FIG. 6E , middle and right) tumor cells. 24 hours later, the supernatants were collected and IFN-gamma secretion was measured using ELISA.
  • the post-REP TIL that were initially expanded with anti-4-1BB antibody exhibited an increase in Interferon-gamma secretion compared to the TIL initially expanded with IL-2 alone ( FIG. 6E ).
  • FIGS. 7A to 7E show that addition of anti-4-1BB antibody to the melanoma fragments increased dendritic cell activation markers and addition of anti-MHC-I antibody decreases expansion of CD8 + TIL.
  • the melanoma tumors were cut up into multiple fragments and placed in culture with IL-2 alone or IL-2+anti-4-1BB antibody. After 1 week in culture, the fragments were mechanically disaggregated, filtered, and stained for DC and their activation markers.
  • the DC expressed 4-1BB and the addition of the anti-4-1BB antibody increased CD86, CD80 and MHC II expression ( FIG. 7A ).
  • FIG. 8 is a bar graph showing dose titration of different concentrations of agonist anti-4-1BB antibody added to melanoma tumor fragment cultures to determine the optimal dose for the anti-4-1BB antibody for initial TIL expansion.
  • Cells counts were done after 3 weeks of culture of the tumor fragments with or without the indicated concentrations of anti-4-1BB (BMS 663513). 10 mg/ml anti-4-1BB antibody was found to optimal and induced the highest TIL expansion in initial experiments on 3 different melanoma patient tumors (5 fragments per condition for each anti-41-BB dose).
  • FIGS. 9A to 9E show that TILs initially expanded with anti-4-1BB antibody continue to expand during secondary expansion.
  • TIL were initially expanded with or without anti-4-1BB antibody
  • the pre-REP TIL were then subjected to the REP.
  • phenotype analysis and cell counts were conducted.
  • Post-REP TIL initially expanded with anti-4-1BB antibody exhibited increased CD8 + expression ( FIG. 9A ) and similar levels of CD27 ( FIG. 9B ) and CD28 ( FIG. 9C ).
  • Total ( FIG. 9D ) and CD8 ( FIG. 9E ) fold expansion was not disrupted despite the loss of CD27 at the pre-REP level in the TIL expanded with anti-4-1BB antibody.
  • FIGS. 10A and 10B are bar graph showing that addition of anti-4-1BB antibody during ex vivo expansion of TIL isolated from B16 melanomas increases anti-tumor reactivity.
  • Mice were injected subcutaneously with 1 ⁇ 10 5 B16 melanoma cells. Tumors were resected on day 21, digested, and T cells were purified by AUTOMACS separation. T cells were cultured in vitro with 6000 IU/ml IL-2 or IL-2 plus anti-4-1BB for 7 days. T cells from B16 tumors were co-cultured with B16 cells ( FIG. 10A ). Supernatants were collected and IFN-gamma was measured by ELISA.
  • mice received i.p. 20 mg/kg normal or non-specific IgG (NrIgG) or anti-PD1 antibody.
  • Tumors were resected, digested and T cells were purified by AUTOMACS (Miltenyi) separation.
  • T cells were cultured in vitro with 6000 IU/ml IL-2 and 10 mg/ml NrIgG or anti-4-1BB antibody.
  • On day 7, T cells were co-cultured with B16 cells for 24 hours. Supernatants were collected and IFN-gamma was measured by ELISA ( FIG. 10B ).
  • FIG. 11 is an illustration of the current adoptive T-cell therapy protocol for melanoma.
  • FIG. 12 is an illustration of a protocol used for tumor processing of Triple Negative Breast Cancers (TNBCs)/Inflammatory Breast Cancers (IBC).
  • TNBCs Triple Negative Breast Cancers
  • IBC Inflammatory Breast Cancers
  • FIGS. 14A to 14B are plots showing that T cell differentiation was not different in TIL growth from surgical samples or biopsy samples.
  • FIGS. 15A to 15B are plots showing addition of anti 4-1BB antibody increases total yield of isolated from breast cancer fragment after 4 weeks of culture with IL-2.
  • FIGS. 16A to 16E show that addition of anti 4-1BB antibody increases the frequency of CD8+ T cells and a CD4-CD8 ⁇ T cell population expanded from tumor fragments.
  • FIGS. 17A to 17C show that addition of anti-4-1BB to tumor fragments facilitates the expansion of more differentiated CD8+ TIL after 4 weeks of culture with IL-2.
  • FIGS. 18A to 18D show that addition of anti 4-1BB to tumor fragments increases killing ability of expanded TIL after 4 weeks of culture with IL-2.
  • FIGS. 19A to 19B show Perforin and Granzyme B expression in CD8+ TIL expanded from tumor fragments with or without added anti-4-1BB.
  • This approach can accelerate the rate of TIL expansion out of the tumor fragments and enrich the tumor-specific T cells at the same time.
  • additional antigenic stimulation that occurs during the early tumor fragment culture from antigen presentation can induce further T-cell co-stimulatory molecule expression that can be further used to enhance T-cell outgrowth.
  • Tumor fragments have been used for years to expand TIL by simply adding exogenous IL-2, but whether other immunomodulators can be added in these tumor fragment cultures to affect TIL expansion and phenotype has basically been ignored.
  • the 4-1BB co-stimulatory pathway can be exploited in melanoma tumor fragment cultures in an active rather than a passive way by activating 4-1BB signaling using a 4-1BB agonist to enhance the output of CD8 + T cells, their tumor reactivity, and memory properties to develop a practical way to improve the TIL therapy product.
  • 4-1BB expression can be seen on resident CD8 + T cells in melanoma metastases and is maintained during the culture of tumor fragments for at least a week in a significant frequency of CD8 + T cells in these early cultures ex vivo.
  • Tumor-infiltrating lymphocyte (TIL) production is a 2-step process: 1) the pre-REP (Rapid Expansion) stage where you the grow the cells in standard lab media such as RPMI and treat the TILs w/reagents such as irradiated feeder cells, and anti-CD3 antibodies to achieve the desired effect; and 2) the REP stage where you expand the TILs in a large enough culture amount for treating the patients.
  • the REP stage requires cGMP grade reagents and 30-40 L of culture medium.
  • the pre-REP stage can utilize lab grade reagents (under the assumption that the lab grade reagents get diluted out during the REP stage), making it easier to incorporate alternative strategies for improving TIL production. Therefore, in some embodiments, the disclosed TLR agonist and/or peptide or peptidomimetics can be included in the culture medium during the pre-REP stage.
  • ACT may be performed by (i) obtaining autologous lymphocytes from a mammal, (ii) culturing the autologous lymphocytes to produce expanded lymphocytes, and (ii) administering the expanded lymphocytes to the mammal.
  • the lymphocytes are tumor-derived, i.e. they are TILs, and are isolated from the mammal to be treated, i.e. autologous transfer.
  • Autologous ACT as described herein may also be performed by (i) culturing autologous lymphocytes to produce expanded lymphocytes; (ii) administering nonmyeloablative lymphodepleting chemotherapy to the mammal; and (iii) after administering nonmyeloablative lymphodepleting chemotherapy, administering the expanded lymphocytes to the mammal.
  • Autologous TILs may be obtained from the stroma of resected tumors. For this, tumor samples are obtained from patients and a single cell suspension is obtained.
  • the single cell suspension can be obtained in any suitable manner, e.g., mechanically (disaggregating the tumor using, e.g., a gentleMACSTM Dissociator, Miltenyi Biotec, Auburn, Calif.) or enzymatically (e.g., collagenase or DNase).
  • mechanically disaggregating the tumor using, e.g., a gentleMACSTM Dissociator, Miltenyi Biotec, Auburn, Calif.
  • enzymatically e.g., collagenase or DNase
  • T cells can be rapidly expanded using non-specific T-cell receptor stimulation in the presence of feeder lymphocytes and interleukin-2 (IL-2), IL-7, IL-15, IL-21, or combinations thereof.
  • the non-specific T-cell receptor stimulus can e.g. include around 30 ng/ml of OKT3, a mouse monoclonal anti-CD3 antibody (available from Ortho-McNeil®, Raritan, N.J. or Miltenyi Biotec, Bergisch Gladbach, Germany).
  • T cells can be rapidly expanded by stimulation of peripheral blood mononuclear cells (PBMC) in vitro with one or more antigens (including antigenic portions thereof, such as epitope(s), or a cell of the cancer, which can be optionally expressed from a vector, such as an human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., approximately 0.3 ⁇ M MART-1:26-35 (27 L) or gp100:209-217 (210M)), in the presence of a T-cell growth factor, such as around 200-400 ⁇ l/ml, such as 300 IU/ml IL-2 or IL-15, with IL-2 being preferred.
  • a vector such as an human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., approximately 0.3 ⁇ M MART-1:26-35 (27 L) or gp100:209-217 (210M)
  • HLA-A2 human leukocyte antigen A2
  • the in vitro-induced T-cells are rapidly expanded by re-stimulation with the same antigen(s) of the cancer pulsed onto HLA-A2-expressing antigen-presenting cells.
  • the T-cells can be re-stimulated with irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2, for example.
  • the autologous ACT method comprises enriching cultured TILs for CD8+ T cells prior to rapid expansion of the cells. Following culture of the TILs in IL-2, the T cells are depleted of CD4+ cells and enriched for CD8+ cells using, for example, a CD8 microbead separation (e.g., using a CliniMACS ⁇ plus>CD8 microbead system (Miltenyi Biotec)).
  • a CD8 microbead separation e.g., using a CliniMACS ⁇ plus>CD8 microbead system (Miltenyi Biotec)
  • a T-cell growth factor that promotes the growth and activation of the autologous T cells is administered to the mammal either concomitantly with the autologous T cells or subsequently to the autologous T cells.
  • the T-cell growth factor can be any suitable growth factor that promotes the growth and activation of the autologous T-cells.
  • T-cell growth factors examples include interleukin (IL)-2, IL-7, IL-15, IL-12 and IL-21, which can be used alone or in various combinations, such as IL-2 and IL-7, IL-2 and IL-15, IL-7 and IL-15, IL-2, IL-7 and IL-15, IL-12 and IL-7, IL-12 and IL-15, or IL-12 and IL2.
  • IL-2 and IL-7 interleukin
  • IL-7 and IL-15 examples include interleukin (IL-2 and IL-15, IL-7 and IL-15, IL-2, IL-7 and IL-15, IL-12 and IL-15, or IL-12 and IL2.
  • tumor fragments placed in culture to expand TIL for adoptive cell therapy are not static tissues, but small, dynamic tumor microenvironments that can be manipulated to alter the yield and phenotype of TILs being expanded for cell therapy as well as enrich for tumor reactivity and improved memory phenotype.
  • 4-1BB co-stimulation enhancement in this system can manipulate these ex vivo tumor microenvironments to expand optimally active TIL for adoptive cell therapy.
  • the method involves culturing tumor fragments from the subject in a culture medium containing IL-2 and a 41BB agonist in an amount effective to expand tumor-infiltrating lymphocytes with enriched tumor-reactivity and specificity.
  • the culture medium can further contain a checkpoint inhibitor, such as anti-PD-1 antibody (e.g., BMS 936558), anti-PD-L1 antibody (e.g., cloneM1H1), anti-CTLA-4 antibody (e.g., Ipilimumab, BMS), or any combination thereof.
  • Also disclosed is a method for treating a tumor in a subject that involves treating the subject with nonmyelo ablative lympho depleting chemotherapy, and administering tumor-infiltrating lymphocytes expanded by the disclosed methods.
  • the 4-1BB glycoprotein is a member of the tumor necrosis factor receptor superfamily and binds to a high-affinity ligand (4-1BBL) expressed on several antigen-presenting cells such as macrophages and activated B cells. Therefore, in some embodiments, the 4-1BB agonist is a 4-1BBL protein, or a fragment or variant thereof capable of ligating 4-1BB on T-cell.
  • the 4-1BB agonist is a recombinant protein, such as recombinant human 4-1BB.
  • the recombinant protein can have the native 4-1BBL sequence.
  • An example protein sequence for human 4-1BBL is provided in UniProtKB/Swiss-Prot Accession No. P41273.
  • the recombinant protein can also be a fragment, variant, or combination thereof so long as it is capable of ligating 4-1BB on T-cell.
  • the recombinant protein is a fusion protein.
  • Fusion proteins also known as chimeric proteins, are proteins created through the joining of two or more genes which originally coded for separate proteins. Translation of this fusion gene results in a single polypeptide with function properties derived from each of the original proteins.
  • Recombinant fusion proteins can be created artificially by recombinant DNA technology for use in biological research or therapeutics. Chimeric mutant proteins occur naturally when a large-scale mutation, typically a chromosomal translocation, creates a novel coding sequence containing parts of the coding sequences from two different genes.
  • fusion proteins are made possible by the fact that many protein functional domains are modular.
  • the linear portion of a polypeptide which corresponds to a given domain, such as a tyrosine kinase domain may be removed from the rest of the protein without destroying its intrinsic enzymatic capability.
  • any of the herein disclosed functional domains can be used to design a fusion protein.
  • a recombinant fusion protein is a protein created through genetic engineering of a fusion gene. This typically involves removing the stop codon from a cDNA sequence coding for the first protein, then appending the cDNA sequence of the second protein in frame through ligation or overlap extension PCR. That DNA sequence will then be expressed by a cell as a single protein.
  • the protein can be engineered to include the full sequence of both original proteins, or only a portion of either.
  • linker or “spacer” peptides are also added which make it more likely that the proteins fold independently and behave as expected.
  • linkers in protein or peptide fusions are sometimes engineered with cleavage sites for proteases or chemical agents which enable the liberation of the two separate proteins.
  • This technique is often used for identification and purification of proteins, by fusing a GST protein, FLAG peptide, or a hexa-his peptide (aka: a 6 ⁇ his-tag) which can be isolated using nickel or cobalt resins (affinity chromatography).
  • Chimeric proteins can also be manufactured with toxins or anti-bodies attached to them in order to study disease development.
  • IRES elements can be used to create multigene, or polycistronic, messages.
  • IRES elements are able to bypass the ribosome scanning model of 5′ methylated Cap dependent translation and begin translation at internal sites (Pelletier and Sonenberg, 1988).
  • IRES elements from two members of the picornavirus family polio and encephalomyocarditis have been described (Pelletier and Sonenberg, 1988), as well an IRES from a mammalian message (Macejak and Sarnow, 1991).
  • IRES elements can be linked to heterologous open reading frames. Multiple open reading frames can be transcribed together, each separated by an IRES, creating polycistronic messages.
  • IRES element By virtue of the IRES element, each open reading frame is accessible to ribosomes for efficient translation. Multiple genes can be efficiently expressed using a single promoter/enhancer to transcribe a single message (U.S. Pat. Nos. 5,925,565 and 5,935,819; PCT/US99/05781). IRES sequences are known in the art and include those from encephalomycarditis virus (EMCV) (Ghattas, I. R. et al., Mol. Cell.
  • EMCV encephalomycarditis virus
  • the 4-1BB agonist is an agonistic anti-4-1BB antibody capable of ligating 4-1BB on T-cells to induce its co-stimulatory activity.
  • Suitable antibodies include both polyclonal and monoclonal antibodies.
  • Antibodies that can be used in the disclosed compositions and methods include whole immunoglobulin (i.e., an intact antibody) of any class, fragments thereof, and synthetic proteins containing at least the antigen binding variable domain of an antibody.
  • the disclosed antibody can be a human antibody and/or a humanized antibody.
  • Many non-human antibodies e.g., those derived from mice, rats, or rabbits
  • are naturally antigenic in humans and thus can give rise to undesirable immune responses when administered to humans. Therefore, the use of human or humanized antibodies in the methods serves to lessen the chance that an antibody administered to a human will evoke an undesirable immune response.
  • a humanized anti-4-1BB antibody is in clinical trials in patients with solid tumors, including melanoma, renal carcinoma, and ovarian cancer, and so far seems to have a favorable toxicity profile.
  • IgA human immunoglobulins
  • IgD immunoglobulins
  • IgE immunoglobulins
  • IgG immunoglobulins
  • Native antibodies are usually heterotetrameric glycoproteins, composed of two identical light (L) chains and two identical heavy (H) chains.
  • each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes.
  • Each heavy and light chain also has regularly spaced intrachain disulfide bridges.
  • Each heavy chain has at one end a variable domain (V(H)) followed by a number of constant domains.
  • Each light chain has a variable domain at one end (V(L)) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
  • Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains.
  • the light chains of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (k) and lambda (l), based on the amino acid sequences of their constant domains.
  • immunoglobulins can be assigned to different classes.
  • variable is used herein to describe certain portions of the variable domains that differ in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen.
  • variability is not usually evenly distributed through the variable domains of antibodies. It is typically concentrated in three segments called complementarity determining regions (CDRs) or hyperyariable regions both in the light chain and the heavy chain variable domains.
  • CDRs complementarity determining regions
  • FR framework
  • the variable domains of native heavy and light chains each comprise four FR regions, largely adopting a b-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the b-sheet structure.
  • the CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the antigen binding site of antibodies.
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity.
  • fragments of antibodies which have bioactivity.
  • the fragments whether attached to other sequences or not, include insertions, deletions, substitutions, or other selected modifications of particular regions or specific amino acids residues, provided the activity of the fragment is not significantly altered or impaired compared to the nonmodified antibody or antibody fragment.
  • a single chain antibody can be created by fusing together the variable domains of the heavy and light chains using a short peptide linker, thereby reconstituting an antigen binding site on a single molecule.
  • Single-chain antibody variable fragments scFvs
  • the linker is chosen to permit the heavy chain and light chain to bind together in their proper conformational orientation.
  • Divalent single-chain variable fragments can be engineered by linking two scFvs. This can be done by producing a single peptide chain with two VH and two VL regions, yielding tandem scFvs. ScFvs can also be designed with linker peptides that are too short for the two variable regions to fold together (about five amino acids), forcing scFvs to dimerize. This type is known as diabodies. Diabodies have been shown to have dissociation constants up to 40-fold lower than corresponding scFvs, meaning that they have a much higher affinity to their target. Still shorter linkers (one or two amino acids) lead to the formation of trimers (triabodies or tribodies). Tetrabodies have also been produced. They exhibit an even higher affinity to their targets than diabodies.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a substantially homogeneous population of antibodies, i.e., the individual antibodies within the population are identical except for possible naturally occurring mutations that may be present in a small subset of the antibody molecules.
  • the antibodies can also be “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, as long as they exhibit the desired antagonistic activity.
  • In vitro methods are also suitable for preparing monovalent antibodies.
  • Digestion of antibodies to produce fragments thereof, particularly, Fab or F(ab) 2 fragments can be accomplished using routine techniques known in the art. For instance, digestion can be performed using papain. Papain digestion of antibodies typically produces two identical antigen binding fragments, called Fab fragments, each with a single antigen binding site, and a residual Fc fragment. Pepsin treatment yields an Fc fragment and an F(ab) 2 fragment that has two antigen combining sites and is still capable of cross-linking antigen.
  • the 4-1BB agonist is an agonistic aptamer capable of ligating 4-1BB on T-cells to induce its co-stimulatory activity.
  • aptamer refers to oligonucleic acid or peptide molecules that bind to a specific target molecule. These molecules are generally selected from a random sequence pool. The selected aptamers are capable of adapting unique tertiary structures and recognizing target molecules with high affinity and specificity.
  • a “nucleic acid aptamer” is a DNA or RNA oligonucleic acid that binds to a target molecule via its conformation, and thereby inhibits or suppresses functions of such molecule.
  • a nucleic acid aptamer may be constituted by DNA, RNA, or a combination thereof.
  • a “peptide aptamer” is a combinatorial protein molecule with a variable peptide sequence inserted within a constant scaffold protein. Identification of peptide aptamers is typically performed under stringent yeast dihybrid conditions, which enhances the probability for the selected peptide aptamers to be stably expressed and correctly folded in an intracellular context.
  • Nucleic acid aptamers are typically oligonucleotides ranging from 15-50 bases in length that fold into defined secondary and tertiary structures, such as stem-loops or G-quartets. Nucleic acid aptamers preferably bind the target molecule with a K d less than 10 ⁇ 6 , 10 ⁇ 8 , 10 ⁇ 10 , or 10 ⁇ 12 . Nucleic acid aptamers can also bind the target molecule with a very high degree of specificity.
  • Nucleic acid aptamers are typically isolated from complex libraries of synthetic oligonucleotides by an iterative process of adsorption, recovery and re-amplification.
  • nucleic acid aptamers may be prepared using the SELEX (Systematic Evolution of Ligands by Exponential Enrichment) method.
  • the SELEX method involves selecting an RNA molecule bound to a target molecule from an RNA pool composed of RNA molecules each having random sequence regions and primer-binding regions at both ends thereof, amplifying the recovered RNA molecule via RT-PCR, performing transcription using the obtained cDNA molecule as a template, and using the resultant as an RNA pool for the subsequent procedure.
  • the base sequence lengths of the random sequence region and the primer binding region are not particularly limited. In general, the random sequence region contains about 20 to 80 bases and the primer binding region contains about 15 to 40 bases. Specificity to a target molecule may be enhanced by prospectively mixing molecules similar to the target molecule with RNA pools and using a pool containing RNA molecules that did not bind to the molecule of interest. An RNA molecule that was obtained as a final product by such technique is used as an RNA aptamer.
  • Peptide aptamers are proteins that are designed to interfere with other protein interactions inside cells. They consist of a variable peptide loop attached at both ends to a scaffold. This double structural constraint greatly increases the binding affinity of the peptide aptamer to levels comparable to an antibody.
  • the variable loop length is typically composed of about ten to twenty amino acids, and the scaffold may be any protein which has good solubility.
  • the bacterial protein Thioredoxin-A is the most used scaffold protein, the variable loop being inserted within the reducing active site, the two Cysteines lateral chains being able to form a disulfide bridge.
  • Peptide aptamer selection can be made using different systems, but the most used is currently the yeast two-hybrid system.
  • Peptide aptamer can also be selected from combinatorial peptide libraries constructed by phage display and other surface display technologies such as mRNA display, ribosome display, bacterial display and yeast display. These experimental procedures are also known as biopannings. Among peptides obtained from biopannings, mimotopes can be considered as a kind of peptide aptamers. All the peptides panned from combinatorial peptide libraries have been stored in a special database with the name MimoDB.
  • the disclosed compounds can be formulated in a physiologically- or pharmaceutically-acceptable form and administered by any suitable route known in the art including, for example, oral, nasal, rectal, topical, and parenteral routes of administration.
  • parenteral includes subcutaneous, intradermal, intravenous, intramuscular, intraperitoneal, and intrasternal administration, such as by injection.
  • Administration of the disclosed compounds or compositions can be a single administration, or at continuous or distinct intervals as can be readily determined by a person skilled in the art.
  • the compounds disclosed herein, and compositions comprising them can also be administered utilizing liposome technology, slow release capsules, implantable pumps, and biodegradable containers. These delivery methods can, advantageously, provide a uniform dosage over an extended period of time.
  • the compounds can also be administered in their salt derivative forms or crystalline forms.
  • the compounds disclosed herein can be formulated according to known methods for preparing pharmaceutically acceptable compositions. Formulations are described in detail in a number of sources which are well known and readily available to those skilled in the art. For example, Remington's Pharmaceutical Science by E. W. Martin (1995) describes formulations that can be used in connection with the disclosed methods. In general, the compounds disclosed herein can be formulated such that an effective amount of the compound is combined with a suitable carrier in order to facilitate effective administration of the compound.
  • the compositions used can also be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspension, suppositories, injectable and infusible solutions, and sprays.
  • compositions also preferably include conventional pharmaceutically-acceptable carriers and diluents which are known to those skilled in the art.
  • carriers or diluents for use with the compounds include ethanol, dimethyl sulfoxide, glycerol, alumina, starch, saline, and equivalent carriers and diluents.
  • compositions disclosed herein can advantageously comprise between about 0.1% and 99%, and especially, 1 and 15% by weight of the total of one or more of the subject compounds based on the weight of the total composition including carrier or diluent.
  • Formulations suitable for administration include, for example, aqueous sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions, which can include suspending agents and thickening agents.
  • the formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a freeze dried (lyophilized) condition requiring only the condition of the sterile liquid carrier, for example, water for injections, prior to use.
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powder, granules, tablets, etc. It should be understood that in addition to the ingredients particularly mentioned above, the compositions disclosed herein can include other agents conventional in the art having regard to the type of formulation in question.
  • Compounds disclosed herein, and compositions comprising them can be delivered to a cell either through direct contact with the cell or via a carrier means.
  • Carrier means for delivering compounds and compositions to cells are known in the art and include, for example, encapsulating the composition in a liposome moiety.
  • Another means for delivery of compounds and compositions disclosed herein to a cell comprises attaching the compounds to a protein or nucleic acid that is targeted for delivery to the target cell.
  • U.S. Pat. No. 6,960,648 and U.S. Application Publication Nos. 20030032594 and 20020120100 disclose amino acid sequences that can be coupled to another composition and that allows the composition to be translocated across biological membranes.
  • compositions for transporting biological moieties across cell membranes for intracellular delivery can also be incorporated into polymers, examples of which include poly (D-L lactide-co-glycolide) polymer for intracranial tumors; poly[bis(p-carboxyphenoxy) propane:sebacic acid] in a 20:80 molar ratio (as used in GLIADEL); chondroitin; chitin; and chitosan.
  • the compounds disclosed herein can be administered to a patient in need of treatment in combination with other antitumor or anticancer substances and/or with radiation and/or photodynamic therapy and/or with surgical treatment to remove a tumor.
  • these other substances or treatments can be given at the same as or at different times from the compounds disclosed herein.
  • the compounds disclosed herein can be used in combination with mitotic inhibitors such as taxol or vinblastine, alkylating agents such as cyclophosamide or ifosfamide, antimetabolites such as 5-fluorouracil or hydroxyurea, DNA intercalators such as adriamycin or bleomycin, topoisomerase inhibitors such as etoposide or camptothecin, antiangiogenic agents such as angiostatin, antiestrogens such as tamoxifen, and/or other anti-cancer drugs or antibodies, such as, for example, GLEEVEC (Novartis Pharmaceuticals Corporation) and HERCEPTIN (Genentech, Inc.), respectively, or an immunotherapeutic such as ipilimumab and bortezomib.
  • mitotic inhibitors such as taxol or vinblastine
  • alkylating agents such as cyclophosamide or ifosfamide
  • antimetabolites such as 5-fluorouracil or hydroxyure
  • compounds and compositions disclosed herein can be locally administered at one or more anatomical sites, such as sites of unwanted cell growth (such as a tumor site or benign skin growth, e.g., injected or topically applied to the tumor or skin growth), optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent.
  • a pharmaceutically acceptable carrier such as an inert diluent
  • Compounds and compositions disclosed herein can be systemically administered, such as intravenously or orally, optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent, or an assimilable edible carrier for oral delivery. They can be enclosed in hard or soft shell gelatin capsules, can be compressed into tablets, or can be incorporated directly with the food of the patient's diet.
  • the active compound can be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, aerosol sprays, and the like.
  • the tablets, troches, pills, capsules, and the like can also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring can be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound can be incorporated into sustained-release preparations and devices.
  • compositions disclosed herein can be administered intravenously, intramuscularly, or intraperitoneally by infusion or injection.
  • Solutions of the active agent or its salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations can contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient, which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various other antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, buffers or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the inclusion of agents that delay absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating a compound and/or agent disclosed herein in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • compounds and agents disclosed herein can be applied in as a liquid or solid. However, it will generally be desirable to administer them topically to the skin as compositions, in combination with a dermatologically acceptable carrier, which can be a solid or a liquid.
  • a dermatologically acceptable carrier which can be a solid or a liquid.
  • Compounds and agents and compositions disclosed herein can be applied topically to a subject's skin to reduce the size (and can include complete removal) of malignant or benign growths, or to treat an infection site.
  • Compounds and agents disclosed herein can be applied directly to the growth or infection site.
  • the compounds and agents are applied to the growth or infection site in a formulation such as an ointment, cream, lotion, solution, tincture, or the like.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers, for example.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Useful dosages of the compounds and agents and pharmaceutical compositions disclosed herein can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art.
  • compositions that comprise a compound disclosed herein in combination with a pharmaceutically acceptable carrier.
  • Pharmaceutical compositions adapted for oral, topical or parenteral administration, comprising an amount of a compound constitute a preferred aspect.
  • the dose administered to a patient, particularly a human should be sufficient to achieve a therapeutic response in the patient over a reasonable time frame, without lethal toxicity, and preferably causing no more than an acceptable level of side effects or morbidity.
  • dosage will depend upon a variety of factors including the condition (health) of the subject, the body weight of the subject, kind of concurrent treatment, if any, frequency of treatment, therapeutic ratio, as well as the severity and stage of the pathological condition.
  • Adoptive cell transfer is a very effective form of immunotherapy and involves the transfer of immune cells with antitumor activity into cancer patients.
  • ACT is a treatment approach that involves the identification, in vitro, of lymphocytes with antitumor activity, the in vitro expansion of these cells to large numbers and their infusion into the cancer-bearing host.
  • Lymphocytes used for adoptive transfer can be derived from the stroma of resected tumors (tumor infiltrating lymphocytes or TILs).
  • ACT in which the lymphocytes originate from the cancer-bearing host to be infused is termed autologous ACT.
  • US 2011/0052530 relates to a method for performing adoptive cell therapy to promote cancer regression, primarily for treatment of patients suffering from metastatic melanoma, which is incorporated by reference in its entirety for these methods.
  • nonmyeloablative lymphodepleting chemotherapy is administered to the mammal prior to administering to the mammal the expanded tumor-infiltrating lymphocytes.
  • the purpose of lymphodepletion is to make room for the infused lymphocytes, in particular by eliminating regulatory T cells and other non-specific T cells which compete for homeostatic cytokines.
  • Nonmyeloablative lymphodepleting chemotherapy can be any suitable such therapy, which can be administered by any suitable route known to a person of skill.
  • the nonmyeloablative lymphodepleting chemotherapy can comprise, for example, the administration of cyclophosphamide and fludarabine, particularly if the cancer is melanoma, which can be metastatic.
  • a preferred route of administering cyclophosphamide and fludarabine is intravenously.
  • any suitable dose of cyclophosphamide and fludarabine can be administered.
  • around 40-80 mg/kg, such as around 60 mg/kg of cyclophosphamide is administered for approximately two days after which around 15-35 mg/m 2 , such as around 25 mg/m 2 fludarabine is administered for around five days, particularly if the cancer is melanoma.
  • expanded lymphocytes produced by the disclosed methods are administered as an intra-arterial or intravenous infusion, which preferably lasts about 30 to about 60 minutes.
  • routes of administration include intraperitoneal, intrathecal and intralymphatic.
  • any suitable dose of lymphocytes can be administered. In one embodiment, about 1 ⁇ 10 10 lymphocytes to about 15 ⁇ 10 10 lymphocytes are administered.
  • compositions and methods can be used in combination with other cancer immunotherapies.
  • immunotherapy uses components of the immune system to direct targeted cytotoxic activity against cancer cells, without necessarily initiating an immune response in the patient, while active immunotherapy actively triggers an endogenous immune response.
  • Passive strategies include the use of the monoclonal antibodies (mAbs) produced by B cells in response to a specific antigen.
  • mAbs monoclonal antibodies
  • mAbs have been the biggest success story for immunotherapy; the top three best-selling anticancer drugs in 2012 were mAbs.
  • rituximab (Rituxan, Genentech), which binds to the CD20 protein that is highly expressed on the surface of B cell malignancies such as non-Hodgkin's lymphoma (NHL).
  • NHL non-Hodgkin's lymphoma
  • CLL chronic lymphocytic leukemia
  • trastuzumab Herceptin; Genentech
  • HER2 human epidermal growth factor receptor 2
  • therapeutic cancer vaccines In order to actively drive an antitumor immune response, therapeutic cancer vaccines have been developed. Unlike the prophylactic vaccines that are used preventatively to treat infectious diseases, therapeutic vaccines are designed to treat established cancer by stimulating an immune response against a specific tumor-associated antigen.
  • sipuleucel-T Provenge; Dendreon Corporation
  • IMPACT Immunotherapy Prostate Adenocarcinoma Treatment
  • the advantage of active immunotherapies is that they have the potential to provide long-lasting anticancer activity by engaging both the innate and adaptive arms of the immune response. While mAbs are typically considered passive immunotherapies, there is increasing evidence that they also induce an adaptive immune response via a “vaccination-like” effect.
  • Generating optimal “killer” CD8 T cell responses also requires T cell receptor activation plus co-stimulation, which can be provided through ligation of tumor necrosis factor receptor family members, including OX40 (CD134) and 4-1BB (CD137).
  • OX40 is of particular interest as treatment with an activating (agonist) anti-OX40 mAb augments T cell differentiation and cytolytic function leading to enhanced anti-tumor immunity against a variety of tumors.
  • anti-cancer drugs are available for combination with the present method and compositions.
  • the following is a non-exhaustive lists of anti-cancer (anti-neoplastic) drugs that can be used in conjunction with irradiation: Acivicin; Aclarubicin; Acodazole Hydrochloride; AcrQnine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Car
  • the cancer treated by the disclosed compositions and methods can in some aspects be any solid tumor for which TILs can be produced.
  • the cancer can also be metastatic and/or recurrent.
  • Non-limiting examples of cancers include acute lymphocytic cancer, acute myeloid leukemia, alveolar rhabdomyosarcoma, bone cancer, brain cancer, breast cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the intrahepatic bile duct, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, cancer of the vulva, chronic lymphocytic leukemia, chronic myeloid cancer, cervical cancer, glioma, Hodgkin lymphoma, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, lung cancer, malignant mesothelioma, melanoma, multiple myeloma, nasopharyn
  • TNBCs Triple Negative Breast Cancers
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2 ERBB2
  • TNBCs do not respond to hormonal therapy such as tamoxifen or aromatase inhibitors or therapies that target HER2 receptors, such as Herceptin (trastuzumab).
  • Herceptin Herceptin
  • the cancer is a triple negative breast cancer (TNBC).
  • subject refers to any individual who is the target of administration or treatment.
  • the subject can be a vertebrate, for example, a mammal.
  • the subject can be a human or veterinary patient.
  • patient refers to a subject under the treatment of a clinician, e.g., physician.
  • terapéuticaally effective refers to the amount of the composition used is of sufficient quantity to ameliorate one or more causes or symptoms of a disease or disorder. Such amelioration only requires a reduction or alteration, not necessarily elimination.
  • treatment refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder.
  • This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder.
  • this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder.
  • inhibitor refers to a decrease in an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This may also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
  • hypoplastic cell or “hyperplasm” refers to a cell undergoing physiological (normal) cell proliferation (“hyperplasia”).
  • Neoplasm refers to a cell undergoing abnormal cell proliferation (“neoplasia”). The growth of neoplastic cells exceeds and is not coordinated with that of the normal tissues around it. The growth typically persists in the same excessive manner even after cessation of the stimuli, and typically causes formation of a tumor. Neoplasms may be benign, premalignant, or malignant.
  • cancer or “malignant neoplasm” refers to a cell that displays uncontrolled growth, invasion upon adjacent tissues, and often metastasis to other locations of the body.
  • tumor refers to an abnormal mass of tissue containing neoplastic cells.
  • tumor refers to the spread of malignant tumor cells from one organ or part to another non-adjacent organ or part. Cancer cells can “break away,” “leak,” or “spill” from a primary tumor, enter lymphatic and blood vessels, circulate through the bloodstream, and settle down to grow within normal tissues elsewhere in the body. When tumor cells metastasize, the new tumor is called a secondary or metastatic cancer or tumor.
  • antibody refers to natural or synthetic antibodies that selectively bind a target antigen.
  • the term includes polyclonal and monoclonal antibodies.
  • fragments or polymers of those immunoglobulin molecules, and human or humanized versions of immunoglobulin molecules that selectively bind the target antigen are fragments or polymers of those immunoglobulin molecules, and human or humanized versions of immunoglobulin molecules that selectively bind the target antigen.
  • peptide “protein,” and “polypeptide” are used interchangeably to refer to a natural or synthetic molecule comprising two or more amino acids linked by the carboxyl group of one amino acid to the alpha amino group of another.
  • peptidomimetic means a mimetic of a peptide which includes some alteration of the normal peptide chemistry. Peptidomimetics typically enhance some property of the original peptide, such as increase stability, increased efficacy, enhanced delivery, increased half life, etc. Use of peptidomimetics can involve the incorporation of a non-amino acid residue with non-amide linkages at a given position.
  • One embodiment of the present invention is a peptidomimetic wherein the compound has a bond, a peptide backbone or an amino acid component replaced with a suitable mimic.
  • unnatural amino acids which may be suitable amino acid mimics include ⁇ -alanine, L- ⁇ -amino butyric acid, L- ⁇ -amino butyric acid, L- ⁇ -amino isobutyric acid, L- ⁇ -amino caproic acid, 7-amino heptanoic acid, L-aspartic acid, L-glutamic acid, N- ⁇ -Boc-N- ⁇ -CBZ-L-lysine, N- ⁇ -Boc-N- ⁇ -Fmoc-L-lysine, L-methionine sulfone, L-norleucine, L-norvaline, N- ⁇ -Boc-N- ⁇ CBZ-L-ornithine, N- ⁇ -Boc-N- ⁇ -CBZ-L-ornithine, Boc-p-nitro-L-phenylalanine, Boc-hydroxyproline, and Boc-L-thioproline.
  • TIL tumor infiltrating lymphocyte
  • regression does not necessarily imply 100% or complete regression. Rather, there are varying degrees of regression of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. The term also encompasses delaying the onset of the disease, or a symptom or condition thereof.
  • a fully-human IgG4 monoclonal agonistic anti-4-1BB antibody (BMS 663513 Lot 6A20383/1187261) was provided by Bristol Myers Squibb at a concentration of 14.4 mg/ml and subsequently stored in aliquots at a concentration of 1 mg/ml at ⁇ 80° C.
  • the anti-4-1BB antibody was added at day 0 of the fragment set up at different concentrations and subsequently each time the TILs were fed. A more detailed description of the fragment set up is described below. 10 ⁇ g/ml was found to be the optimal dose and this concentration used for the remainder of the experiments.
  • tumor fragments were cultured in the presence of 10 ⁇ g/ml anti-human PD-1 antibody (BMS 936558), anti-human PD-L1 antibody (cloneM1H1) (eBioscience, San Diego, Calif.), or 10 ⁇ g/ml anti-human CTLA-4 antibody (Ipilimumab, BMS) in some experiments.
  • the TIL-CM contained RPMI 1640 with Glutamax (Gibco/Invitrogen; Carlsbad, Calif.), 1 ⁇ Pen-Strep (Gibco/Invitrogen; Carlsbad, Calif.), 50 ⁇ m 2-mercaptoethanol (Gibco/Invitrogen; Carlsbad, Calif.), 20 ⁇ g/ml Gentamicin (Gibco/Invitrogen; Carlsbad, Calif.), and 1 mM pyruvate (Gibco/Invitrogen; Carlsbad, Calif.). Each well contained 1 fragment and the fragments were kept in culture for a period of 3 weeks.
  • IL-2 For control cultures, the fragments were placed in 6,000 IU/ml IL-2 with a fully-human IgG4 Isotype control (Eureka therapeutics ET904) in some experiments.
  • the control cultures are referred to as ‘IL-2’ in this Example.
  • the TIL isolated after 3 weeks from the tumor fragment cultures were subjected to secondary expansion using a Rapid Expansion Protocol (REP).
  • the TIL were washed in TIL-CM for 5 minutes at 1400 rpm and 1.3 ⁇ 10 5 cells were placed in a T25 flask with 10 ml TIL-CM and 10 ml AIM-V, 30 ng/ml anti-CD3 (OKT3) and 26 ⁇ 10 6 gamma-irradiated (2,500 Rads) allogeneic PBMC referred to as ‘feeder cells’.
  • TIL-2 On day 2 of the REP, 3,000 IU/ml IL-2 was added to the cultures and subsequently each time the TIL were fed (day 5, 7, 9, and 12) with fresh media in order to maintain the cultures between 1-2 ⁇ 10 6 viable cells per ml. After the 2 week expansion, the TIL were referred as ‘post-REP’ TIL.
  • FWB Melanoma fragments were cut up into 4-6 mm 2 pieces and mechanically disaggregated using glass slides, filtered, re-suspended in 2 ml sterile FACS Wash Buffer (FWB), and washed for 5 minutes at 1400 rpm.
  • the FWB contained 1 ⁇ Dulbecco's Phosphate Buffered Saline (D-PBS) and 1% Bovine Serum Albumin (BSA) (Sigma Aldrich).
  • D-PBS Dulbecco's Phosphate Buffered Saline
  • BSA Bovine Serum Albumin
  • the cells that grew out of the fragment were also collected and re-suspended in 2 ml FWB and washed for 5 min at 1400 rpm. The cells were then stained with fluorochrome-conjugated monoclonal antibodies that recognized surface markers CD11c, CD80, CD86, 4-1BB, 4-1BBL, HLA-DR, CD3, CD8, CD56, CD19, CD20, CD4, (eBioscience, BD Biosciences, and BD Pharmingen) and AmCyan Aqua live/dead fixable dye (Molecular Probes by Life Technologies; Lot 1413034) in 0.1 ml FACS Stain Buffer (FSB) for 25 minutes on ice.
  • FACS Stain Buffer FACS Stain Buffer
  • the FSB contained 1 ⁇ D-PBS, 1% BSA, and 5% goat serum.
  • the cells were then washed in FWB for 5 minutes at 1400 rpm and fixed in 1 ⁇ D-PBS, 1% para-formaldehyde solution.
  • the cells were then acquired using a BD FACScanto II flow cytometer machine using FACSDiva software.
  • Tumors were cut up into multiple fragments, placed in TIL-CM, IL-2 ⁇ anti-4-1BB antibody. After 1 week, the fragments and the cells that migrated out of the tumor fragments were harvested. The fragments were mechanically disaggregated and filtered. The fragments and the cells that migrated out of the fragment were combined, washed with FWB, and subsequently stained for cell surface markers CD3 FITC (BD Biosciences; Catalog number 555916; Lot 17763) and CD8 Pacific Blue (BD Pharmingen; Catalog number 558207; Clone RPA-T8) on ice for 25 minutes. After the surface staining was done, the cells were washed in 2 ml FWB two times.
  • CD3 FITC BD Biosciences; Catalog number 555916; Lot 17763
  • CD8 Pacific Blue BD Pharmingen; Catalog number 558207; Clone RPA-T8
  • the supernatant was aspirated and 1 ml fixation buffer (BD Bioscience; Catalog 554655) was added to each tube and the tubes were placed at room temperature (RT) for 20 minutes. After 20 minutes, the cells were washed with FWB for 5 minutes at 1400 rpm, 4° C.
  • the supernatant was aspirated and 100 ⁇ l Perm Buffer III (BD; Catalog 558050; Lot 2128930) was added to each tube while vortexing each tube. The tubes were then placed on ice for 30 minutes and washed twice in 1 ml FWB for 5 minutes, 1400 rpm, 4° C.
  • the cells were then stained using anti-NF ⁇ B (p65) Alexa 647 (BD Pharmingen; Catalog number 560335) or anti-I ⁇ Ba Alexa 647 (BD Pharmingen; Catalog number 560817) and left in the dark at RT for 1 hour.
  • the cells were then washed with 3 ml FWB for 5 minutes, 1400 rpm, 4° C., then re-suspended in 300 ⁇ l FWB and samples were subsequently acquired using a BD FACScanto II flow cytometer machine.
  • the melanoma tumors were cut up into multiple fragments and placed in a 24-well plate with TIL-CM, IL-2 ⁇ anti-4-1BB antibody. After 3 weeks, the TIL were harvested, washed twice in media for 5 minutes at 1400 rpm at 4° C. After, 250,000 TIL per well were placed in a 48-well plate and kept at 37° C. for 3-4 hours. After the 3-4 hour incubation period, 200 IU/ml IL-2 was added to the cultures. As a control, half the cultures were not given IL-2. The plate was then placed in 37° C. for 20 minutes. The flow cytometry tubes were labeled and 1 ml fixation buffer was added to each tube.
  • the TIL were then added to the tubes, re-suspended and placed in 37° C. incubator for 10 minutes. After 10 minutes, the cells were spun for 5 minutes at 1400 rpm at 4° C. After the wash, the supernatant was aspirated and 1 ml Perm Buffer III was added to the tubes, re-suspended and placed in 4° C. for 20 minutes. The cells were then washed twice in 1 ml stain buffer for 5 minutes at 1400 rpm, 4° C. The cells were then stained with antibodies against CD8, pSTAT5, and CD3 in a total volume of 100 ⁇ l for 30 minutes at RT.
  • the cells were washed with 3 ml stain buffer for 5 minutes, 1400 rpm at 4° C., then re-suspended with 300 ⁇ l stain buffer and the samples were acquired using a BD FACScanto II flow cytometer machine.
  • Fragments from melanoma tumors were set up as described earlier with or without the addition of agonistic anti-4-1BB antibody.
  • the TIL were harvested and stained for surface and intracellular markers using flow cytometry. Briefly, the TIL were harvested, counted, and washed with 2 ml FWB for 5 minutes at 1400 rpm. The cells were stained with surface markers CD3, CD4, CD25, CD27, CD28, and CD8 (eBiosciences, BD Pharmingen, and BD Biosciences).
  • the viability dye AmCyan Aqua was added to gate out dead cells while acquiring the stained samples. The cells were stained in FSB for 25 minutes on ice.
  • the cells were then washed with 2 ml FWB and BD Cytofix/Cytoperm Fixation and Permeabilization Solution (BD Biosciences Cat: 554722) (used for Granzyme B, bcl-2, and EOMES) or Perm Buffer III (BD Biosciences Cat: 558050) was used for bcl-6.
  • the buffers were added to the samples for 20 minutes at RT in the dark.
  • the cells were washed with 1 ⁇ BD Perm/Wash Buffer (Cat: 554723) for 5 minutes at 1400 rpm, followed by intracellular staining for Foxp3, Granzyme B, bcl-2, EOMES, and bcl-6 (eBiosciences, BD Biosciences) in 1 ⁇ Fixation Buffer for 25 minutes on ice.
  • the samples were then washed with FWB for 5 minutes at 1400 rpm and fixed in 1 ⁇ D-PBS, 1% para-formaldehyde solution.
  • the samples were acquired using a BD FACScanto II flow cytometer machine.
  • harvested cells after 3 weeks of culture were stained for cell surface markers together with a MART-1 peptide HLA-A0201 tetramer (Beckman-Coulter) to enumerate the frequency of CD8 + MART-1-specific T cells.
  • TIL TIL media
  • IL-2 IL-2
  • anti-4-1BB antibody an anti-4-1BB antibody
  • the TIL were set up at a 1:1 ratio with HLA-matched melanoma tumor lines in a 96-well plate for 1 hour at 37° C.
  • Golgi-Stop BD Biosciences Catalog 554724
  • CD107a antibody BD Biosciences
  • the cells were then stained for surface markers CD3 and CD8 for 25 minutes at 4° C. After, the cells were washed with FWB again, re-suspended in fixation buffer (added while vortexing the tubes) and incubated for 15 minutes at room temperature. After, the cells were washed with FWB and re-suspended in Cytofix/Cytoperm buffer and incubated at room temperature for 20 minutes. The cells were then washed with FWB and stained for IFN-gamma for 25 minutes in 4° C. The cells were then washed again and the samples were acquired using a BD FACScanto II flow cytometer machine.
  • the tumor fragments were set up as previously described with or without anti-4-1BB antibody. After 1 week in culture, the cells were collected and washed twice in 1 ⁇ D-PBS for 5 minutes at 1400 rpm. The cells were re-suspended in 1 ml 4% para-formaldehyde for 20 minutes at RT. The cells were washed again in 1 ⁇ D-PBS and re-suspend in 1 ml and placed at 4° C. The washed cells were cytospun onto glass slides at 750 rpm for 3 minutes using Shandon Cytospin II centrifuge.
  • NF ⁇ B (p65) staining analysis was also done using the LAS V4.2 software, but additional analysis was conducted using Vectra Intelligent Slide Analysis System (Vectra, Perkin Elmer) Nuance software 3.0.1.2, using composite coloring style “Fluorescence.”
  • the melanoma tumor was surgically resected and cut up into multiple fragments.
  • the fragments were then placed in culture with an anti-HLA-ABC blocking antibody (anti-HLA antibody) at a concentration of 80 ⁇ g/ml for 3 hours at 37° C.
  • IL-2 alone or IL-2 plus anti-4-1BB antibody (10 ⁇ g/ml) was then added.
  • the cultures were subsequently fed with additional anti-MHC antibody once more and all cultures were fed over a 3 week period with media, IL-2 ⁇ anti-4-1BB antibody.
  • the viable cells after 3 weeks were counted and stained for CD3 and CD8 using flow cytometry.
  • TIL tumor target cells from autologous or HLA-A-matched melanoma tumor cell lines (1 ⁇ 10 5 TIL and 1 ⁇ 10 5 tumor targets).
  • Tumor target cells from HLA-A unmatched tumor cell lines were used as negative controls for specificity. Triplicate wells were set up for each condition.
  • the supernatants were collected after 24 hr and IFN-gamma secretion was measured using a human IFN-gamma ELISA kit (Thermo Scientific KB132422).
  • a 96-well ELISA plate reader (ELx808, Bio-Tek Instruments Inc., Houston, Tex.) was used to read the plate.
  • mice Six to eight week-old C57BL/6 mice were purchased from Harlan Laboratories Indianapolis, Ind.) and were housed at the Animal Research Facility of the H. Lee Moffitt Cancer Center and Research Institute. All animal protocols were reviewed and approved by the Institutional Animal Care and Use Committee at the University of South Florida.
  • B16 melanoma cancer cells were maintained by serial in vitro passages in complete medium (CM) consisting of RPMI 1640 supplemented with 10% heat-inactivated FCS, 0.1 mM non-essential amino acids, 1 mM sodium pyruvate, 2 mM fresh L-glutamine, 100 ⁇ g/ml streptomycin, 100 U/mL penicillin, 50 ⁇ g/ml gentamycin, 0.5 ⁇ g/mL fungizone (all from Life Technologies, Rockville, Md.), and 0.05 mM 2-ME (Sigma-Aldrich, St. Louis, Mo.). A total of 1 ⁇ 10 5 B16 cells were injected subcutaneously (s.c.).
  • mice received 20 mg/kg of rat IgG (rIgG) or anti-mouse PD-1 antibodies (clone RMP1-14, BioXcell, West Lebanon, N.H.) intraperitoneally (i.p.) every 3 days. Tumors were collected on days 21-24. Tumor cell suspensions were prepared from solid tumors by enzymatic digestion in HBSS (Life Technologies) containing 1 mg/ml collagenase, 0.1 mg/ml DNAse I, and 2.5 U/ml of hyaluronidase (all from Sigma-Aldrich) with constant stirring for 2 hours at RT.
  • HBSS Life Technologies
  • TIL TIL-1 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ IL-2 ⁇ g/ml non-specific control IgG or anti-mouse 4-1BB antibodies (clone 3H3, BioXcell). On day 7, TIL were collected and co-cultured with MC-38 or B16 cells at a 10:1 ratio. After 24 hours, supernatants were collected and IFN-gamma secretion was measured by ELISA.
  • 4-1BB is expressed on freshly isolated melanoma tumor-infiltrating lymphocytes and after 1 week of tumor fragment culture.
  • Some CD3 + CD8 + TIL expanded from tumor fragments expressed 4-1BB [Hernandez-Chacon, J A, et al (2011) J Immunother 34(3):236-50], so experiments were conducted to determine whether 4-1BB was expressed on the freshly isolated TIL from melanoma tumors used to derive tumor fragment cultures.
  • Melanoma tumors were surgically resected from metastatic melanoma patients and single cell suspensions were prepared and stained for 4-1BB on the CD3 + CD8 + subset.
  • 4-1BB was mainly expressed on CD8 + TIL, whereas OX40 was also expressed but to a lesser extent ( FIG. 1A ).
  • the expression of 4-1BB and OX40 was also examined on CD3 + CD8 + subset after the TIL were in culture for 1 week ( FIG. 1B ).
  • Some TIL still expressed 4-1BB even after 1 week of culture, whereas OX40 was also expressed but at lower levels as expected on CD8 + T cells ( FIG. 1B ).
  • 4-1BB is expressed on CD8 + T cells in tumor tissue ex vivo with a population of 4-1BB + cells still detectable after a week of culturing the tumor fragments.
  • Agonistic Anti-4-1BB Antibody Increases TIL Expansion In Vitro.
  • anti-4-1BB To further test the effects of anti-4-1BB, experiments were performed on an additional seven tumor samples (28 separate tumor fragments), which revealed this enhanced outgrowth of TIL after 3 weeks from a consistent phenomenon in a larger sampling of patient tumors ( FIG. 2D ). Experiments were then conducted to determine how anti-4-1BB facilitates TIL outgrowth compared to three other immunomodulatory (checkpoint) antibodies, anti-PD-1, anti-PD-L1, and anti-CTLA-4 at the same dose, currently being used in clinical trials and standard of care for melanoma (anti-CTLA-4). As shown in FIG. 2D , anti-4-1BB antibody was superior in inducing TIL outgrowth from the tumor fragments compared to IL-2 alone or IL-2 plus these other checkpoint antibodies.
  • FIG. 3B Staining for NF ⁇ B in TILs from 3 different patient tumor samples found a consistent increase in translocated p65 in cultures treated with anti-4-1BB, after quantifying the staining using this pseudo-immunofluorescent imaging approach ( FIG. 3B ).
  • fragments were isolated 1 week after culture initiation and stained for NF ⁇ B (p65) and I ⁇ B ⁇ in the TILs using flow cytometry after the isolated fragments were mechanically disaggregated into single cell suspensions using frosted glass. After gating on viable CD3 + CD8 + T cells, an increase in NF ⁇ B (p65) staining ( FIG.
  • the phenotype of the CD8 + TIL expanded from tumor fragments in the presence of anti-4-1BB was characterized further.
  • Melanoma tumor fragments were cultured as before with or without anti-4-1BB antibody.
  • the TIL were stained using flow cytometry for effector-memory markers (CD28 and CD27), markers of effector CTL (Granzyme B and Eomesodermin), and T-cell survival/memory markers (bcl-2 and bcl-6) [Huang, J, et al (2006) J Immunol 176(12):7726-35; Powell, D J, et al (2005) Blood 105(1):241-50; Ichii, H, et al (2004) J Immunol 173(2):883-91; Ichii, H, et al (2002) Nat Immunol 3(6):558-63; Pearce, E L, et al (2003) Science 302(5647):1041-3; Li, G, et al (2013) PLoS One 8
  • CD28 expression was unaltered ( FIG. 5A ), but CD27 expression decreased somewhat ( FIG. 5B ). This was associated with an increase in CD70 expression (ligand for CD27) on the harvested CD8 + TIL.
  • BTLA another marker on CD8 + TIL associated with clinical response to TIL therapy in melanoma patients [Radvanyi, L G, et al (2012) Clin Cancer Res 18(24):6758-70], was also unaltered.
  • Other T-cell negative co-stimulatory molecules, such as PD-1 were also unaltered.
  • TILs that grew out of tumor fragments over the 3-week period with anti-4-1BB co-stimulation could further expand after re-stimulation through the T-Cell Receptor (TCR) in a REP.
  • TCR T-Cell Receptor
  • 4-1BB co-stimulation during the initial TIL outgrowth did not affect the fold-expansion of TIL after TCR re-stimulation in the REP ( FIG. 9A ) and did not alter the phenotype of the resulting post-REP cells in terms of CD8, CD28, and CD27 expression, although there was a trend towards an increased frequency of CD8 + T cells ( FIG. 9B-E ).
  • Granzyme B is a key cytolytic granule molecule in CD8 + T cells, while Eomes is a T-box transcription factors helping drive Granzyme B expression as well as being involved in memory T-cell maintenance [Ichii, H, et al (2004) J Immunol 173(2):883-91; Ichii, H, et al (2002) Nat Immunol 3(6):558-63; Pearce, E L, et al (2003) Science 302(5647):1041-3].
  • Both bcl-2 and bcl-6 are cell survival (anti-apoptotic) molecules in memory T cells, with bcl-6 being also key transcription factor in the maintenance of memory status that inhibits Blimp-1 expression and terminal CD8 + T-cell differentiation [Ichii, H, et al (2004) J Immunol 173(2):883-91; Ichii, H, et al (2002) Nat Immunol 3(6):558-63; Yoshida, K, et al (2006) Eur J Immunol 36(12):3146-56; Scheeren, F A, et al (2005) Nat Immunol 6(3): 303-13].
  • IL-2 signaling could have been altered.
  • ICS assays were performed for IFN-gamma production on a single cell level using flow cytometry of TIL isolated after 3 weeks from fragment cultures from HLA-A2 + patients using HLA-A2 + melanoma tumor targets, revealing a significant increase in the frequency in IFN-gamma-positive CD8 + T cells in TIL isolated from fragment cultures treated with anti-4-1BB antibody ( FIG. 6B ; data from 11 experiments shown).
  • the frequency of CD107a release (a measure for target-specific T-cell degranulation) was also measured using flow cytometry.
  • TIL expanded with IL-2 plus anti-4-1BB versus IL-2 alone exhibited an increase in CD107a + frequency after co-incubation with HLA-A-matched tumor targets in the CD3 + CD8 + subset ( FIG. 6C ; data from 6 patients).
  • TIL that grew out from the fragments for CD8 and MART-1-specific TCR expression was stained using MART-1 peptide tetramers, revealing a marked increase in the frequency of MART-1-specific T cells when anti-4-1BB was added to the tumor fragment cultures ( FIG. 6D ).
  • TILs originally expanded from tumor fragments with or without added anti-4-1BB after secondary rapid expansion (REP) using anti-CD3 plus IL-2 were analyzed for tumor-specific reactivity.
  • an enhanced anti-tumor reactivity of the post-REP product was found when 4-1BB co-stimulation was provided in the initial tumor fragment cultures ( FIG. 6E ).
  • provision of agonist anti-4-1BB antibodies to the initial melanoma tumor fragment cultures significantly increases the outgrowth of tumor-specific CD8 + T cells for adoptive cell therapy.
  • mice were challenged with subcutaneous B16 melanoma, the tumors isolated, and the isolated cells cultured with IL-2 with or without an agonist anti-mouse 4-1BB antibody (clone 3H3) for seven days.
  • TILs grown with the anti-4-1BB antibody exhibited increased the anti-tumor reactivity of the expanded TILs, as determined using IFN-gamma-release after co-culture with B16 tumor targets ( FIG. 10A ).
  • PD-1 blockade has been shown to facilitate anti-tumor T-cell priming, increase T-cell infiltration into tumors and anti-tumor effector function [Peng, W, et al (2012) Cancer Res 72(20):5209-18; Pilon-Thomas, S, et al (2010) J Immunol 184(7): 3442-9].
  • Clinical trials in melanoma have shown increased T-cell infiltration into tumors with an impressive 30% clinical response rate [Topalian, S L, et al (2014) J Clin Oncol 32:1020-1030].
  • the effects of prior PD-1 blockade in vivo were next determined on the expansion of tumor-specific TIL from B16 melanomas.
  • mice bearing B16 tumors were treated via intraperitoneal injection with 20 mg/kg non-specific or normal IgG (NrIgG) or blocking anti-PD-1 antibodies. Tumors were collected on day 21 and the isolated TIL were cultured in vitro in NrIgG or anti-4-1BB antibodies for seven days. Addition of anti-41BB antibodies to the in vitro culture without prior anti-PD-1 in vivo led to enhanced anti-tumor activity of the cultured T cells ( FIG. 10B ). However, this increased anti-tumor activity of the short-term cultured B16 TIL with anti-4-1BB was enhanced even more when mice were treated with anti-PD-1 in vivo prior to isolation of the tumors and culture of the TIL ex vivo ( FIG. 10B ). Thus, PD-1 blockade in vivo prior to TIL expansion from resected tumors improves the effects of agonist anti-4-1BB antibody in enhancing anti-tumor reactivity of expanded TIL.
  • FIG. 7A left side contour plots
  • addition of the agonist anti-4-1BB during the tumor fragment culture set-up seemed to activate these DC, as found by a marked increase in CD86 and HLA class II expression in the gated live CD3 ⁇ CD11c + subset in a representative set of fragments ( FIG. 7A ).
  • MHC HLA
  • FIG. 7C CD86
  • CD80 FIG. 7D
  • Adoptive cell therapy (ACT) with autologous TIL has shown considerable efficacy in mediating tumor regression and increasing the survival of metastatic melanoma patients that have failed first and second line therapies [Dudley, M E (2011) J Cancer 2:360-2; Rosenberg, S A, et al (2008) Nat Rev Cancer 8(4):299-308; Radvanyi, L G, et al (2012) Clin Cancer Res 18(24):6758-70].
  • CD8 + T cells have emerged to be a key subset in mediating clinical response, although CD4 + T cells in TIL products can also play sometimes a dominant role in tumor regression in less frequent cases [Friedman, K M, et al (2012) J Immunother 35(5):400-8].
  • a number of biological issues arise in making the TIL initial expansion process not only faster to reduce the wait times for patients, but also to enrich for tumor-reactive T cells, especially CD8 + that also maintain effector-memory phenotypic properties for persistence after infusion.
  • IL-2 has been used for years to expand TIL from these tumor fragments, but other non-cytokine immunomodulators that may play a role have not been investigated.
  • CD8 + TIL tumor-reactive CD8 + TIL in ACT
  • TIL tumor-specific TIL subsets
  • the above experiments were conducted to determine whether modulation of T-cell co-stimulatory pathways on these activated TIL, such as 4-1BB, could have profound effects on the yield of TIL and their phenotypic and functional properties that may be beneficial for ACT approaches.
  • the anti-4-1BB antibody was shown not only increases the rate of TIL expansion from tumor fragments, but also significantly increases the rate yield of CD8 + T cells from the tumors while markedly enriching for tumor-specific CD8 + TIL.
  • these properties were also maintained after further secondary rapid expansion (REP) with anti-CD3, irradiated PBMC feeder cells and IL-2 commonly used to generate the final TIL infusion products in clinical trials.
  • REP secondary rapid expansion
  • the increased tumor reactivity observed in the TIL isolated from tumor fragment cultures treated with anti-4-1BB suggests that this may be an alternative approach at enriching TIL products for tumor specificity in adoptive transfer clinical trials to the direct isolation and purification of 4-1BB + CD8 + T cells from single cells digests of melanoma tumors.
  • the above data showing that blockade of HLA class I during early tumor fragment cultures can inhibit the increased outgrowth of CD8 + T cells with anti-4-1BB also suggests that ongoing antigen presentation may actually occur in these tumor fragments that stimulate resident CD8 + T cells which up-regulate 4-1BB and then get co-stimulated with the agonist antibody.
  • the tumor fragment culture system is an extension of this process which can be further enhanced by addition of immunomodulators such as 4-1BB agonists to improve the output and phenotype of TIL for adoptive cell therapy.
  • the tumor microenvironment was manipulated within early tumor fragment cultures from melanoma patients used to expand TIL for adoptive cell therapy. This can profoundly affect the outgrowth of tumor-reactive TIL associated with the modulation of tumor fragment-resident DC.
  • This approach can be easily applied clinically to more rapidly expand TIL enriched for tumor specificity due to the availability of clinical-grade human or humanized anti-4-1BB antibodies such as the one used here.
  • the disclosed approach can be applied to improve the output of TILs from other solid tumors other than melanoma that have proven more difficult to expand tumor-reactive CD8 + T cells maintaining effector and memory properties.
  • TNBC Triple Negative Breast Cancer
  • TNBC Triple negative breast cancer
  • TIL tumor-infiltrating lymphocytes
  • TNF-R family member signaling such as 41BB/CD137
  • TNBC tumor samples were obtained by surgical resection or core biopsy after neoadjuvant chemotherapy.
  • Small tumor fragments were cultured in 24-well plates in medium containing 3000 IU/ml IL-2, or 3000 IU/ml IL-2 plus 10 ⁇ g/ml added agonistic anti-41BB IgG4 (BM5663513).
  • Viable cell numbers and the expression of CD3, CD8, CD4, CD27, CD28, CCR7, CD45RA, CD56, CD16, Granzyme B, and Perforin was determined by flow cytometry on days 7, 14, 21, 28, and 35 after culture set-up. Cytotoxic function of the TIL was evaluated by measuring Caspase 3 cleavage in target cells.
  • TILs were expanded with IL-2 (3,000 IU/ml) alone for 4 weeks. T cell differentiation was not different in TIL growth from surgical samples or biopsy samples ( FIGS. 14A, 14B ). As shown in FIGS. 15A to 15B , 4-1BB ligation increases the TIL expansion from both surgical and biopsy specimen. TILs were expanded with IL-2 from tumor fragments with or without a single addition of anti-4-1BB on day 0.
  • TILs after 4 weeks of expansion were assayed for activity using flow-cytometry-based assay that measures caspase-3 cleavage using Fc receptor+P815 mastocytoma cells coated with anti-CD3 antibody in a redirected CTL assay ( FIGS. 18A-18D ).
  • TILs expanded from TNBC tumor fragments treated with anti-4-1BB had increased the cleavage of CTL activity. No significant difference in Perforin or Granzyme B expression was found suggesting other mechanisms (e.g., enhanced degranulation ability), or increased expression of other cytolytic molecules, account for the increased CTL activity in TIL expanded with anti-4-1BB ( FIGS. 19A-19B ).
  • TILs were successfully expanded from the tumor fragments in 4/4 tumor samples, IL-2 alone yielded fewer cells than IL-2 together with 41BB costimulation provided early during the culture.
  • Anti-41BB markedly increased the percentage and yield of CD8 + CD3 + T cells at all times (6.34 ⁇ 10 6 vs 202 ⁇ 10 6 ).
  • the resulting CD8 + cells had decreased CD27 expression coincident with increased CD70 when anti-41BB was added.
  • 41BB costimulation also increased the cytotoxic T-cell activity of the expanded TIL.
  • the expanded TIL could also be cryopreserved and later expanded using anti-CD3 and IL-2.
  • Provision of 41BB costimulation immediately upon tumor fragment culture initiation enhanced the rate and yield of CD8 + T cells with increased effector function; these could be cryopreserved and later thawed with high viability for secondary expansion.
  • the disclosed results support further development of an autologous TIL expansion protocol after neoadjuvant therapy for use in an adoptive cell therapy approach to treat TNBC recurrence or metastasis.
US15/303,201 2014-04-10 2015-04-10 Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy Abandoned US20170044496A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/303,201 US20170044496A1 (en) 2014-04-10 2015-04-10 Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201461978112P 2014-04-10 2014-04-10
PCT/US2015/025313 WO2015157636A1 (fr) 2014-04-10 2015-04-10 Expansion améliorée des lymphocytes infiltrants des tumeurs pour thérapie cellulaire adoptive
US15/303,201 US20170044496A1 (en) 2014-04-10 2015-04-10 Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/025313 A-371-Of-International WO2015157636A1 (fr) 2014-04-10 2015-04-10 Expansion améliorée des lymphocytes infiltrants des tumeurs pour thérapie cellulaire adoptive

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/227,824 Continuation US11401506B2 (en) 2014-04-10 2018-12-20 Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy

Publications (1)

Publication Number Publication Date
US20170044496A1 true US20170044496A1 (en) 2017-02-16

Family

ID=54288436

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/303,201 Abandoned US20170044496A1 (en) 2014-04-10 2015-04-10 Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy
US16/227,824 Active US11401506B2 (en) 2014-04-10 2018-12-20 Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy
US17/851,458 Pending US20220340874A1 (en) 2014-04-10 2022-06-28 Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/227,824 Active US11401506B2 (en) 2014-04-10 2018-12-20 Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy
US17/851,458 Pending US20220340874A1 (en) 2014-04-10 2022-06-28 Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy

Country Status (3)

Country Link
US (3) US20170044496A1 (fr)
EP (1) EP3154350B1 (fr)
WO (1) WO2015157636A1 (fr)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170107490A1 (en) * 2014-06-11 2017-04-20 Polybiocept Ab Expansion of lymphocytes with a cytokine composition for active cellular immunotherapy
US10130659B2 (en) 2017-03-29 2018-11-20 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10517894B2 (en) 2016-10-26 2019-12-31 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
WO2020023361A1 (fr) * 2018-07-23 2020-01-30 H. Lee Moffitt Cancer Center And Research Institute Inc. Renforcement de la réponse anti-tumorale dans des cellules de mélanome présentant une signalisation sting défectueuse
WO2020068816A1 (fr) * 2018-09-24 2020-04-02 H. Lee Moffitt Cancer Center And Research Institute, Inc. Culture de lymphocytes infiltrant les tumeurs issus d'une digestion de tumeur
WO2020185658A1 (fr) * 2019-03-08 2020-09-17 North Carolina State University Thérapie photothermique favorisant l'infiltration tumorale et l'activité antitumorale des cellules car t
CN111849892A (zh) * 2020-07-07 2020-10-30 南方医科大学深圳医院 一种胶质瘤来源肿瘤浸润淋巴细胞(til)的体外扩增方法及用途
JP2021503281A (ja) * 2017-11-17 2021-02-12 アイオバンス バイオセラピューティクス,インコーポレイテッド 細針吸引及び小生検からのtil拡大培養
CN112513254A (zh) * 2018-03-29 2021-03-16 艾欧凡斯生物治疗公司 肿瘤浸润淋巴细胞的制备方法及其在免疫治疗中的用途
CN113215099A (zh) * 2021-04-28 2021-08-06 广东康盾创新产业集团股份公司 一种til细胞扩增培养基及其使用方法
US11111493B2 (en) 2018-03-15 2021-09-07 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
US11141434B2 (en) 2016-07-07 2021-10-12 Iovance Biotherapeutics, Inc. Programmed death 1 ligand 1 (PD-L1) binding proteins and methods of use thereof
WO2021239083A1 (fr) * 2020-05-29 2021-12-02 上海君赛生物科技有限公司 Milieu pour cellules d'ensemencement de lymphocytes infiltrant les tumeurs et son application
US11220670B2 (en) 2016-11-17 2022-01-11 Iovance Biotherapeutics, Inc. Remnant tumor infiltrating lymphocytes and methods of preparing and using the same
US11254913B1 (en) 2017-03-29 2022-02-22 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
WO2022105816A1 (fr) * 2020-11-19 2022-05-27 苏州沙砾生物科技有限公司 Procédé de culture de lymphocytes infiltrant les tumeurs et son utilisation
US11357841B2 (en) 2017-01-06 2022-06-14 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof
US11384337B2 (en) 2018-04-27 2022-07-12 Iovance Biotherapeutics, Inc. Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11401506B2 (en) 2014-04-10 2022-08-02 H. Lee Moffitt Cancer Center And Research Institute, Inc. Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy
US11433097B2 (en) 2017-06-05 2022-09-06 Iovance Biotherapeutics, Inc. Methods of using tumor infiltrating lymphocytes in double-refractory melanoma
US20220315892A1 (en) * 2016-10-31 2022-10-06 Iovance Biotherapeutics, Inc. Engineered Artificial Antigen Presenting Cells for Tumor Infiltrating Lymphocyte Expansion
WO2023284721A1 (fr) * 2021-07-13 2023-01-19 苏州沙砾生物科技有限公司 Méthode de mise en culture de cellules immunitaires et son utilisation
US11631483B2 (en) 2020-04-22 2023-04-18 Iovance Biotherapeutics, Inc. Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
WO2023115011A1 (fr) * 2021-12-17 2023-06-22 Instil Bio, Inc. Traitement de lymphocytes infiltrant une tumeur
US11713446B2 (en) 2018-01-08 2023-08-01 Iovance Biotherapeutics, Inc. Processes for generating TIL products enriched for tumor antigen-specific T-cells
WO2023219873A1 (fr) * 2022-05-10 2023-11-16 H. Lee Moffitt Cancer Center And Research Institute, Inc. Procédés de culture de lymphocytes infiltrant les tumeurs
WO2023172623A3 (fr) * 2022-03-08 2024-01-25 Elephas Biosciences Corporation Fragments d'anticorps et leurs utilisations pour l'imagerie d'une activité cellulaire
US11981921B2 (en) 2023-06-20 2024-05-14 Iovance Biotherapeutics, Inc. TIL expansion processes using specific cytokine combinations and/or AKTi treatment

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190028664A (ko) * 2016-05-25 2019-03-19 더 카운실 오브 더 퀸즐랜드 인스티튜트 오브 메디컬 리서치 암의 치료를 위한 면역 체크포인트 억제제와 세포독성 t 세포
EP3532078A4 (fr) * 2016-10-31 2020-06-03 H. Lee Moffitt Cancer Center And Research Institute, Inc. Cellules présentatrices d'antigène artificielles utilisées pour l'expansion de cellules immunitaires pour l'immunothérapie
KR20190104048A (ko) * 2017-01-06 2019-09-05 이오반스 바이오테라퓨틱스, 인크. 종양 괴사 인자 수용체 슈퍼패밀리 (tnfrsf) 효능제를 사용한 종양 침윤 림프구 (til)의 확장 및 til과 tnfrsf 효능제의 치료 조합물
CN107502589A (zh) * 2017-08-04 2017-12-22 北京世纪劲得生物技术有限公司 一种肿瘤浸润淋巴细胞与单个核细胞共培养方法
JP2021508104A (ja) * 2017-12-15 2021-02-25 アイオバンス バイオセラピューティクス,インコーポレイテッド 腫瘍浸潤リンパ球の有益な投与を決定するシステム及び方法並びにその使用方法、並びに腫瘍浸潤リンパ球の有益な投与及びその使用方法
WO2019136459A1 (fr) * 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Procédés de génération de produits de til enrichis pour des lymphocytes t spécifiques d'un antigène tumoral
EP3737743A1 (fr) * 2018-01-08 2020-11-18 Iovance Biotherapeutics, Inc. Procédés de génération de produits de til enrichis pour des lymphocytes t spécifiques d'un antigène tumoral
KR20210136050A (ko) * 2019-03-01 2021-11-16 이오반스 바이오테라퓨틱스, 인크. 액상 종양으로부터의 종양 침윤 림프구의 확장 및 그의 치료 용도
EP4073236A1 (fr) * 2019-12-11 2022-10-19 Iovance Biotherapeutics, Inc. Procédés pour la production de lymphocytes infiltrant les tumeurs (til) et leurs procédés d'utilisation
CA3168932A1 (fr) * 2020-02-28 2021-09-02 Micah BENSON Procedes d'activation et de multiplication de lymphocytes infiltrant les tumeurs
KR20220016670A (ko) * 2020-08-03 2022-02-10 (주) 테라베스트 종양 침윤성 림프구로부터 역분화된 줄기세포 기억 t세포를 포함하는 세포치료제 조성물 및 그 제조 방법
EP4247939A2 (fr) 2020-11-23 2023-09-27 Lyell Immunopharma, Inc. Méthodes de culture de cellules immunitaires
CN112552393B (zh) * 2020-12-31 2022-02-01 西安德诺海思医疗科技有限公司 一种重组人源iii型胶原蛋白及其毕赤酵母重组表达系统
WO2022182915A1 (fr) 2021-02-25 2022-09-01 Lyell Immunopharma, Inc. Procédés de culture cellulaire
AU2022292928A1 (en) * 2021-06-17 2024-01-25 CBio A/S Multistep process for culturing tumor-infiltrating lymphocytes for therapeutic use
AU2022377637A1 (en) 2021-10-28 2024-05-02 Lyell Immunopharma, Inc. Methods for culturing immune cells

Family Cites Families (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU7873187A (en) 1986-08-08 1988-02-24 University Of Minnesota Method of culturing leukocytes
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US7479269B2 (en) 1988-11-23 2009-01-20 Genetics Institute, Llc Methods for selectively enriching TH1 and TH2 cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US5126132A (en) 1989-08-21 1992-06-30 The United States Of America As Represented By The Department Of Health And Human Services Tumor infiltrating lymphocytes as a treatment modality for human cancer
DE4228457A1 (de) 1992-08-27 1994-04-28 Beiersdorf Ag Herstellung von heterodimerem PDGF-AB mit Hilfe eines bicistronischen Vektorsystems in Säugerzellen
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
FR2722208B1 (fr) 1994-07-05 1996-10-04 Inst Nat Sante Rech Med Nouveau site interne d'entree des ribosomes, vecteur le contenant et utilisation therapeutique
AU767880B2 (en) 1998-03-16 2003-11-27 Introgen Therapeutics, Inc. Multigene vectors
WO2000032235A1 (fr) 1998-11-26 2000-06-08 Pentapharm Ag Conjugues systeme de transport
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
US7033597B2 (en) 2000-10-13 2006-04-25 Université de Lausanne Intracellular delivery of biological effectors
JP4387669B2 (ja) 2000-10-13 2009-12-16 ザイジェン エス.アー. 新規なトランスポーターペプチド配列による生物学的エフェクターの細胞内送達
AU2002322211A1 (en) * 2001-07-12 2003-01-29 Canvac Methods and compisitions for activation human t cells in vitro
US20050084967A1 (en) 2002-06-28 2005-04-21 Xcyte Therapies, Inc. Compositions and methods for eliminating undesired subpopulations of T cells in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation
EP1539929B1 (fr) 2002-06-28 2013-04-10 Life Technologies Corporation Techniques permettant de restaurer le repertoire immunologique de patients qui presente des defauts immunologiques lies a l'auto-immunite et a un organe ou a une transplantation de cellules souche hematopoietiques multipotentes
US8034334B2 (en) 2002-09-06 2011-10-11 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Immunotherapy with in vitro-selected antigen-specific lymphocytes after non-myeloablative lymphodepleting chemotherapy
US9255243B2 (en) 2003-10-08 2016-02-09 Wilson Wolf Manufacturing Corporation Cell culture methods and devices utilizing gas permeable materials
CA2970873C (fr) 2005-05-09 2022-05-17 E. R. Squibb & Sons, L.L.C. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d'autres immunotherapies
CN104356236B (zh) 2005-07-01 2020-07-03 E.R.施贵宝&圣斯有限责任公司 抗程序性死亡配体1(pd-l1)的人单克隆抗体
JP5205275B2 (ja) 2005-12-21 2013-06-05 セントクローネ インターナショナル エービー 散在性ガンの治療法
WO2007071390A1 (fr) 2005-12-21 2007-06-28 Sentoclone Ab Méthode de traitement de mélanome malin
US8007785B2 (en) 2005-12-21 2011-08-30 Sentoclone International Ab Method for treating colon cancer with tumour-reactive T-lymphocytes
AU2006328943B2 (en) 2005-12-21 2011-07-21 Sentoclone International Ab Improved method for expansion of tumour-reactive T-lymphocytes for immunotherapy of patients with cancer
US7951365B2 (en) 2007-06-27 2011-05-31 Deifiera Falun Ab Method for expansion of tumour-reactive T-lymphocytes for immunotherapy of patients with specific cancer types
US8475790B2 (en) 2008-10-06 2013-07-02 Bristol-Myers Squibb Company Combination of CD137 antibody and CTLA-4 antibody for the treatment of proliferative diseases
US20150320798A1 (en) 2012-11-27 2015-11-12 The Johns Hopkins University Use of Post-Transplant Cyclophosphamide Treated Allogeneic Marrow Infiltrating Lymphocytes to Augment Anti-Tumor Immunity
US8383099B2 (en) 2009-08-28 2013-02-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Adoptive cell therapy with young T cells
EP2493486A1 (fr) * 2009-10-30 2012-09-05 University Of Arkansas For Medical Science Utilisation de cellules effectrices autologues et d'anticorps pour le traitement d'un myélome multiple
US8956860B2 (en) 2009-12-08 2015-02-17 Juan F. Vera Methods of cell culture for adoptive cell therapy
US20130115617A1 (en) 2009-12-08 2013-05-09 John R. Wilson Methods of cell culture for adoptive cell therapy
SG181559A1 (en) 2009-12-08 2012-07-30 Wolf Wilson Mfg Corp Improved methods of cell culture for adoptive cell therapy
US9512401B2 (en) * 2010-10-01 2016-12-06 Board Of Regents, The University Of Texas System B and T lymphocyte attenuator marker for use in adoptive T-cell therapy
PT2637694T (pt) 2010-11-12 2021-05-05 Nektar Therapeutics Conjugados de uma fração de il-2 e um polímero
WO2012129201A1 (fr) 2011-03-22 2012-09-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Procédés de mise en croissance de lymphocytes infiltrant une tumeur dans contenants perméables au gaz
CN103502439B (zh) 2011-04-13 2016-10-12 因缪尼卡姆股份公司 用于抗原特异性t细胞增殖的方法
KR101499929B1 (ko) 2011-10-21 2015-03-06 셀 메디카 리미티드 세포의 무균 증식 장치
GB201121308D0 (en) 2011-12-12 2012-01-25 Cell Medica Ltd Process
JP6386447B2 (ja) 2012-05-18 2018-09-05 ウィルソン ウォルフ マニュファクチャリング コーポレイションWilson Wolf Manufacturing Corporation 養子細胞療法のための改良された細胞培養法
IL302514A (en) 2012-06-11 2023-07-01 Wilson Wolf Mfg Corporation Improved cell culture methods for stress cell therapy
DK2925329T3 (da) 2012-11-27 2021-03-22 Ivan Marques Borrello Anvendelse af cyclophosphamid-behandlede knoglemarvsinfiltrerende lymfocytter efter transplantationer til øgning af antitumorimmunitet
EP2961415B1 (fr) 2013-03-01 2021-01-06 The United States of America, as represented by The Secretary, Department of Health and Human Services Procédés de production de populations enrichies de lymphocytes t réactifs à une tumeur à partir d'une tumeur
EP3628322A1 (fr) 2013-03-01 2020-04-01 The United States of America, as represented by the Secretary, Department of Health and Human Services Cellules t cd8 + qui expriment également pd-1 et / ou tim-3 pour le traitement du cancer
WO2014210036A1 (fr) 2013-06-24 2014-12-31 Wilson Wolf Manufacturing Corporation Dispositif à système clos et procédés de processus de culture de cellules perméable aux gaz
AR097306A1 (es) 2013-08-20 2016-03-02 Merck Sharp & Dohme Modulación de la inmunidad tumoral
US10233425B2 (en) 2013-09-16 2019-03-19 The Trustees Of The University Of Pennsylvania CD137 enrichment for efficient tumor infiltrating lymphocyte selection
ES2776407T3 (es) 2014-03-20 2020-07-30 H Lee Moffitt Cancer Ct & Res Linfocitos infiltrantes de tumor para terapia celular adoptiva
WO2015157636A1 (fr) 2014-04-10 2015-10-15 H. Lee Moffitt Cancer Center And Research Institute, Inc. Expansion améliorée des lymphocytes infiltrants des tumeurs pour thérapie cellulaire adoptive
EP3838288A1 (fr) 2014-06-11 2021-06-23 polybiocept GmbH Expansion de lymphocytes avec une composition de cytokine pour immunothérapie cellulaire active
CA2959994C (fr) 2014-09-04 2023-04-11 The Johns Hopkins University Activation des lymphocytes infiltrant la moelle osseuse sous des conditions hypotoxiques en alternance avec des conditions normotoxiques
AU2014407539B2 (en) 2014-10-02 2020-10-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of isolating T cell receptors having antigenic specificity for a cancer-specific mutation
EP3034092A1 (fr) 2014-12-17 2016-06-22 Université de Lausanne Immunothérapie adoptive pour traiter le cancer

Cited By (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11401506B2 (en) 2014-04-10 2022-08-02 H. Lee Moffitt Cancer Center And Research Institute, Inc. Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy
US20170107490A1 (en) * 2014-06-11 2017-04-20 Polybiocept Ab Expansion of lymphocytes with a cytokine composition for active cellular immunotherapy
US11141434B2 (en) 2016-07-07 2021-10-12 Iovance Biotherapeutics, Inc. Programmed death 1 ligand 1 (PD-L1) binding proteins and methods of use thereof
US11266694B2 (en) 2016-10-26 2022-03-08 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11344581B2 (en) 2016-10-26 2022-05-31 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11058728B1 (en) 2016-10-26 2021-07-13 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11123371B2 (en) 2016-10-26 2021-09-21 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11975028B2 (en) 2016-10-26 2024-05-07 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US10517894B2 (en) 2016-10-26 2019-12-31 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11865140B2 (en) 2016-10-26 2024-01-09 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11369637B2 (en) 2016-10-26 2022-06-28 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11364266B2 (en) 2016-10-26 2022-06-21 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11141438B2 (en) 2016-10-26 2021-10-12 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11351198B2 (en) 2016-10-26 2022-06-07 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11351199B2 (en) 2016-10-26 2022-06-07 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11351197B2 (en) 2016-10-26 2022-06-07 Iovante Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11857573B2 (en) 2016-10-26 2024-01-02 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11344580B2 (en) 2016-10-26 2022-05-31 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11311578B2 (en) 2016-10-26 2022-04-26 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11304980B2 (en) 2016-10-26 2022-04-19 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11969444B2 (en) 2016-10-26 2024-04-30 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11026974B2 (en) 2016-10-26 2021-06-08 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US11179419B2 (en) 2016-10-26 2021-11-23 Iovance Biotherapeutics, Inc. Restimulation of cryopreserved tumor infiltrating lymphocytes
US20220315892A1 (en) * 2016-10-31 2022-10-06 Iovance Biotherapeutics, Inc. Engineered Artificial Antigen Presenting Cells for Tumor Infiltrating Lymphocyte Expansion
US11667890B2 (en) 2016-10-31 2023-06-06 Iovance Biotherapeutics, Inc. Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
US11220670B2 (en) 2016-11-17 2022-01-11 Iovance Biotherapeutics, Inc. Remnant tumor infiltrating lymphocytes and methods of preparing and using the same
US11293009B2 (en) 2016-11-17 2022-04-05 Iovance Biotherapeutics, Inc. Remnant tumor infiltrating lymphocytes and methods of preparing and using the same
US11401507B2 (en) 2016-11-17 2022-08-02 Iovance Biotherapeutics, Inc. Remnant tumor infiltrating lymphocytes and methods of preparing and using the same
US11357841B2 (en) 2017-01-06 2022-06-14 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof
US10946045B2 (en) 2017-03-29 2021-03-16 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11517592B1 (en) 2017-03-29 2022-12-06 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11013770B1 (en) 2017-03-29 2021-05-25 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11007226B2 (en) 2017-03-29 2021-05-18 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11040070B2 (en) 2017-03-29 2021-06-22 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11052115B2 (en) 2017-03-29 2021-07-06 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11052116B2 (en) 2017-03-29 2021-07-06 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10953046B2 (en) 2017-03-29 2021-03-23 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10130659B2 (en) 2017-03-29 2018-11-20 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11083752B2 (en) 2017-03-29 2021-08-10 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10166257B2 (en) 2017-03-29 2019-01-01 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10953047B2 (en) 2017-03-29 2021-03-23 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10946044B2 (en) 2017-03-29 2021-03-16 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11939596B2 (en) 2017-03-29 2024-03-26 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11168303B2 (en) 2017-03-29 2021-11-09 Iovance Biotherapeutics Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11168304B2 (en) 2017-03-29 2021-11-09 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10933094B2 (en) 2017-03-29 2021-03-02 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10272113B2 (en) 2017-03-29 2019-04-30 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11202804B2 (en) 2017-03-29 2021-12-21 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11202803B1 (en) 2017-03-29 2021-12-21 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10925900B2 (en) 2017-03-29 2021-02-23 lovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11241456B2 (en) 2017-03-29 2022-02-08 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11254913B1 (en) 2017-03-29 2022-02-22 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10918666B2 (en) 2017-03-29 2021-02-16 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11273181B2 (en) 2017-03-29 2022-03-15 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11273180B2 (en) 2017-03-29 2022-03-15 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11291687B2 (en) 2017-03-29 2022-04-05 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10363273B2 (en) 2017-03-29 2019-07-30 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10905718B2 (en) 2017-03-29 2021-02-02 lovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11304979B2 (en) 2017-03-29 2022-04-19 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10894063B2 (en) 2017-03-29 2021-01-19 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11337998B2 (en) 2017-03-29 2022-05-24 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10398734B2 (en) 2017-03-29 2019-09-03 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11541077B2 (en) 2017-03-29 2023-01-03 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11344579B2 (en) 2017-03-29 2022-05-31 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11529372B1 (en) 2017-03-29 2022-12-20 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10695372B2 (en) 2017-03-29 2020-06-30 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10653723B1 (en) 2017-03-29 2020-05-19 Iovance, Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
US10646517B2 (en) 2017-03-29 2020-05-12 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10639330B2 (en) 2017-03-29 2020-05-05 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11007225B1 (en) 2017-03-29 2021-05-18 lovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10420799B2 (en) 2017-03-29 2019-09-24 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10463697B2 (en) 2017-03-29 2019-11-05 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US10537595B2 (en) 2017-03-29 2020-01-21 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11433097B2 (en) 2017-06-05 2022-09-06 Iovance Biotherapeutics, Inc. Methods of using tumor infiltrating lymphocytes in double-refractory melanoma
US11819517B2 (en) 2017-06-05 2023-11-21 Iovance Biotherapeutics, Inc. Methods of using tumor infiltrating lymphocytes in double-refractory melanoma
JP2021503281A (ja) * 2017-11-17 2021-02-12 アイオバンス バイオセラピューティクス,インコーポレイテッド 細針吸引及び小生検からのtil拡大培養
US11713446B2 (en) 2018-01-08 2023-08-01 Iovance Biotherapeutics, Inc. Processes for generating TIL products enriched for tumor antigen-specific T-cells
US11111493B2 (en) 2018-03-15 2021-09-07 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
US11421228B2 (en) 2018-03-15 2022-08-23 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
US11608500B2 (en) 2018-03-15 2023-03-21 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
CN112513254A (zh) * 2018-03-29 2021-03-16 艾欧凡斯生物治疗公司 肿瘤浸润淋巴细胞的制备方法及其在免疫治疗中的用途
US11384337B2 (en) 2018-04-27 2022-07-12 Iovance Biotherapeutics, Inc. Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
US11866688B2 (en) 2018-04-27 2024-01-09 Iovance Biotherapeutics, Inc. Closed process for expansion and gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
WO2020023361A1 (fr) * 2018-07-23 2020-01-30 H. Lee Moffitt Cancer Center And Research Institute Inc. Renforcement de la réponse anti-tumorale dans des cellules de mélanome présentant une signalisation sting défectueuse
WO2020068816A1 (fr) * 2018-09-24 2020-04-02 H. Lee Moffitt Cancer Center And Research Institute, Inc. Culture de lymphocytes infiltrant les tumeurs issus d'une digestion de tumeur
WO2020185658A1 (fr) * 2019-03-08 2020-09-17 North Carolina State University Thérapie photothermique favorisant l'infiltration tumorale et l'activité antitumorale des cellules car t
US11631483B2 (en) 2020-04-22 2023-04-18 Iovance Biotherapeutics, Inc. Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
WO2021239083A1 (fr) * 2020-05-29 2021-12-02 上海君赛生物科技有限公司 Milieu pour cellules d'ensemencement de lymphocytes infiltrant les tumeurs et son application
CN111849892A (zh) * 2020-07-07 2020-10-30 南方医科大学深圳医院 一种胶质瘤来源肿瘤浸润淋巴细胞(til)的体外扩增方法及用途
WO2022105816A1 (fr) * 2020-11-19 2022-05-27 苏州沙砾生物科技有限公司 Procédé de culture de lymphocytes infiltrant les tumeurs et son utilisation
CN114901805A (zh) * 2020-11-19 2022-08-12 苏州沙砾生物科技有限公司 肿瘤浸润淋巴细胞的培养方法及其用途
CN113215099A (zh) * 2021-04-28 2021-08-06 广东康盾创新产业集团股份公司 一种til细胞扩增培养基及其使用方法
WO2023284721A1 (fr) * 2021-07-13 2023-01-19 苏州沙砾生物科技有限公司 Méthode de mise en culture de cellules immunitaires et son utilisation
WO2023115011A1 (fr) * 2021-12-17 2023-06-22 Instil Bio, Inc. Traitement de lymphocytes infiltrant une tumeur
WO2023172623A3 (fr) * 2022-03-08 2024-01-25 Elephas Biosciences Corporation Fragments d'anticorps et leurs utilisations pour l'imagerie d'une activité cellulaire
WO2023219873A1 (fr) * 2022-05-10 2023-11-16 H. Lee Moffitt Cancer Center And Research Institute, Inc. Procédés de culture de lymphocytes infiltrant les tumeurs
US11981921B2 (en) 2023-06-20 2024-05-14 Iovance Biotherapeutics, Inc. TIL expansion processes using specific cytokine combinations and/or AKTi treatment

Also Published As

Publication number Publication date
EP3154350A4 (fr) 2018-04-18
US20200032209A1 (en) 2020-01-30
US20220340874A1 (en) 2022-10-27
US11401506B2 (en) 2022-08-02
WO2015157636A1 (fr) 2015-10-15
EP3154350B1 (fr) 2024-03-27
EP3154350A1 (fr) 2017-04-19

Similar Documents

Publication Publication Date Title
US20220340874A1 (en) Enhanced Expansion of Tumor-Infiltrating Lymphocytes for Adoptive Cell Therapy
US20230092130A1 (en) Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
AU2018211278B2 (en) Agents for treatment of claudin expressing cancer diseases
JP2018507208A (ja) 癌治療のための免疫療法とサイトカイン制御療法との組み合わせ
WO2022165260A9 (fr) Procédés de fabrication de lymphocytes infiltrant les tumeurs modifiés et leur utilisation dans la thérapie cellulaire adoptive
EP3843759B1 (fr) Utilisation de lymphocytes infiltrant les tumeurs pour traiter les patients souffrant de nsclc réfractaires à un anticorps anti-pd-1
KR20210091212A (ko) 항-pd-1 항체에 불응성인 nsclc 환자의 치료
EP4262811A1 (fr) Traitement avec des thérapies de lymphocytes infiltrant les tumeurs en combinaison avec des inhibiteurs de ctla-4 et de pd-1
JP2023516636A (ja) 腫瘍反応性t細胞のエクスビボ富化および増大のための方法ならびに関連するその組成物
EP2920209B1 (fr) Agents de traitement de maladies cancéreuses exprimant claudine
CA3231508A1 (fr) Methodes d'amplification de populations immunitaires reagissant aux tumeurs a l'aide d'organoides
WO2023009716A1 (fr) Traitement de patients atteints d'un cancer avec des thérapies de lymphocytes infiltrant les tumeurs en combinaison avec des inhibiteurs de kras
WO2023086803A1 (fr) Procédés de traitement de multiplication utilisant des lymphocytes infiltrant les tumeurs cd8
WO2023220608A1 (fr) Traitement de patients atteints d'un cancer avec des thérapies lymphocytaires infiltrant les tumeurs en combinaison avec un agoniste d'il-15r
CN117940557A (zh) 制备经修饰的肿瘤浸润性淋巴细胞的方法及其在过继性细胞治疗中的应用

Legal Events

Date Code Title Description
AS Assignment

Owner name: H. LEE MOFFITT CANCER CENTER AND RESEARCH INSTITUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SARNAIK, AMOD A.;WEBER, JEFFREY S.;PILON-THOMAS, SHARI;AND OTHERS;SIGNING DATES FROM 20161202 TO 20170501;REEL/FRAME:043062/0693

Owner name: BOARD OF REGENTS, THE UNIVERSITY OF TEXAS SYSTEM,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:RADVANYI, LASZLO;CHACON, JESSICA;SIGNING DATES FROM 20170501 TO 20170523;REEL/FRAME:043062/0795

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION