74 results on '"Rosenberg, Steven"'
Search Results
2. Neoantigen-specific stimulation of tumor-infiltrating lymphocytes enables effective TCR isolation and expansion while preserving stem-like memory phenotypes.
- Author
-
Levin N, Kim SP, Marquardt CA, Vale NR, Yu Z, Sindiri S, Gartner JJ, Parkhurst M, Krishna S, Lowery FJ, Zacharakis N, Levy L, Prickett TD, Benzine T, Ray S, Masi RV, Gasmi B, Li Y, Islam R, Bera A, Goff SL, Robbins PF, and Rosenberg SA
- Subjects
- Humans, Mice, Immunologic Memory, Animals, Female, Phenotype, Neoplasms immunology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Antigens, Neoplasm immunology, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism
- Abstract
Background: Tumor-infiltrating lymphocytes (TILs) targeting neoantigens can effectively treat a selected set of metastatic solid cancers. However, harnessing TILs for cancer treatments remains challenging because neoantigen-reactive T cells are often rare and exhausted, and ex vivo expansion can further reduce their frequencies. This complicates the identification of neoantigen-reactive T-cell receptors (TCRs) and the development of TIL products with high reactivity for patient treatment., Methods: We tested whether TILs could be in vitro stimulated against neoantigens to achieve selective expansion of neoantigen-reactive TILs. Given their prevalence, mutant p53 or RAS were studied as models of human neoantigens. An in vitro stimulation method, termed "NeoExpand", was developed to provide neoantigen-specific stimulation to TILs. 25 consecutive patient TILs from tumors harboring p53 or RAS mutations were subjected to NeoExpand., Results: We show that neoantigenic stimulation achieved selective expansion of neoantigen-reactive TILs and broadened the neoantigen-reactive CD4
+ and CD8+ TIL clonal repertoire. This allowed the effective isolation of novel neoantigen-reactive TCRs. Out of the 25 consecutive TIL samples, neoantigenic stimulation enabled the identification of 16 unique reactivities and 42 TCRs, while conventional TIL expansion identified 9 reactivities and 14 TCRs. Single-cell transcriptome analysis revealed that neoantigenic stimulation increased neoantigen-reactive TILs with stem-like memory phenotypes expressing IL-7R, CD62L, and KLF2. Furthermore, neoantigenic stimulation improved the in vivo antitumor efficacy of TILs relative to the conventional OKT3-induced rapid TIL expansion in p53-mutated or KRAS-mutated xenograft mouse models., Conclusions: Taken together, neoantigenic stimulation of TILs selectively expands neoantigen-reactive TILs by frequencies and by their clonal repertoire. NeoExpand led to improved phenotypes and functions of neoantigen-reactive TILs. Our data warrant its clinical evaluation., Trial Registration Number: NCT00068003, NCT01174121, and NCT03412877., Competing Interests: Competing interests: NL, SPK and SAR have a pending patent application. The rest of the authors report no competing interest., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2024
- Full Text
- View/download PDF
3. Internal checkpoint regulates T cell neoantigen reactivity and susceptibility to PD1 blockade.
- Author
-
Palmer DC, Webber BR, Patel Y, Johnson MJ, Kariya CM, Lahr WS, Parkhurst MR, Gartner JJ, Prickett TD, Lowery FJ, Kishton RJ, Gurusamy D, Franco Z, Vodnala SK, Diers MD, Wolf NK, Slipek NJ, McKenna DH, Sumstad D, Viney L, Henley T, Bürckstümmer T, Baker O, Hu Y, Yan C, Meerzaman D, Padhan K, Lo W, Malekzadeh P, Jia L, Deniger DC, Patel SJ, Robbins PF, McIvor RS, Choudhry M, Rosenberg SA, Moriarity BS, and Restifo NP
- Subjects
- Adoptive Transfer, Animals, Cytokines metabolism, Humans, Immunotherapy, Adoptive methods, Mice, Lymphocytes, Tumor-Infiltrating, T-Lymphocytes
- Abstract
Background: Adoptive transfer of tumor-infiltrating lymphocytes (TIL) fails to consistently elicit tumor rejection. Manipulation of intrinsic factors that inhibit T cell effector function and neoantigen recognition may therefore improve TIL therapy outcomes. We previously identified the cytokine-induced SH2 protein (CISH) as a key regulator of T cell functional avidity in mice. Here, we investigate the mechanistic role of CISH in regulating human T cell effector function in solid tumors and demonstrate that CRISPR/Cas9 disruption of CISH enhances TIL neoantigen recognition and response to checkpoint blockade., Methods: Single-cell gene expression profiling was used to identify a negative correlation between high CISH expression and TIL activation in patient-derived TIL. A GMP-compliant CRISPR/Cas9 gene editing process was developed to assess the impact of CISH disruption on the molecular and functional phenotype of human peripheral blood T cells and TIL. Tumor-specific T cells with disrupted Cish function were adoptively transferred into tumor-bearing mice and evaluated for efficacy with or without checkpoint blockade., Findings: CISH expression was associated with T cell dysfunction. CISH deletion using CRISPR/Cas9 resulted in hyper-activation and improved functional avidity against tumor-derived neoantigens without perturbing T cell maturation. Cish knockout resulted in increased susceptibility to checkpoint blockade in vivo., Conclusions: CISH negatively regulates human T cell effector function, and its genetic disruption offers a novel avenue to improve the therapeutic efficacy of adoptive TIL therapy., Funding: This study was funded by Intima Bioscience, U.S. and in part through the Intramural program CCR at the National Cancer Institute., Competing Interests: Declaration of interests M.C. is a co-founder of Intima Bioscience. B.R.W., S.A.R., and B.S.M. have received sponsored research support from Intima Bioscience. D.C.P., B.R.W., M.C., S.A.R., B.S.M., and N.P.R. have patents filed based on the findings described here., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
4. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers.
- Author
-
Lowery FJ, Krishna S, Yossef R, Parikh NB, Chatani PD, Zacharakis N, Parkhurst MR, Levin N, Sindiri S, Sachs A, Hitscherich KJ, Yu Z, Vale NR, Lu YC, Zheng Z, Jia L, Gartner JJ, Hill VK, Copeland AR, Nah SK, Masi RV, Gasmi B, Kivitz S, Paria BC, Florentin M, Kim SP, Hanada KI, Li YF, Ngo LT, Ray S, Shindorf ML, Levi ST, Shepherd R, Toy C, Parikh AY, Prickett TD, Kelly MC, Beyer R, Goff SL, Yang JC, Robbins PF, and Rosenberg SA
- Subjects
- CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Gene Regulatory Networks, Humans, Lymphocytes, Tumor-Infiltrating metabolism, Neoplasms genetics, Neoplasms metabolism, RNA-Seq, Single-Cell Analysis, Antigens, Neoplasm immunology, Lymphocytes, Tumor-Infiltrating immunology, Neoplasm Metastasis, Neoplasms immunology, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology, Transcriptome
- Abstract
The accurate identification of antitumor T cell receptors (TCRs) represents a major challenge for the engineering of cell-based cancer immunotherapies. By mapping 55 neoantigen-specific TCR clonotypes (NeoTCRs) from 10 metastatic human tumors to their single-cell transcriptomes, we identified signatures of CD8
+ and CD4+ neoantigen-reactive tumor-infiltrating lymphocytes (TILs). Neoantigen-specific TILs exhibited tumor-specific expansion with dysfunctional phenotypes, distinct from blood-emigrant bystanders and regulatory TILs. Prospective prediction and testing of 73 NeoTCR signature-derived clonotypes demonstrated that half of the tested TCRs recognized tumor antigens or autologous tumors. NeoTCR signatures identified TCRs that target driver neoantigens and nonmutated viral or tumor-associated antigens, suggesting a common metastatic TIL exhaustion program. NeoTCR signatures delineate the landscape of TILs across metastatic tumors, enabling successful TCR prediction based purely on TIL transcriptomic states for use in cancer immunotherapy.- Published
- 2022
- Full Text
- View/download PDF
5. Identification of neoantigen-reactive T lymphocytes in the peripheral blood of a patient with glioblastoma.
- Author
-
Leko V, Cafri G, Yossef R, Paria B, Hill V, Gurusamy D, Zheng Z, Gartner JJ, Prickett TD, Goff SL, Robbins P, Lu YC, and Rosenberg SA
- Subjects
- Humans, Glioblastoma immunology, Lymphocytes, Tumor-Infiltrating immunology
- Abstract
The adoptive transfer of naturally occurring T cells that recognize cancer neoantigens has led to durable tumor regressions in select patients with cancer. However, it remains unknown whether such T cells can be isolated from and used to treat patients with glioblastoma, a cancer that is refractory to currently available therapies. To answer this question, we stimulated patient blood-derived memory T cells in vitro using peptides and minigenes that represented point mutations unique to patients' tumors (ie, candidate neoantigens) and then tested their ability to specifically recognize these mutations. In a cohort of five patients with glioblastoma, we found that circulating CD4
+ memory T cells from one patient recognized a cancer neoantigen harboring a mutation in the EED gene (EEDH189N ) that was unique to that patient's tumor. This finding suggests that neoantigen-reactive T cells could indeed be isolated from patients with glioblastoma, thereby providing a rationale for further efforts to develop neoantigen-directed adoptive T cell therapy for this disease., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
6. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer.
- Author
-
Krishna S, Lowery FJ, Copeland AR, Bahadiroglu E, Mukherjee R, Jia L, Anibal JT, Sachs A, Adebola SO, Gurusamy D, Yu Z, Hill V, Gartner JJ, Li YF, Parkhurst M, Paria B, Kvistborg P, Kelly MC, Goff SL, Altan-Bonnet G, Robbins PF, and Rosenberg SA
- Subjects
- Animals, Antigens, CD analysis, Antigens, Differentiation, T-Lymphocyte analysis, Apyrase analysis, CD8-Positive T-Lymphocytes chemistry, Female, Humans, Lectins, C-Type analysis, Melanoma immunology, Mice, Mice, Mutant Strains, Skin Neoplasms immunology, CD8-Positive T-Lymphocytes immunology, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating transplantation, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Adoptive T cell therapy (ACT) using ex vivo-expanded autologous tumor-infiltrating lymphocytes (TILs) can mediate complete regression of certain human cancers. The impact of TIL phenotypes on clinical success of TIL-ACT is currently unclear. Using high-dimensional analysis of human ACT products, we identified a memory-progenitor CD39-negative stem-like phenotype (CD39
- CD69- ) associated with complete cancer regression and TIL persistence and a terminally differentiated CD39-positive state (CD39+ CD69+ ) associated with poor TIL persistence. Most antitumor neoantigen-reactive TILs were found in the differentiated CD39+ state. However, ACT responders retained a pool of CD39- stem-like neoantigen-specific TILs that was lacking in ACT nonresponders. Tumor-reactive stem-like TILs were capable of self-renewal, expansion, persistence, and superior antitumor response in vivo. These data suggest that TIL subsets mediating ACT response are distinct from TIL subsets enriched for antitumor reactivity., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
7. Enhanced efficacy and limited systemic cytokine exposure with membrane-anchored interleukin-12 T-cell therapy in murine tumor models.
- Author
-
Zhang L, Davies JS, Serna C, Yu Z, Restifo NP, Rosenberg SA, Morgan RA, and Hinrichs CS
- Subjects
- Animals, Cell Line, Tumor, Cell Membrane genetics, Cell Membrane metabolism, Genetic Vectors administration & dosage, Genetic Vectors genetics, Genetic Vectors immunology, Humans, Interferon-gamma metabolism, Interleukin-12 administration & dosage, Interleukin-12 genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Xenograft Model Antitumor Assays, Antigens, Neoplasm immunology, Immunotherapy, Adoptive methods, Interleukin-12 immunology, Lymphocytes, Tumor-Infiltrating immunology, Melanoma, Experimental immunology, Melanoma, Experimental therapy, Receptors, Antigen, T-Cell immunology
- Abstract
Background: Interleukin-12 (IL-12) is a potent, proinflammatory cytokine that holds promise for cancer immunotherapy, but its clinical use has been limited by its toxicity. To minimize systemic exposure and potential toxicity while maintaining the beneficial effects of IL-12, we developed a novel IL-12-based therapeutic system that combines tumor-specific T-cell-mediated delivery of IL-12 with membrane-restricted IL-12 localization and inducible IL-12 expression., Methods: Therapeutic T cells targeting a tumor antigen were genetically engineered to express membrane-anchored IL-12 (aIL-12). Expression, function, and shedding of the aIL-12 molecule was assessed in vitro. Tumor treatment efficacy was assessed in vivo with T cell receptor (TCR) transgenic murine tumor models and a tumor xenograft model. Key outcomes were change in tumor size, circulating levels of IL-12 and other cytokines, and survival. Toxicity was assessed via change in body weight. Tumor growth curve measurements were compared using repeated-measures two-way analyses of variance., Results: Retroviral gene transfer resulted in cell membrane expression of aIL-12 by transduced T cells. In each of two transgenic murine tumor models, tumor-specific T cells constitutively expressing aIL-12 demonstrated increased antitumor efficacy, low circulating IL-12 and interferon-γ, and no weight loss. Expression of aIL-12 via a NFAT- inducible promoter resulted in coordinate expression of aIL-12 with T cell activation. In an OT-I TCR transgenic murine tumor model, the NFAT -inducible aIL-12 molecule improved tumor treatment and did not result in detectable levels of IL-12 in serum or in weight loss. In a human tumor xenograft model, the NFAT -inducible aIL-12 molecule improved antitumor responses by human T cells coexpressing a tumor-specific engineered TCR. Serum IL-12 levels were undetectable with the NFAT -inducible construct in both models., Conclusion: Expression of aIL-12 by tumor-targeting therapeutic T cells demonstrated low systemic exposure and improved efficacy. This treatment strategy may have broad applications to cellular therapy with tumor-infiltrating lymphocytes, chimeric antigen receptor T cells, and TCR T cells., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
8. Single-Cell Transcriptome Analysis Reveals Gene Signatures Associated with T-cell Persistence Following Adoptive Cell Therapy.
- Author
-
Lu YC, Jia L, Zheng Z, Tran E, Robbins PF, and Rosenberg SA
- Subjects
- Biomarkers metabolism, Clone Cells immunology, Clone Cells metabolism, Colorectal Neoplasms immunology, Colorectal Neoplasms pathology, Colorectal Neoplasms therapy, Gene Expression Profiling, Gene Expression Regulation, Humans, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating transplantation, Membrane Proteins genetics, Membrane Proteins metabolism, Single-Cell Analysis, Transcription Factors genetics, Transcription Factors metabolism, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating immunology
- Abstract
Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TIL) can mediate responses in some patients with metastatic epithelial cancer. Identifying gene signatures associated with successful ACT might enable the development of improved therapeutic approaches. The persistence of transferred T cells in the peripheral blood is one indication of clinical effectiveness, but many T-cell and host factors may influence T-cell persistence. To limit these variables, we previously studied a patient with metastatic colorectal cancer treated with polyclonal TILs targeting the KRAS (G12D) hotspot mutation, who experienced a partial response for 9 months. Three dominant clonotypes specifically recognizing KRAS(G12D) epitopes were identified, but we found that only two clonotypes persisted 40 days after ACT. Because of these findings, in this study, we performed the single-cell transcriptome analysis of the infused TILs. The analysis revealed a total of 472 genes that were differentially expressed between clonotypes 9.1-NP and 9.2-P single cells, and 528 genes between 9.1-NP and 10-P. Following these clonotypes in the peripheral blood after ACT, the gene expression patterns changed, but IL7R, ITGB1, KLF2 , and ZNF683 remained expressed in the persistent 9.2-P and 10-P cells, compared with the nonpersistent 9.1-NP cells. In addition, four autologous TILs, which were used for treatment but persisted poorly 1 month after ACT, did not express the gene profiles associated with persistence. These results suggest that certain TIL populations possess a unique gene expression profile that can lead to the persistence of T cells. Thus, this single-patient study provides insight into how to improve ACT for solid cancer., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
9. Identification of Neoantigen-Reactive Tumor-Infiltrating Lymphocytes in Primary Bladder Cancer.
- Author
-
Leko V, McDuffie LA, Zheng Z, Gartner JJ, Prickett TD, Apolo AB, Agarwal PK, Rosenberg SA, and Lu YC
- Subjects
- Adult, Aged, Alcohol Oxidoreductases genetics, Alcohol Oxidoreductases immunology, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, Cells, Cultured, Cytokines metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins immunology, Humans, Lymphocyte Activation, Male, Middle Aged, Mutation genetics, Alcohol Oxidoreductases metabolism, Antigens, Neoplasm metabolism, Cancer Vaccines immunology, DNA-Binding Proteins metabolism, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Urinary Bladder Neoplasms immunology
- Abstract
Immune checkpoint inhibitors are effective in treating a variety of malignancies, including metastatic bladder cancer. A generally accepted hypothesis suggests that immune checkpoint inhibitors induce tumor regressions by reactivating a population of endogenous tumor-infiltrating lymphocytes (TILs) that recognize cancer neoantigens. Although previous studies have identified neoantigen-reactive TILs from several types of cancer, no study to date has shown whether neoantigen-reactive TILs can be found in bladder tumors. To address this, we generated TIL cultures from patients with primary bladder cancer and tested their ability to recognize tumor-specific mutations. We found that CD4
+ TILs from one patient recognized mutated C-terminal binding protein 1 in an MHC class II-restricted manner. This finding suggests that neoantigen-reactive TILs reside in bladder cancer, which may help explain the effectiveness of immune checkpoint blockade in this disease and also provides a rationale for the future use of adoptive T cell therapy targeting neoantigens in bladder cancer.- Published
- 2019
- Full Text
- View/download PDF
10. A Phase II Study of Tumor-infiltrating Lymphocyte Therapy for Human Papillomavirus-associated Epithelial Cancers.
- Author
-
Stevanović S, Helman SR, Wunderlich JR, Langhan MM, Doran SL, Kwong MLM, Somerville RPT, Klebanoff CA, Kammula US, Sherry RM, Yang JC, Rosenberg SA, and Hinrichs CS
- Subjects
- Adult, Carcinoma diagnosis, Carcinoma metabolism, Female, Humans, Male, Middle Aged, Papillomavirus Infections virology, Tomography, X-Ray Computed, Treatment Outcome, Carcinoma etiology, Carcinoma therapy, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Papillomaviridae, Papillomavirus Infections complications
- Abstract
Purpose: Cellular therapy is an emerging cancer treatment modality, but its application to epithelial cancers has been limited. This clinical trial evaluated tumor-infiltrating lymphocyte (TIL) therapy for the treatment of patients with metastatic human papillomavirus (HPV)-associated carcinomas., Patients and Methods: The trial was a phase II design with two cohorts, cervical cancers and noncervical cancers. Cell infusion was preceded by a lymphocyte-depleting conditioning regimen and followed by systemic high-dose aldesleukin., Results: Objective tumor responses occurred in 5 of 18 (28%) patients in the cervical cancer cohort and 2 of 11 (18%) patients in the noncervical cancer cohort. Two of the responses in cervical cancer were complete and are ongoing 67 and 53 months after treatment. Responses in the noncervical cancer cohort were in anal cancer and oropharyngeal cancer. The HPV reactivity of the infused T cells correlated with clinical response. Peripheral blood repopulation with HPV-reactive T cells also correlated with clinical response., Conclusions: These findings support the concept that cellular therapy can mediate the regression of epithelial cancers, and they suggest the importance of predictive biomarkers and novel treatment platforms for more effective therapies., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
11. Combined Analysis of Antigen Presentation and T-cell Recognition Reveals Restricted Immune Responses in Melanoma.
- Author
-
Kalaora S, Wolf Y, Feferman T, Barnea E, Greenstein E, Reshef D, Tirosh I, Reuben A, Patkar S, Levy R, Quinkhardt J, Omokoko T, Qutob N, Golani O, Zhang J, Mao X, Song X, Bernatchez C, Haymaker C, Forget MA, Creasy C, Greenberg P, Carter BW, Cooper ZA, Rosenberg SA, Lotem M, Sahin U, Shakhar G, Ruppin E, Wargo JA, Friedman N, Admon A, and Samuels Y
- Subjects
- Animals, Antigens, Neoplasm metabolism, Histocompatibility Antigens Class I metabolism, Humans, Melanoma metabolism, Melanoma pathology, Mice, Mice, Inbred NOD, Mice, SCID, T-Lymphocytes metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antigen Presentation immunology, Antigens, Neoplasm immunology, Histocompatibility Antigens Class I immunology, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, T-Lymphocytes immunology
- Abstract
The quest for tumor-associated antigens (TAA) and neoantigens is a major focus of cancer immunotherapy. Here, we combine a neoantigen prediction pipeline and human leukocyte antigen (HLA) peptidomics to identify TAAs and neoantigens in 16 tumors derived from seven patients with melanoma and characterize their interactions with their tumor-infiltrating lymphocytes (TIL). Our investigation of the antigenic and T-cell landscapes encompassing the TAA and neoantigen signatures, their immune reactivity, and their corresponding T-cell identities provides the first comprehensive analysis of cancer cell T-cell cosignatures, allowing us to discover remarkable antigenic and TIL similarities between metastases from the same patient. Furthermore, we reveal that two neoantigen-specific clonotypes killed 90% of autologous melanoma cells, both in vitro and in vivo , showing that a limited set of neoantigen-specific T cells may play a central role in melanoma tumor rejection. Our findings indicate that combining HLA peptidomics with neoantigen predictions allows robust identification of targetable neoantigens, which could successfully guide personalized cancer immunotherapies. Significance: As neoantigen targeting is becoming more established as a powerful therapeutic approach, investigating these molecules has taken center stage. Here, we show that a limited set of neoantigen-specific T cells mediates tumor rejection, suggesting that identifying just a few antigens and their corresponding T-cell clones could guide personalized immunotherapy. Cancer Discov; 8(11); 1366-75. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 1333 ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
12. Enhanced detection of neoantigen-reactive T cells targeting unique and shared oncogenes for personalized cancer immunotherapy.
- Author
-
Yossef R, Tran E, Deniger DC, Gros A, Pasetto A, Parkhurst MR, Gartner JJ, Prickett TD, Cafri G, Robbins PF, and Rosenberg SA
- Subjects
- Adult, Aged, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Carcinogenesis genetics, Female, Humans, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Male, Middle Aged, Mutation, Neoplasms genetics, Neoplasms immunology, Oncogenes genetics, Oncogenes immunology, Programmed Cell Death 1 Receptor immunology, Programmed Cell Death 1 Receptor metabolism, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism, Sensitivity and Specificity, Tumor Cells, Cultured, Cell Separation methods, Flow Cytometry methods, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating transplantation, Neoplasms therapy
- Abstract
Adoptive cell transfer (ACT) of tumor-infiltrating lymphocytes (TILs) targeting neoantigens can mediate tumor regression in selected patients with metastatic epithelial cancer. However, effectively identifying and harnessing neoantigen-reactive T cells for patient treatment remains a challenge and it is unknown whether current methods to detect neoantigen-reactive T cells are missing potentially clinically relevant neoantigen reactivities. We thus investigated whether the detection of neoantigen-reactive TILs could be enhanced by enriching T cells that express PD-1 and/or T cell activation markers followed by microwell culturing to avoid overgrowth of nonreactive T cells. In 6 patients with metastatic epithelial cancer, this method led to the detection of CD4+ and CD8+ T cells targeting 18 and 1 neoantigens, respectively, compared with 6 and 2 neoantigens recognized by CD4+ and CD8+ T cells, respectively, when using our standard TIL fragment screening approach. In 2 patients, no recognition of mutated peptides was observed using our conventional screen, while our high-throughput approach led to the identification of 5 neoantigen-reactive T cell receptors (TCRs) against 5 different mutations from one patient and a highly potent MHC class II-restricted KRASG12V-reactive TCR from a second patient. In addition, in a metastatic tumor sample from a patient with serous ovarian cancer, we isolated 3 MHC class II-restricted TCRs targeting the TP53G245S hot-spot mutation. In conclusion, this approach provides a highly sensitive platform to isolate clinically relevant neoantigen-reactive T cells or their TCRs for cancer treatment.
- Published
- 2018
- Full Text
- View/download PDF
13. Outcomes of Adoptive Cell Transfer With Tumor-infiltrating Lymphocytes for Metastatic Melanoma Patients With and Without Brain Metastases.
- Author
-
Mehta GU, Malekzadeh P, Shelton T, White DE, Butman JA, Yang JC, Kammula US, Goff SL, Rosenberg SA, and Sherry RM
- Subjects
- Adolescent, Adult, Aged, Brain Neoplasms immunology, Brain Neoplasms secondary, Female, Follow-Up Studies, Humans, Lymphocytes, Tumor-Infiltrating transplantation, Male, Melanoma immunology, Melanoma secondary, Middle Aged, Neoplasm Metastasis, Neoplasm Staging, Retrospective Studies, Skin Neoplasms immunology, Skin Neoplasms pathology, Skin Neoplasms secondary, Treatment Outcome, Young Adult, Melanoma, Cutaneous Malignant, Brain Neoplasms therapy, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Brain metastases cause significant morbidity and mortality in patients with metastatic melanoma. Although adoptive cell therapy (ACT) with tumor-infiltrating lymphocytes (TIL) can achieve complete and durable remission of advanced cutaneous melanoma, the efficacy of this therapy for brain metastases is unclear. Records of patients with M1c melanoma treated with ACT using TIL, including patients with treated and untreated brain metastases, were analyzed. Treatment consisted of preparative chemotherapy, autologous TIL infusion, and high-dose interleukin-2. Treatment outcomes, sites of initial tumor progression, and overall survival were analyzed. Among 144 total patients, 15 patients with treated and 18 patients with untreated brain metastases were identified. Intracranial objective responses (OR) occurred in 28% patients with untreated brain metastases. The systemic OR rates for patients with M1c disease without identified brain disease, treated brain disease, and untreated brain disease, and were 49%, 33% and 33%, respectively, of which 59%, 20% and 16% were durable at last follow-up. The site of untreated brain disease was the most likely site of initial tumor progression (61%) in patients with untreated brain metastases. Overall, we found that ACT with TIL can eliminate small melanoma brain metastases. However, following TIL therapy these patients frequently progress in the brain at a site of untreated brain disease. Patients with treated or untreated brain disease are less likely to achieve durable systemic ORs following TIL therapy compared with M1c disease and no history of brain disease. Melanoma brain metastases likely require local therapy despite the systemic effect of ACT.
- Published
- 2018
- Full Text
- View/download PDF
14. Characterization of an Immunogenic Mutation in a Patient with Metastatic Triple-Negative Breast Cancer.
- Author
-
Assadipour Y, Zacharakis N, Crystal JS, Prickett TD, Gartner JJ, Somerville RPT, Xu H, Black MA, Jia L, Chinnasamy H, Kriley I, Lu L, Wunderlich JR, Zheng Z, Lu YC, Robbins PF, Rosenberg SA, Goff SL, and Feldman SA
- Subjects
- Antigen-Presenting Cells immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes transplantation, Female, HLA-DRB1 Chains immunology, Humans, Lymphocytes, Tumor-Infiltrating transplantation, Middle Aged, Mutation, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Tumor Necrosis Factor Receptor Superfamily, Member 9 immunology, Exome Sequencing, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Triple Negative Breast Neoplasms immunology, Triple Negative Breast Neoplasms therapy
- Abstract
Purpose: The administration of autologous tumor-infiltrating lymphocytes (TILs) can mediate durable tumor regressions in patients with melanoma likely based on the recognition of immunogenic somatic mutations expressed by the cancer. There are limited data regarding the immunogenicity of mutations in breast cancer. We sought to identify immunogenic nonsynonymous mutations in a patient with triple-negative breast cancer (TNBC) to identify and isolate mutation-reactive TILs for possible use in adoptive cell transfer. Experimental Design: A TNBC metastasis was resected for TIL generation and whole-exome sequencing. Tandem minigenes or long 25-mer peptides encoding selected mutations were electroporated or pulsed onto autologous antigen-presenting cells, and reactivity of TIL was screened by upregulation of CD137 and IFNγ ELISPOT. The nature of the T-cell response against a unique nonsynonymous mutation was characterized. Results: We identified 72 nonsynonymous mutations from the tumor of a patient with TNBC. CD4
+ and HLA-DRB1*1501-restricted TILs isolated from this tumor recognized a single mutation in RBPJ (recombination signal binding protein for immunoglobulin kappa J region). Analysis of 16 metastatic sites revealed that the mutation was ubiquitously present in all samples. Conclusions: Breast cancers can express naturally processed and presented unique nonsynonymous mutations that are recognized by a patient's immune system. TILs recognizing these immunogenic mutations can be isolated from a patient's tumor, suggesting that adoptive cell transfer of mutation-reactive TILs could be a viable treatment option for patients with breast cancer. Clin Cancer Res; 23(15); 4347-53. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
15. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study.
- Author
-
Chandran SS, Somerville RPT, Yang JC, Sherry RM, Klebanoff CA, Goff SL, Wunderlich JR, Danforth DN, Zlott D, Paria BC, Sabesan AC, Srivastava AK, Xi L, Pham TH, Raffeld M, White DE, Toomey MA, Rosenberg SA, and Kammula US
- Subjects
- Adult, Anemia chemically induced, Eye Enucleation, Female, GTP-Binding Protein alpha Subunits genetics, GTP-Binding Protein alpha Subunits, Gq-G11 genetics, Humans, Infections chemically induced, Lymphopenia chemically induced, Male, Melanoma genetics, Melanoma secondary, Metastasectomy, Middle Aged, Neutropenia chemically induced, Radiotherapy, Response Evaluation Criteria in Solid Tumors, Thrombocytopenia chemically induced, Transplantation Conditioning adverse effects, Transplantation, Autologous, Uveal Neoplasms genetics, Uveal Neoplasms pathology, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating transplantation, Melanoma therapy, Uveal Neoplasms therapy
- Abstract
Background: Uveal melanoma is a rare tumour with no established treatments once metastases develop. Although a variety of immune-based therapies have shown efficacy in metastatic cutaneous melanoma, their use in ocular variants has been disappointing. Recently, adoptive T-cell therapy has shown salvage responses in multiple refractory solid tumours. Thus, we sought to determine if adoptive transfer of autologous tumour-infiltrating lymphocytes (TILs) could mediate regression of metastatic uveal melanoma., Methods: In this ongoing single-centre, two-stage, phase 2, single-arm trial, patients (aged ≥16 years) with histologically confirmed metastatic ocular melanoma were enrolled. Key eligibility criteria were an Eastern Cooperative Oncology Group performance status of 0 or 1, progressive metastatic disease, and adequate haematological, renal, and hepatic function. Metastasectomies were done to procure tumour tissue to generate autologous TIL cultures, which then underwent large scale ex-vivo expansion. Patients were treated with lymphodepleting conditioning chemotherapy (intravenous cyclophosphamide [60 mg/kg] daily for 2 days followed by fludarabine [25 mg/m
2 ] daily for 5 days, followed by a single intravenous infusion of autologous TILs and high-dose interleukin-2 [720 000 IU/kg] every 8 h). The primary endpoint was objective tumour response in evaluable patients per protocol using Response to Evaluation Criteria in Solid Tumors, version 1.0. An interim analysis of this trial is reported here. The trial is registered at ClinicalTrials.gov, number NCT01814046., Findings: From the completed first stage and ongoing expansion stage of this trial, a total of 21 consecutive patients with metastatic uveal melanoma were enrolled between June 7, 2013, and Sept 9, 2016, and received TIL therapy. Seven (35%, 95% CI 16-59) of 20 evaluable patients had objective tumour regression. Among the responders, six patients achieved a partial response, two of which are ongoing and have not reached maximum response. One patient achieved complete response of numerous hepatic metastases, currently ongoing at 21 months post therapy. Three of the responders were refractory to previous immune checkpoint blockade. Common grade 3 or worse toxic effects were related to the lymphodepleting chemotherapy regimen and included lymphopenia, neutropenia, and thrombocytopenia (21 [100%] patients for each toxicity); anaemia (14 [67%] patients); and infection (six [29%] patients). There was one treatment-related death secondary to sepsis-induced multiorgan failure., Interpretation: To our knowledge, this is the first report describing adoptive transfer of autologous TILs to mediate objective tumour regression in patients with metastatic uveal melanoma. These initial results challenge the belief that metastatic uveal melanoma is immunotherapy resistant and support the further investigation of immune-based therapies for this cancer. Refinement of this T-cell therapy is crucial to improve the frequency of clinical responses and the general applicability of this treatment modality., Funding: Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
16. Isolation of T-Cell Receptors Specifically Reactive with Mutated Tumor-Associated Antigens from Tumor-Infiltrating Lymphocytes Based on CD137 Expression.
- Author
-
Parkhurst M, Gros A, Pasetto A, Prickett T, Crystal JS, Robbins P, and Rosenberg SA
- Subjects
- Adoptive Transfer methods, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Exome genetics, Gene Expression Regulation, Neoplastic genetics, Gene Expression Regulation, Neoplastic immunology, High-Throughput Nucleotide Sequencing, Humans, Melanoma genetics, Melanoma immunology, Mutation, RNA, Messenger genetics, Receptors, Antigen, T-Cell genetics, Tumor Necrosis Factor Receptor Superfamily, Member 9 immunology, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy, Receptors, Antigen, T-Cell immunology
- Abstract
Purpose: The adoptive transfer of lymphocytes genetically modified to express tumor reactive T-cell receptors (TCR) can mediate tumor regression. Some tumor-infiltrating lymphocytes (TIL) recognize somatic mutations expressed only in the patient's tumors, and evidence suggests that clinically effective TILs target tumor-specific neoantigens. Here we attempted to isolate neoantigen-reactive TCRs as a prelude to the treatment of patients with autologous T cells genetically modified to express such TCRs. Experimental Design: Mutations expressed by tumors were identified using whole-exome and RNA sequencing. Tandem minigene (TMG) constructs encoding 12-24 mutated gene products were synthesized, each encoding the mutated amino acid flanked by 12 amino acids of the normal protein sequence. TILs were cultured with autologous dendritic cells (DC) transfected with in vitro transcribed (IVT) mRNAs encoding TMGs and were evaluated for IFNγ secretion and CD137 expression. Neoantigen-reactive T cells were enriched from TILs by sorting for CD137
+ CD8+ T cells and expanded in vitro Dominant TCR α and β chains were identified in the enriched populations using a combination of 5' rapid amplification of cDNA ends, deep sequencing of genomic DNA, PairSeq analysis, and single-cell RT-PCR analysis. Human PBL retrovirally transduced to express the TCRs were evaluated for recognition of relevant neoantigens. Results: We identified 27 TCRs from 6 patients that recognized 14 neoantigens expressed by autologous tumor cells. Conclusions: This strategy provides the means to generate T cells expressing neoantigen-reactive TCRs for use in future adoptive cell transfer immunotherapy trials for patients with cancer. Clin Cancer Res; 23(10); 2491-505. ©2016 AACR ., (©2016 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
17. Tumor- and Neoantigen-Reactive T-cell Receptors Can Be Identified Based on Their Frequency in Fresh Tumor.
- Author
-
Pasetto A, Gros A, Robbins PF, Deniger DC, Prickett TD, Matus-Nicodemos R, Douek DC, Howie B, Robins H, Parkhurst MR, Gartner J, Trebska-McGowan K, Crystal JS, and Rosenberg SA
- Subjects
- Adult, Aged, Biomarkers, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Clonal Evolution, Female, Humans, Immunophenotyping, Lymphocyte Count, Lymphocytes, Tumor-Infiltrating pathology, Male, Middle Aged, Mutation, Neoplasm Metastasis, Neoplasm Staging, Neoplasms pathology, T-Lymphocyte Subsets pathology, Young Adult, Antigens, Neoplasm metabolism, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Neoplasms immunology, Neoplasms metabolism, Receptors, Antigen, T-Cell metabolism, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism
- Abstract
Adoptive transfer of T cells with engineered T-cell receptor (TCR) genes that target tumor-specific antigens can mediate cancer regression. Accumulating evidence suggests that the clinical success of many immunotherapies is mediated by T cells targeting mutated neoantigens unique to the patient. We hypothesized that the most frequent TCR clonotypes infiltrating the tumor were reactive against tumor antigens. To test this hypothesis, we developed a multistep strategy that involved TCRB deep sequencing of the CD8(+)PD-1(+) T-cell subset, matching of TCRA-TCRB pairs by pairSEQ and single-cell RT-PCR, followed by testing of the TCRs for tumor-antigen specificity. Analysis of 12 fresh metastatic melanomas revealed that in 11 samples, up to 5 tumor-reactive TCRs were present in the 5 most frequently occurring clonotypes, which included reactivity against neoantigens. These data show the feasibility of developing a rapid, personalized TCR-gene therapy approach that targets the unique set of antigens presented by the autologous tumor without the need to identify their immunologic reactivity. Cancer Immunol Res; 4(9); 734-43. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
18. Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion Before Adoptive Transfer of Tumor-Infiltrating Lymphocytes for Patients With Metastatic Melanoma.
- Author
-
Goff SL, Dudley ME, Citrin DE, Somerville RP, Wunderlich JR, Danforth DN, Zlott DA, Yang JC, Sherry RM, Kammula US, Klebanoff CA, Hughes MS, Restifo NP, Langhan MM, Shelton TE, Lu L, Kwong ML, Ilyas S, Klemen ND, Payabyab EC, Morton KE, Toomey MA, Steinberg SM, White DE, and Rosenberg SA
- Subjects
- Adolescent, Adult, Aged, Female, Humans, Male, Melanoma immunology, Melanoma mortality, Middle Aged, Neoplasm Metastasis, Prospective Studies, Whole-Body Irradiation, Immunotherapy, Adoptive, Lymphocyte Depletion, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy
- Abstract
Purpose: Adoptive cell transfer, the infusion of large numbers of activated autologous lymphocytes, can mediate objective tumor regression in a majority of patients with metastatic melanoma (52 of 93; 56%). Addition and intensification of total body irradiation (TBI) to the preparative lymphodepleting chemotherapy regimen in sequential trials improved objective partial and complete response (CR) rates. Here, we evaluated the importance of adding TBI to the adoptive transfer of tumor-infiltrating lymphocytes (TIL) in a randomized fashion., Patients and Methods: A total of 101 patients with metastatic melanoma, including 76 patients with M1c disease, were randomly assigned to receive nonmyeloablative chemotherapy with or without 1,200 cGy TBI before transfer of tumor-infiltrating lymphcytes. Primary end points were CR rate (as defined by Response Evaluation Criteria in Solid Tumors v1.0) and overall survival (OS). Clinical and laboratory data were analyzed for correlates of response., Results: CR rates were 24% in both groups (12 of 50 v 12 of 51), and OS was also similar (median OS, 38.2 v 36.6 months; hazard ratio, 1.11; 95% CI, 0.65 to 1.91; P = .71). Thrombotic microangiopathy was an adverse event unique to the TBI arm and occurred in 13 of 48 treated patients. With a median potential follow-up of 40.9 months, only one of 24 patients who achieved a CR recurred., Conclusion: Adoptive cell transfer can mediate durable complete regressions in 24% of patients with metastatic melanoma, with median survival > 3 years. Results were similar using chemotherapy preparative regimens with or without addition of TBI., (© 2016 by American Society of Clinical Oncology.)
- Published
- 2016
- Full Text
- View/download PDF
19. Adoptive T-Cell Therapy for Cancer.
- Author
-
Yang JC and Rosenberg SA
- Subjects
- Animals, Antigens, Neoplasm genetics, Humans, Immunotherapy, Adoptive trends, Mice, Mutation, Neoplasms genetics, Antigens, Neoplasm immunology, Cell Cycle Checkpoints immunology, Cell Engineering, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms therapy, T-Lymphocytes immunology
- Abstract
Recent developments have demonstrated that immunotherapies are capable of achieving durable antitumor responses in patients with metastatic cancer. One modality that has been able to induce durable complete regressions in patients with melanoma has been adoptive cell therapy (ACT). This has slowly been expanded to other cancer types using new approaches such as genetically engineered T-cells and other methods of antigen targeting. It now appears that immune targeting of mutated "neoantigens" plays a major role in successful ACT, as well as in other immunotherapies such as checkpoint inhibitors. This realization presents not only new challenges to ACT but also new opportunities in that all tumors now may have potential antigens to attack that can be revealed by tumor genomic sequencing. There are a variety of exciting approaches to translate these new findings into clinical trials applying ACT to the majority of cancer types., (2016 Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
20. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes.
- Author
-
Cohen CJ, Gartner JJ, Horovitz-Fried M, Shamalov K, Trebska-McGowan K, Bliskovsky VV, Parkhurst MR, Ankri C, Prickett TD, Crystal JS, Li YF, El-Gamil M, Rosenberg SA, and Robbins PF
- Subjects
- Adolescent, Adult, Algorithms, Amino Acid Sequence, Antigen-Antibody Reactions, Antigens, Neoplasm classification, Antigens, Neoplasm genetics, Cells, Cultured, DNA, Neoplasm genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins immunology, Epitopes genetics, Epitopes immunology, Female, Genes, erbB-2, HLA-A1 Antigen chemistry, HLA-A1 Antigen immunology, HLA-A2 Antigen chemistry, HLA-A2 Antigen immunology, Humans, Interferon-gamma Release Tests, Male, Melanoma genetics, Middle Aged, Molecular Sequence Data, Mutation, Neoplasm Proteins genetics, Neoplasm Proteins immunology, Nuclear Proteins genetics, Nuclear Proteins immunology, Peptide Fragments immunology, Receptor, ErbB-2 immunology, TEA Domain Transcription Factors, Transcription Factors genetics, Transcription Factors immunology, Antigens, Neoplasm immunology, Exome, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Melanoma secondary, RNA, Neoplasm genetics, T-Cell Antigen Receptor Specificity, T-Lymphocytes immunology
- Abstract
Adoptively transferred tumor-infiltrating T lymphocytes (TILs) that mediate complete regression of metastatic melanoma have been shown to recognize mutated epitopes expressed by autologous tumors. Here, in an attempt to develop a strategy for facilitating the isolation, expansion, and study of mutated antigen-specific T cells, we performed whole-exome sequencing on matched tumor and normal DNA isolated from 8 patients with metastatic melanoma. Candidate mutated epitopes were identified using a peptide-MHC-binding algorithm, and these epitopes were synthesized and used to generate panels of MHC tetramers that were evaluated for binding to tumor digests and cultured TILs used for the treatment of patients. This strategy resulted in the identification of 9 mutated epitopes from 5 of the 8 patients tested. Cells reactive with 8 of the 9 epitopes could be isolated from autologous peripheral blood, where they were detected at frequencies that were estimated to range between 0.4% and 0.002%. To the best of our knowledge, this represents the first demonstration of the successful isolation of mutation-reactive T cells from patients' peripheral blood prior to immune therapy, potentially providing the basis for designing personalized immunotherapies to treat patients with advanced cancer.
- Published
- 2015
- Full Text
- View/download PDF
21. Clinical Scale Zinc Finger Nuclease-mediated Gene Editing of PD-1 in Tumor Infiltrating Lymphocytes for the Treatment of Metastatic Melanoma.
- Author
-
Beane JD, Lee G, Zheng Z, Mendel M, Abate-Daga D, Bharathan M, Black M, Gandhi N, Yu Z, Chandran S, Giedlin M, Ando D, Miller J, Paschon D, Guschin D, Rebar EJ, Reik A, Holmes MC, Gregory PD, Restifo NP, Rosenberg SA, Morgan RA, and Feldman SA
- Subjects
- Alleles, Animals, Cell Separation, Cytokines metabolism, Female, Flow Cytometry, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Green Fluorescent Proteins metabolism, Humans, Immunologic Memory, Immunotherapy, Adoptive, Interferon-gamma metabolism, Lymphocyte Activation immunology, Mice, Neoplasm Metastasis, Neoplasm Transplantation, Phenotype, Programmed Cell Death 1 Receptor metabolism, Tumor Necrosis Factor-alpha metabolism, Endoribonucleases genetics, Gene Expression Regulation, Neoplastic, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy, Programmed Cell Death 1 Receptor genetics, Zinc Fingers
- Abstract
Programmed cell death-1 (PD-1) is expressed on activated T cells and represents an attractive target for gene-editing of tumor targeted T cells prior to adoptive cell transfer (ACT). We used zinc finger nucleases (ZFNs) directed against the gene encoding human PD-1 (PDCD-1) to gene-edit melanoma tumor infiltrating lymphocytes (TIL). We show that our clinical scale TIL production process yielded efficient modification of the PD-1 gene locus, with an average modification frequency of 74.8% (n = 3, range 69.9-84.1%) of the alleles in a bulk TIL population, which resulted in a 76% reduction in PD-1 surface-expression. Forty to 48% of PD-1 gene-edited cells had biallelic PD-1 modification. Importantly, the PD-1 gene-edited TIL product showed improved in vitro effector function and a significantly increased polyfunctional cytokine profile (TNFα, GM-CSF, and IFNγ) compared to unmodified TIL in two of the three donors tested. In addition, all donor cells displayed an effector memory phenotype and expanded approximately 500-2,000-fold in vitro. Thus, further study to determine the efficiency and safety of adoptive cell transfer using PD-1 gene-edited TIL for the treatment of metastatic melanoma is warranted.
- Published
- 2015
- Full Text
- View/download PDF
22. Adoptive Cell Therapy--Tumor-Infiltrating Lymphocytes, T-Cell Receptors, and Chimeric Antigen Receptors.
- Author
-
Feldman SA, Assadipour Y, Kriley I, Goff SL, and Rosenberg SA
- Subjects
- Animals, Antigens, Neoplasm immunology, Cell- and Tissue-Based Therapy methods, Humans, Immunotherapy, Adoptive methods, T-Lymphocytes immunology, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms immunology, Neoplasms therapy, Receptors, Antigen immunology, Receptors, Antigen, T-Cell immunology
- Published
- 2015
- Full Text
- View/download PDF
23. Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma.
- Author
-
Zhang L, Morgan RA, Beane JD, Zheng Z, Dudley ME, Kassim SH, Nahvi AV, Ngo LT, Sherry RM, Phan GQ, Hughes MS, Kammula US, Feldman SA, Toomey MA, Kerkar SP, Restifo NP, Yang JC, and Rosenberg SA
- Subjects
- Adult, Aged, Cells, Cultured, Female, Genetic Engineering, Humans, Immunotherapy, Interleukin-12 biosynthesis, Lymphocytes, Tumor-Infiltrating transplantation, Male, Melanoma immunology, Melanoma secondary, Middle Aged, Skin Neoplasms immunology, Skin Neoplasms pathology, Transcriptional Activation, Treatment Outcome, Young Adult, Interleukin-12 genetics, Lymphocytes, Tumor-Infiltrating physiology, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Purpose: Infusion of interleukin-12 (IL12) can mediate antitumor immunity in animal models, yet its systemic administration to patients with cancer results in minimal efficacy and severe toxicity. Here, we evaluated the antitumor activity of adoptively transferred human tumor-infiltrating lymphocytes (TILs) genetically engineered to secrete single-chain IL12 selectively at the tumor site., Experimental Design: Thirty-three patients with metastatic melanoma were treated in a cell dose-escalation trial of autologous TILs transduced with a gene encoding a single-chain IL12 driven by a nuclear factor of the activated T cells promoter (NFAT.IL12). No IL2 was administered., Results: The administration of 0.001 to 0.1 × 10(9) NFAT.IL12-transduced TILs to 17 patients resulted in a single, objective response (5.9%). However, at doses between 0.3 and 3 × 10(9) cells, 10 of 16 patients (63%) exhibited objective clinical responses. The responses tended to be short, and the administered IL12-producing cells rarely persisted at 1 month. Increasing cell doses were associated with high serum levels of IL12 and IFNγ as well as clinical toxicities, including liver dysfunction, high fevers, and sporadic life-threatening hemodynamic instability., Conclusions: In this first-in-man trial, administration of TILs transduced with an inducible IL12 gene mediated tumor responses in the absence of IL2 administration using cell doses 10- to 100-fold lower than conventional TILs. However, due to toxicities, likely attributable to the secreted IL12, further refinement will be necessary before this approach can be safely used in the treatment of cancer patients., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
24. Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics.
- Author
-
Crompton JG, Sukumar M, Roychoudhuri R, Clever D, Gros A, Eil RL, Tran E, Hanada K, Yu Z, Palmer DC, Kerkar SP, Michalek RD, Upham T, Leonardi A, Acquavella N, Wang E, Marincola FM, Gattinoni L, Muranski P, Sundrud MS, Klebanoff CA, Rosenberg SA, Fearon DT, and Restifo NP
- Subjects
- Animals, Humans, Immunologic Memory, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating pathology, Melanoma immunology, Melanoma, Experimental immunology, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Transgenic, Proto-Oncogene Proteins c-akt immunology, Random Allocation, Tumor Cells, Cultured, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy, Melanoma, Experimental therapy, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors
- Abstract
Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) results in complete regression of advanced cancer in some patients, but the efficacy of this potentially curative therapy may be limited by poor persistence of TIL after adoptive transfer. Pharmacologic inhibition of the serine/threonine kinase Akt has recently been shown to promote immunologic memory in virus-specific murine models, but whether this approach enhances features of memory (e.g., long-term persistence) in TIL that are characteristically exhausted and senescent is not established. Here, we show that pharmacologic inhibition of Akt enables expansion of TIL with the transcriptional, metabolic, and functional properties characteristic of memory T cells. Consequently, Akt inhibition results in enhanced persistence of TIL after adoptive transfer into an immunodeficient animal model and augments antitumor immunity of CD8 T cells in a mouse model of cell-based immunotherapy. Pharmacologic inhibition of Akt represents a novel immunometabolomic approach to enhance the persistence of antitumor T cells and improve the efficacy of cell-based immunotherapy for metastatic cancer., (©2014 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
25. Human melanoma metastases demonstrate nonstochastic site-specific antigen heterogeneity that correlates with T-cell infiltration.
- Author
-
Bartlett EK, Fetsch PA, Filie AC, Abati A, Steinberg SM, Wunderlich JR, White DE, Stephens DJ, Marincola FM, Rosenberg SA, and Kammula US
- Subjects
- CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes pathology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes pathology, Cluster Analysis, Cohort Studies, Histocompatibility Antigens Class I metabolism, Histocompatibility Antigens Class II metabolism, Humans, Immunohistochemistry, Lymphocytes, Tumor-Infiltrating pathology, MART-1 Antigen metabolism, Melanoma pathology, Melanoma-Specific Antigens classification, Monophenol Monooxygenase metabolism, Neoplasm Metastasis, T-Lymphocytes pathology, gp100 Melanoma Antigen metabolism, Lymphocytes, Tumor-Infiltrating metabolism, Melanoma metabolism, Melanoma-Specific Antigens metabolism, T-Lymphocytes metabolism
- Abstract
Purpose: Metastasis heterogeneity presents a significant obstacle to the development of targeted cancer therapeutics. In this study, we sought to establish from a large series of human melanoma metastases whether there exists a determined pattern in tumor cellular heterogeneity that may guide the development of future targeted immunotherapies., Experimental Design: From a cohort of 1,514 patients with metastatic melanoma, biopsies were procured over a 17-year period from 3,086 metastatic tumors involving various anatomic sites. To allow specific tumor cell profiling, we used established immunohistochemical methods to perform semiquantitative assessment for a panel of prototypic melanocyte differentiation antigens (MDA), including gp100, MART-1, and tyrosinase. To gain insight into the endogenous host immune response against these tumors, we further characterized tumor cell expression of MHC I and MHC II and, also, the concomitant CD4(+) and CD8(+) T-cell infiltrate., Results: Tumor cell profiling for MDA expression demonstrated an anatomic site-specific pattern of antigen expression that was highest in brain, intermediate in soft tissues/lymph nodes, and lowest in visceral metastases. Hierarchical clustering analysis supported that melanoma metastases have a phylogenetically determined, rather than a stochastic, pattern of antigen expression that varies by anatomic site. Furthermore, tyrosinase expression was more frequently lost in metastatic sites outside of the brain and was uniquely correlated with both endogenous CD8(+) and CD4(+) T-cell infiltrates., Conclusion: Site-specific antigen heterogeneity represents a novel attribute for human melanoma metastases that should be considered in future therapy development and when assessing the responsiveness to antigen-specific immunotherapies., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
26. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer.
- Author
-
Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP, Hogan K, Hinrichs CS, Parkhurst MR, Yang JC, and Rosenberg SA
- Subjects
- Adult, Bile Duct Neoplasms genetics, Cholangiocarcinoma genetics, Clinical Trials, Phase II as Topic, Exome, Female, Humans, Mutation, Receptor, ErbB-2 metabolism, Adaptor Proteins, Signal Transducing genetics, Adoptive Transfer methods, Bile Duct Neoplasms therapy, Bile Ducts, Intrahepatic, CD4-Positive T-Lymphocytes immunology, Cholangiocarcinoma therapy, Lymphocytes, Tumor-Infiltrating transplantation, Th1 Cells transplantation
- Abstract
Limited evidence exists that humans mount a mutation-specific T cell response to epithelial cancers. We used a whole-exomic-sequencing-based approach to demonstrate that tumor-infiltrating lymphocytes (TIL) from a patient with metastatic cholangiocarcinoma contained CD4+ T helper 1 (T(H)1) cells recognizing a mutation in erbb2 interacting protein (ERBB2IP) expressed by the cancer. After adoptive transfer of TIL containing about 25% mutation-specific polyfunctional T(H)1 cells, the patient achieved a decrease in target lesions with prolonged stabilization of disease. Upon disease progression, the patient was retreated with a >95% pure population of mutation-reactive T(H)1 cells and again experienced tumor regression. These results provide evidence that a CD4+ T cell response against a mutated antigen can be harnessed to mediate regression of a metastatic epithelial cancer.
- Published
- 2014
- Full Text
- View/download PDF
27. Tumor-reactive CD8+ T cells in metastatic gastrointestinal cancer refractory to chemotherapy.
- Author
-
Turcotte S, Gros A, Tran E, Lee CC, Wunderlich JR, Robbins PF, and Rosenberg SA
- Subjects
- Adult, Autografts, CD8-Positive T-Lymphocytes metabolism, Cell Line, Tumor, Female, Gastrointestinal Neoplasms genetics, Histocompatibility Antigens Class I immunology, Humans, Lymphocytes, Tumor-Infiltrating metabolism, Male, Middle Aged, Neoplasm Metastasis, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, Transduction, Genetic, CD8-Positive T-Lymphocytes immunology, Gastrointestinal Neoplasms immunology, Gastrointestinal Neoplasms pathology, Lymphocytes, Tumor-Infiltrating immunology
- Abstract
Purpose: To evaluate whether patients with metastatic gastrointestinal adenocarcinomas refractory to chemotherapy harbor tumor-reactive cytotoxic T cells., Experimental Design: Expansion of CD8(+) tumor-infiltrating lymphocytes (TIL) and cancer cell lines was attempted from gastrointestinal cancer metastases in 16 consecutive patients for the study of antitumor immune recognition. Retroviral transduction of genes encoding T-cell receptors (TCR) was used to define HLA-restriction elements and specific reactivity., Results: TIL were expanded from metastases in all patients, and new tumor cell lines were generated in 5 patients. Autologous tumor recognition without cross-reactivity against allogeneic HLA-matched gastrointestinal tumors was found in CD8(+) TIL from 3 of these 5 patients. In a patient with gastric cancer liver metastases, the repertoire of CD8(+) TIL was dominated by cytolytic sister clones reactive to 2 out of 4 autologous cancer cell lines restricted by HLA-C*0701. From the same patient, a rare CD8(+) TIL clone with a distinct TCR recognized all four cancer cell lines restricted by HLA-B*4901. In a patient with bile duct cancer, two distinct antitumor cytolytic clones were isolated from a highly polyclonal CD8(+) TIL repertoire. TCRs isolated from these clones recognized epitopes restricted by HLA-A*0201. In a third patient, CD8(+) TIL reactivity was progressively lost against an autologous colon cancer cell line that displayed loss of HLA haplotype., Conclusions: This study provides a basis for the development of immunotherapy for patients with advanced gastrointestinal malignancies by first establishing the presence of naturally occurring tumor-reactive CD8(+) TIL at the molecular level., (©2013 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
28. Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy.
- Author
-
Turcotte S, Gros A, Hogan K, Tran E, Hinrichs CS, Wunderlich JR, Dudley ME, and Rosenberg SA
- Subjects
- Female, Flow Cytometry, Gastrointestinal Neoplasms pathology, Humans, Immunohistochemistry, Lymphocytes, Tumor-Infiltrating cytology, Male, Melanoma pathology, Middle Aged, Neoplasm Metastasis immunology, Neoplasm Metastasis pathology, Phenotype, T-Lymphocytes cytology, Gastrointestinal Neoplasms immunology, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, T-Lymphocytes immunology
- Abstract
Adoptive cell transfer of tumor-infiltrating lymphocytes (TILs) can mediate cancer regression in patients with metastatic melanoma, but whether this approach can be applied to common epithelial malignancies remains unclear. In this study, we compared the phenotype and function of TILs derived from liver and lung metastases from patients with gastrointestinal (GI) cancers (n = 14) or melanoma (n = 42). Fewer CD3(+) T cells were found to infiltrate GI compared with melanoma metastases, but the proportions of CD8(+) cells, T cell differentiation stage, and expression of costimulatory molecules were similar for both tumor types. Clinical-scale expansion up to ~50 × 10(9) T cells on average was obtained for all patients with GI cancer and melanoma. From GI tumors, however, TIL outgrowth in high-dose IL-2 yielded 22 ± 1.4% CD3(+)CD8(+) cells compared with 63 ± 2.4% from melanoma (p < 0.001). IFN-γ ELISA demonstrated MHC class I-mediated reactivity of TIL against autologous tumor in 5 of 7 GI cancer patients tested (9% of 188 distinct TIL cultures) and in 9 of 10 melanoma patients (43% of 246 distinct TIL cultures). In these assays, MHC class I-mediated up-regulation of CD137 (4-1BB) expression on CD8(+) cells suggested that 0-3% of TILs expanded from GI cancer metastases were tumor-reactive. This study implies that the main challenge to the development of TIL adoptive cell transfer for metastatic GI cancers may not be the in vitro expansion of bulk TILs, but the ability to select and enrich for tumor-reactive T cells.
- Published
- 2013
- Full Text
- View/download PDF
29. Mutated PPP1R3B is recognized by T cells used to treat a melanoma patient who experienced a durable complete tumor regression.
- Author
-
Lu YC, Yao X, Li YF, El-Gamil M, Dudley ME, Yang JC, Almeida JR, Douek DC, Samuels Y, Rosenberg SA, and Robbins PF
- Subjects
- Base Sequence, Clinical Trials as Topic, Flow Cytometry, Gene Knockdown Techniques, Gene Library, Humans, Lymphocyte Activation immunology, Male, Melanoma genetics, Molecular Sequence Data, Mutation, Phosphoprotein Phosphatases genetics, Protein Phosphatase 1 genetics, Reverse Transcriptase Polymerase Chain Reaction, Immunodominant Epitopes immunology, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Phosphoprotein Phosphatases immunology, Protein Phosphatase 1 immunology
- Abstract
Adoptive cell therapy with tumor-infiltrating lymphocytes (TILs) represents an effective treatment for patients with metastatic melanoma. However, most of the Ag targets recognized by effective melanoma-reactive TILs remain elusive. In this study, patient 2369 experienced a complete response, including regressions of bulky liver tumor masses, ongoing beyond 7 y following adoptive TIL transfer. The screening of a cDNA library generated from the autologous melanoma cell line resulted in the isolation of a mutated protein phosphatase 1, regulatory (inhibitor) subunit 3B (PPP1R3B) gene product. The mutated PPP1R3B peptide represents the immunodominant epitope recognized by tumor-reactive T cells in TIL 2369. Five years following adoptive transfer, peripheral blood T lymphocytes obtained from patient 2369 recognized the mutated PPP1R3B epitope. These results demonstrate that adoptive T cell therapy targeting a tumor-specific Ag can mediate long-term survival for a patient with metastatic melanoma. This study also provides an impetus to develop personalized immunotherapy targeting tumor-specific, mutated Ags.
- Published
- 2013
- Full Text
- View/download PDF
30. Randomized selection design trial evaluating CD8+-enriched versus unselected tumor-infiltrating lymphocytes for adoptive cell therapy for patients with melanoma.
- Author
-
Dudley ME, Gross CA, Somerville RP, Hong Y, Schaub NP, Rosati SF, White DE, Nathan D, Restifo NP, Steinberg SM, Wunderlich JR, Kammula US, Sherry RM, Yang JC, Phan GQ, Hughes MS, Laurencot CM, and Rosenberg SA
- Subjects
- Adolescent, Adult, Aged, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes pathology, CD8-Positive T-Lymphocytes transplantation, Child, Female, Humans, Immunotherapy, Adoptive adverse effects, Interferon-gamma immunology, Interleukin-2 administration & dosage, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating pathology, Lymphocytes, Tumor-Infiltrating transplantation, Male, Melanoma immunology, Middle Aged, Prospective Studies, Tumor Cells, Cultured, Young Adult, CD8-Positive T-Lymphocytes immunology, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy
- Abstract
Purpose: Adoptive cell therapy (ACT) with autologous tumor-infiltrating lymphocytes (TILs) and high-dose interleukin-2 (IL-2) administered to lymphodepleted patients with melanoma can cause durable tumor regressions. The optimal TIL product for ACT is unknown., Patients and Methods: Patients with metastatic melanoma were prospectively assigned to receive unselected young TILs versus CD8(+)-enriched TILs. All patients received lymphodepleting chemotherapy and high-dose IL-2 therapy and were assessed for response, toxicity, survival, and immunologic end points., Results: Thirty-four patients received unselected young TILs with a median of 8.0% CD4(+) lymphocytes, and 35 patients received CD8(+)-enriched TILs with a median of 0.3% CD4(+) lymphocytes. One month after TIL infusion, patients who received CD8(+)-enriched TILs had significantly fewer CD4(+) peripheral blood lymphocytes (P = .01). Twelve patients responded to therapy with unselected young TILs (according to Response Evaluation Criteria in Solid Tumors [RECIST]), and seven patients responded to CD8(+)-enriched TILs (35% v 20%; not significant). Retrospective studies showed a significant association between response to treatment and interferon gamma secretion by the infused TILs in response to autologous tumor (P = .04), and in the subgroup of patients who received TILs from subcutaneous tumors, eight of 15 patients receiving unselected young TILs responded but none of eight patients receiving CD8(+)-enriched TILs responded., Conclusion: A randomized selection design trial was feasible for improving individualized TIL therapy. Since the evidence indicates that CD8(+)-enriched TILs are not more potent therapeutically and they are more laborious to prepare, future studies should focus on unselected young TILs.
- Published
- 2013
- Full Text
- View/download PDF
31. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells.
- Author
-
Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E, Samuels Y, and Rosenberg SA
- Subjects
- Adult, Antigens, Neoplasm immunology, Epitopes, T-Lymphocyte, Female, HLA-A Antigens metabolism, Humans, Male, Melanoma immunology, Middle Aged, Adoptive Transfer, Antigens, Neoplasm genetics, Exome, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy, Mutation
- Abstract
Substantial regressions of metastatic lesions have been observed in up to 70% of patients with melanoma who received adoptively transferred autologous tumor-infiltrating lymphocytes (TILs) in phase 2 clinical trials. In addition, 40% of patients treated in a recent trial experienced complete regressions of all measurable lesions for at least 5 years following TIL treatment. To evaluate the potential association between the ability of TILs to mediate durable regressions and their ability to recognize potent antigens that presumably include mutated gene products, we developed a new screening approach involving mining whole-exome sequence data to identify mutated proteins expressed in patient tumors. We then synthesized and evaluated candidate mutated T cell epitopes that were identified using a major histocompatibility complex-binding algorithm for recognition by TILs. Using this approach, we identified mutated antigens expressed on autologous tumor cells that were recognized by three bulk TIL lines from three individuals with melanoma that were associated with objective tumor regressions following adoptive transfer. This simplified approach for identifying mutated antigens recognized by T cells avoids the need to generate and laboriously screen cDNA libraries from tumors and may represent a generally applicable method for identifying mutated antigens expressed in a variety of tumor types.
- Published
- 2013
- Full Text
- View/download PDF
32. Modulating the differentiation status of ex vivo-cultured anti-tumor T cells using cytokine cocktails.
- Author
-
Yang S, Ji Y, Gattinoni L, Zhang L, Yu Z, Restifo NP, Rosenberg SA, and Morgan RA
- Subjects
- Animals, CD8-Positive T-Lymphocytes cytology, Cell Differentiation drug effects, Cell Differentiation immunology, Humans, Interleukins immunology, Interleukins pharmacology, Lymphocytes, Tumor-Infiltrating cytology, Melanoma immunology, Melanoma pathology, Mice, Mice, Inbred NOD, Mice, SCID, Phenotype, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, Cytokines immunology, Cytokines pharmacology, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology
- Abstract
The genetic modification of CD8+ T cells using anti-tumor T-cell receptors (TCR) or chimeric antigen receptors is a promising approach for the adoptive cell therapy of patients with cancer. We previously developed a simplified method for the clinical-scale generation of central memory-like (Tcm) CD8+ T cells following transduction with lentivirus encoding anti-tumor TCR and culture in the presence of IL-2. In this study, we compared different cytokines or combinations of IL-2, IL-7, IL-12, IL-15, and IL-21 to expand genetically engineered CD8+ T cells. We demonstrated that specific cytokine combinations IL-12 plus IL-7 or IL-21 for 3 days followed by withdrawal of IL-12 yielded the phenotype of CD62L(high)CD28(high) CD127(high)CD27(high)CCR7(high), which is associated with less-differentiated T cells. Genes associated with stem cells (SOX2, NANOG, OCT4, and LIN28A), were also up-regulated by this cytokine cocktail. Moreover, the use of IL-12 plus IL-7 or IL-21 yielded CD8 T cells showing enhanced persistence in the NOD/SCID/γc-/- mouse model. This defined cytokine combination could also alter highly differentiated TIL from melanoma patients into cells with a less-differentiated phenotype. The methodology that we developed for generating a less-differentiated anti-tumor CD8+ T cells ex vivo may be ideal for the adoptive immunotherapy of cancer.
- Published
- 2013
- Full Text
- View/download PDF
33. Minimally invasive liver resection to obtain tumor-infiltrating lymphocytes for adoptive cell therapy in patients with metastatic melanoma.
- Author
-
Alvarez-Downing MM, Inchauste SM, Dudley ME, White DE, Wunderlich JR, Rosenberg SA, and Kammula US
- Subjects
- Adult, Aged, Combined Modality Therapy, Feasibility Studies, Female, Follow-Up Studies, Humans, Interleukin-10 therapeutic use, Interleukin-2 therapeutic use, Laparoscopy, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Male, Melanoma immunology, Melanoma mortality, Melanoma secondary, Middle Aged, Prognosis, Prospective Studies, Remission Induction, Retrospective Studies, Survival Rate, Cell- and Tissue-Based Therapy, Hepatectomy, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating transplantation, Melanoma therapy, Minimally Invasive Surgical Procedures
- Abstract
Background: Adoptive cell therapy (ACT) with tumor-infiltrating lymphocytes (TIL) in patients with metastatic melanoma has been reported to have a 56% overall response rate with 20% complete responders. To increase the availability of this promising therapy in patients with advanced melanoma, a minimally invasive approach to procure tumor for TIL generation is warranted., Methods: A feasibility study was performed to determine the safety and efficacy of laparoscopic liver resection to generate TIL for ACT. Retrospective review of a prospectively maintained database identified 22 patients with advanced melanoma and visceral metastasis (AJCC Stage M1c) who underwent laparoscopic liver resection between 1 October 2005 and 31 July 2011. The indication for resection in all patients was to receive postoperative ACT with TIL., Results: Twenty patients (91%) underwent resection utilizing a closed laparoscopic technique, one required hand-assistance and another required conversion to open resection. Median intraoperative blood loss was 100 mL with most cases performed without a Pringle maneuver. Median hospital stay was 3 days. Three (14%) patients experienced a complication from resection with no mortality. TIL were generated from 18 of 22 (82%) patients. Twelve of 15 (80%) TIL tested were found to have in vitro tumor reactivity. Eleven patients (50%) received the intended ACT. Two patients were rendered no evidence of disease after surgical resection, with one undergoing delayed ACT with generated TIL after relapse. Objective tumor response was seen in 5 of 11 patients (45%) who received TIL, with one patient experiencing an ongoing complete response (32+ months)., Conclusions: Laparoscopic liver resection can be performed with minimal morbidity and serve as an effective means to procure tumor to generate therapeutic TIL for ACT to patients with metastatic melanoma.
- Published
- 2012
- Full Text
- View/download PDF
34. Tumor-specific CD4+ melanoma tumor-infiltrating lymphocytes.
- Author
-
Friedman KM, Prieto PA, Devillier LE, Gross CA, Yang JC, Wunderlich JR, Rosenberg SA, and Dudley ME
- Subjects
- CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes transplantation, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Cell Proliferation, Female, HLA-DR Antigens immunology, Humans, Interferon-gamma biosynthesis, Interferon-gamma immunology, Interleukin-2 administration & dosage, Interleukin-2 therapeutic use, Liver Neoplasms immunology, Liver Neoplasms secondary, Lymphocyte Depletion, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Melanoma pathology, Organ Specificity, Skin Neoplasms immunology, Skin Neoplasms pathology, Splenic Neoplasms immunology, Splenic Neoplasms secondary, Tumor Cells, Cultured, CD4-Positive T-Lymphocytes immunology, Immunotherapy, Adoptive methods, Liver Neoplasms therapy, Lymphocytes, Tumor-Infiltrating transplantation, Melanoma therapy, Skin Neoplasms therapy, Splenic Neoplasms therapy
- Abstract
Adoptive cell therapy using tumor-infiltrating lymphocytes (TIL) can mediate objective and durable tumor regressions in patients with metastatic melanoma. CD8+ tumor-reactive TIL are well studied in humans and animals, yet the function of tumor-infiltrating CD4+ T lymphocytes in patient treatments remains controversial. We recently demonstrated that CD4+ TILs are not necessary for objective responses in patients. Coinfusion with tumor-specific CD4 TIL may enhance or increase the durability of tumor regressions, but the number of patients with tumor-reactive CD4 TIL is unknown. We screened 44 CD8+-depleted TIL for in vitro reactivity against autologous tumor. Nine (20%) showed specific reactivity by interferon-γ release assay, of which 8 were specifically blocked by an anti-HLA-DR antibody. Flow-cytometric analysis of these reactive TIL confirmed a high CD4+ composition (median 89%). Highlighting the contribution of CD4+ TIL to tumor regression, a patient with widespread metastatic disease was administered TIL containing HLA class II-restricted tumor activity with high-dose interleukin-2 therapy after lymphodepletion that mediated regression of extensive metastatic disease in the liver and spleen. These results demonstrate that at least 20% of metastatic melanomas contain CD4+ lymphocytes with specific tumor recognition and suggest a possible role for CD4+ cells in the effectiveness of adoptive cell therapy.
- Published
- 2012
- Full Text
- View/download PDF
35. Simplified method of the growth of human tumor infiltrating lymphocytes in gas-permeable flasks to numbers needed for patient treatment.
- Author
-
Jin J, Sabatino M, Somerville R, Wilson JR, Dudley ME, Stroncek DF, and Rosenberg SA
- Subjects
- Humans, Primary Cell Culture standards, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating cytology, Primary Cell Culture instrumentation, Primary Cell Culture methods
- Abstract
Adoptive cell therapy of metastatic melanoma with autologous tumor infiltrating lymphocytes (TIL) is clinically effective, but TIL production can be challenging. Here we describe a simplified method for initial TIL culture and rapid expansion in gas-permeable flasks. TIL were initially cultured from tumor digests and fragments in 40 mL capacity flasks with a 10 cm² gas-permeable silicone bottom, G-Rex10. A TIL rapid expansion protocol (REP) was developed using 500 mL capacity flasks with a 100 cm² gas-permeable silicone bottom, G-Rex100. TIL growth was successfully initiated in G-Rex10 flasks from tumor digests from 13 of 14 patients and from tumor fragments in all 11 tumor samples tested. TIL could then be expanded to 8-10×10⁹ cells in a 2-step REP that began by seeding 5×10⁶ TIL into a G-Rex100 flask, followed by expansion at day 7 into 3 G-Rex100 flasks. To obtain the 30-60×10⁹ cells used for patient treatment, we seeded 6 G-Rex100 flasks with 5×10⁶ cells and expanded into 18 G-Rex100 flasks. Large-scale TIL REP in gas-permeable flasks requires approximately 9-10 L of media, about 3-4 times less than other methods. In conclusion, TIL initiation and REP in gas-permeable G-Rex flasks require fewer total vessels, less media, less incubator space, and less labor than initiation and REP in 24-well plates, tissue culture flasks, and bags. TIL culture in G-Rex flasks will facilitate the production of TIL at the numbers required for patient treatment at most cell processing laboratories.
- Published
- 2012
- Full Text
- View/download PDF
36. Augmented lymphocyte expansion from solid tumors with engineered cells for costimulatory enhancement.
- Author
-
Friedman KM, Devillier LE, Feldman SA, Rosenberg SA, and Dudley ME
- Subjects
- 4-1BB Ligand genetics, 4-1BB Ligand metabolism, Antigens, CD genetics, Antigens, CD metabolism, Cell Count, Cell Proliferation, Genetic Engineering, Humans, Immunologic Memory, Immunophenotyping, Interferon-gamma metabolism, K562 Cells immunology, Killer Cells, Natural pathology, Lymphocyte Activation, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Melanoma immunology, Melanoma pathology, Neoplasm Metastasis, Receptor Cross-Talk immunology, Skin Neoplasms immunology, Skin Neoplasms pathology, Transgenes genetics, CD8-Positive T-Lymphocytes pathology, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating metabolism, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Treatment of patients with adoptive T-cell therapy requires expansion of unique tumor-infiltrating lymphocyte (TIL) cultures from single-cell suspensions processed from melanoma biopsies. Strategies which increase the expansion and reliability of TIL generation from tumor digests are necessary to improve access to TIL therapy. Previous studies have evaluated artificial antigen presenting cells for their antigen-specific and costimulatory properties. We investigated engineered cells for costimulatory enhancement (ECCE) consisting of K562 cells that express 4-1BBL in the absence of artificial antigen stimulation. ECCE accelerated TIL expansion and significantly improved TIL numbers (P=0.001) from single-cell melanoma suspensions. TIL generated with ECCE contain significantly more CD8CD62L and CD8CD27 T cells then comparable interleukin-2-expanded TIL and maintained antitumor reactivity. Moreover, ECCE improved TIL expansion from nonmelanoma-cell suspensions similar to that seen with melanoma tumors. These data demonstrate that the addition of ECCE to TIL production will enable the treatment of patients that are ineligible using current methods.
- Published
- 2011
- Full Text
- View/download PDF
37. Cell transfer immunotherapy for metastatic solid cancer--what clinicians need to know.
- Author
-
Rosenberg SA
- Subjects
- Adoptive Transfer, Humans, Neoplasm Metastasis, Neoplasms immunology, T-Lymphocytes cytology, Immunotherapy, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms pathology, Neoplasms therapy, T-Lymphocytes transplantation
- Abstract
Cancer immunotherapy using the adoptive transfer of autologous tumor-infiltrating lymphocytes results in objective cancer regression in 49-72% of patients with metastatic melanoma. In a pilot trial combining cell transfer with a maximum lymphodepleting regimen, complete durable responses were seen in 40% of patients, with complete responses ongoing beyond 3 to 7 years. Current approaches to cell transfer therapy using autologous cells genetically engineered to express conventional or chimeric T-cell receptors have mediated cancer regression in patients with metastatic melanoma, synovial sarcoma, neuroblastoma and refractory lymphoma. Adoptive cell transfer immunotherapy is a rapidly developing new approach to the therapy of metastatic cancer in humans. This Review will emphasize the current available applications of cell transfer immunotherapy for patients with cancer.
- Published
- 2011
- Full Text
- View/download PDF
38. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy.
- Author
-
Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, and Dudley ME
- Subjects
- Adolescent, Adult, Aged, Combined Modality Therapy, Female, Humans, Lymphocyte Count, Male, Melanoma mortality, Melanoma pathology, Middle Aged, Survival Analysis, Telomere genetics, Treatment Outcome, Young Adult, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy
- Abstract
Purpose: Most treatments for patients with metastatic melanoma have a low rate of complete regression and thus overall survival in these patients is poor. We investigated the ability of adoptive cell transfer utilizing autologous tumor-infiltrating lymphocytes (TIL) to mediate durable complete regressions in heavily pretreated patients with metastatic melanoma., Experimental Design: Ninety-three patients with measurable metastatic melanoma were treated with the adoptive transfer of autologous TILs administered in conjunction with interleukin-2 following a lymphodepleting preparative regimen on three sequential clinical trials. Ninety-five percent of these patients had progressive disease following a prior systemic treatment. Median potential follow-up was 62 months., Results: Objective response rates by Response Evaluation Criteria in Solid Tumors (RECIST) in the 3 trials using lymphodepleting preparative regimens (chemotherapy alone or with 2 or 12 Gy irradiation) were 49%, 52%, and 72%, respectively. Twenty of the 93 patients (22%) achieved a complete tumor regression, and 19 have ongoing complete regressions beyond 3 years. The actuarial 3- and 5-year survival rates for the entire group were 36% and 29%, respectively, but for the 20 complete responders were 100% and 93%. The likelihood of achieving a complete response was similar regardless of prior therapy. Factors associated with objective response included longer telomeres of the infused cells, the number of CD8(+)CD27(+) cells infused, and the persistence of the infused cells in the circulation at 1 month (all P(2) < 0.001)., Conclusions: Cell transfer therapy with autologous TILs can mediate durable complete responses in patients with metastatic melanoma and has similar efficacy irrespective of prior treatment. Clin Cancer Res; 17(13); 4550-7. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
39. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1.
- Author
-
Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, Nahvi AV, Helman LJ, Mackall CL, Kammula US, Hughes MS, Restifo NP, Raffeld M, Lee CC, Levy CL, Li YF, El-Gamil M, Schwarz SL, Laurencot C, and Rosenberg SA
- Subjects
- Adult, Cancer Vaccines immunology, Epigenesis, Genetic, Female, Genetic Engineering, Humans, Male, Melanoma immunology, Melanoma mortality, Melanoma secondary, Middle Aged, Neoplasm Metastasis, Neoplasm Staging, Prognosis, Risk Assessment, Sarcoma, Synovial immunology, Sarcoma, Synovial mortality, Sarcoma, Synovial secondary, Skin Neoplasms immunology, Skin Neoplasms mortality, Skin Neoplasms pathology, Survival Analysis, Treatment Outcome, Young Adult, Cancer Vaccines administration & dosage, Immunotherapy methods, Lymphocytes, Tumor-Infiltrating immunology, Melanoma therapy, Sarcoma, Synovial therapy, Skin Neoplasms therapy
- Abstract
Purpose: Adoptive immunotherapy using tumor-infiltrating lymphocytes represents an effective cancer treatment for patients with metastatic melanoma. The NY-ESO-1 cancer/testis antigen, which is expressed in 80% of patients with synovial cell sarcoma and approximately 25% of patients with melanoma and common epithelial tumors, represents an attractive target for immune-based therapies. The current trial was carried out to evaluate the ability of adoptively transferred autologous T cells transduced with a T-cell receptor (TCR) directed against NY-ESO-1 to mediate tumor regression in patients with metastatic melanoma and synovial cell sarcoma., Patients and Methods: A clinical trial was performed in patients with metastatic melanoma or metastatic synovial cell sarcoma refractory to all standard treatments. Patients with NY-ESO-1-positive tumors were treated with autologous TCR-transduced T cells plus 720,000 iU/kg of interleukin-2 to tolerance after preparative chemotherapy. Objective clinical responses were evaluated using Response Evaluation Criteria in Solid Tumors (RECIST)., Results: Objective clinical responses were observed in four of six patients with synovial cell sarcoma and five of 11 patients with melanoma bearing tumors expressing NY-ESO-1. Two of 11 patients with melanoma demonstrated complete regressions that persisted after 1 year. A partial response lasting 18 months was observed in one patient with synovial cell sarcoma., Conclusion: These observations indicate that TCR-based gene therapies directed against NY-ESO-1 represent a new and effective therapeutic approach for patients with melanoma and synovial cell sarcoma. To our knowledge, this represents the first demonstration of the successful treatment of a nonmelanoma tumor using TCR-transduced T cells.
- Published
- 2011
- Full Text
- View/download PDF
40. CD8+ enriched "young" tumor infiltrating lymphocytes can mediate regression of metastatic melanoma.
- Author
-
Dudley ME, Gross CA, Langhan MM, Garcia MR, Sherry RM, Yang JC, Phan GQ, Kammula US, Hughes MS, Citrin DE, Restifo NP, Wunderlich JR, Prieto PA, Hong JJ, Langan RC, Zlott DA, Morton KE, White DE, Laurencot CM, and Rosenberg SA
- Subjects
- Adult, Aged, Aged, 80 and over, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Cells, Cultured, Combined Modality Therapy, Female, Humans, Lymphocyte Count, Lymphocytes, Tumor-Infiltrating cytology, Lymphocytes, Tumor-Infiltrating immunology, Male, Melanoma immunology, Melanoma pathology, Middle Aged, Neoplasm Metastasis, Tumor Burden immunology, CD8-Positive T-Lymphocytes transplantation, Cytotoxicity, Immunologic physiology, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating transplantation, Melanoma therapy
- Abstract
Purpose: Tumor-infiltrating lymphocytes (TIL) and interleukin (IL)-2 administered following lymphodepletion can cause the durable complete regression of bulky metastatic melanoma in patients refractory to approved treatments. However, the generation of a unique tumor-reactive TIL culture for each patient may be prohibitively difficult. We therefore investigated the clinical and immunologic impact of unscreened, CD8+ enriched "young" TIL., Experimental Design: Methods were developed for generating TIL that minimized the time in culture and eliminated the individualized tumor-reactivity screening step. Thirty-three patients were treated with these CD8+ enriched young TIL and IL-2 following nonmyeloablative lymphodepletion (NMA). Twenty-three additional patients were treated with CD8+ enriched young TIL and IL-2 after lymphodepletion with NMA and 6 Gy of total body irradiation., Results: Young TIL cultures for therapy were successfully established from 83% of 122 consecutive melanoma patients. Nineteen of 33 patients (58%) treated with CD8+ enriched young TIL and NMA had an objective response (Response Evaluation Criteria in Solid Tumors) including 3 complete responders. Eleven of 23 patients (48%) treated with TIL and 6 Gy total body irradiation had an objective response including 2 complete responders. At 1 month after TIL infusion the absolute CD8+ cell numbers in the periphery were highly correlated with response., Conclusions: This study shows that a rapid and simplified method can be used to reliably generate CD8+ enriched young TIL for administration as an individualized therapy for advanced melanoma, and may allow this potentially effective treatment to be applied at other institutions and to reach additional patients., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
41. Identification and characterization of a tumor infiltrating CD56(+)/CD16 (-) NK cell subset with specificity for pancreatic and prostate cancer cell lines.
- Author
-
Frankel TL, Burns W, Riley J, Morgan RA, Davis JL, Hanada K, Quezado M, Rosenberg SA, and Royal RE
- Subjects
- Cell Line, Tumor, Female, GPI-Linked Proteins, HLA-A2 Antigen immunology, Humans, Immunophenotyping, Immunotherapy, Interferon-gamma biosynthesis, Male, Pancreatic Neoplasms pathology, Pancreatic Neoplasms therapy, Prostatic Neoplasms pathology, Prostatic Neoplasms therapy, TNF-Related Apoptosis-Inducing Ligand physiology, CD56 Antigen analysis, Killer Cells, Natural immunology, Lymphocytes, Tumor-Infiltrating immunology, Pancreatic Neoplasms immunology, Prostatic Neoplasms immunology, Receptors, IgG analysis
- Abstract
In a recent clinical trial, a patient exhibited regression of several pancreatic cancer metastases following the administration of the immune modulator Ipilimumab (anti-CTLA-4 antibody). We sought to characterize the immune cells responsible for this regression. Tumor infiltrating lymphocytes (TIL-2742) and an autologous tumor line (TC-2742) were expanded from a regressing metastatic lesion excised from this patient. Natural killer (NK) cells predominated in the TIL (92% CD56(+)) with few T cells (12% CD3(+)). A majority (88%) of the NK cells were CD56(bright)CD16(-). TIL-2742 secreted IFN-γ and GM-CSF following co-culture with TC-2742 and major histocompatibility complex mismatched pancreatic tumor lines. After sorting TIL-2742, the purified CD56(+)CD16(-)CD3(-) subset showed reactivity similar to TIL-2742 while the CD56(-)CD16(-)CD3(+) cells exhibited no tumor recognition. In co-culture assays, TIL-2742 and the NK subset expressed high reactivity to several pancreatic and prostate cancer cell lines and could lyse the autologous tumor as well as pancreas and prostate cancer lines. Reactivity was partially abrogated by blockade of TRAIL. We thus identified a unique subset of NK cells (CD56(bright)CD16(dim)) isolated from a regressing metastatic pancreatic cancer in a patient responding to Ipilimumab. This represents the first report of CD56(+)CD16(-) NK cells with apparent specificity for pancreatic and prostate cancer cell lines and associated with tumor regression following the treatment with an immune modulating agent.
- Published
- 2010
- Full Text
- View/download PDF
42. Tumor infiltrating lymphocyte therapy for metastatic melanoma: analysis of tumors resected for TIL.
- Author
-
Goff SL, Smith FO, Klapper JA, Sherry R, Wunderlich JR, Steinberg SM, White D, Rosenberg SA, Dudley ME, and Yang JC
- Subjects
- Adolescent, Adult, Aged, Antigens, Neoplasm immunology, Cell Line, Tumor, Cell Proliferation, Female, Gastrointestinal Neoplasms immunology, Gastrointestinal Neoplasms secondary, Gastrointestinal Neoplasms surgery, HLA-A Antigens metabolism, HLA-A2 Antigen, Humans, Interferon-gamma metabolism, Lung Neoplasms immunology, Lung Neoplasms secondary, Lung Neoplasms surgery, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Lymphocytes, Tumor-Infiltrating transplantation, Male, Melanoma immunology, Melanoma secondary, Melanoma surgery, Middle Aged, Remission Induction, Gastrointestinal Neoplasms therapy, Immunotherapy, Adoptive, Lung Neoplasms therapy, Lymphocytes, Tumor-Infiltrating metabolism, Melanoma therapy
- Abstract
Adoptive cell transfer of tumor-infiltrating lymphocytes (TILs) can mediate objective tumor regression in 49% to 72% of patients with many long-term durable responses. To undergo treatment a patient must have (1) a resectable tumor from which (2) TIL can be generated that (3) exhibit tumor-specific reactivity. From July 2002 to July 2007, 787 tumors from 402 patients were processed for possible use in the generation of TIL, leading to the eventual treatment of 107 patients (27%). Viable TILs were generated in 376 patients (94%), and active, specific TILs were identified in 269 patients (67%). Patient demographics and tumor characteristics were analyzed for possible prognostic factors for growth and activity. Gastrointestinal-derived TIL grew less frequently, whereas lymph node and lung-derived TIL exhibited specific activity more often. TIL that grew and exhibited specific reactivity were from tumors that were larger in diameter and digests that had a higher percentage of lymphocytes. Despite these considerations, active, specific TIL could be generated from almost any site of metastasis. As more centers begin exploring the use of adoptive transfer with TIL, this compendium may provide a framework for therapeutic decision making and future investigation.
- Published
- 2010
- Full Text
- View/download PDF
43. Tumor-specific CD8+ T cells expressing interleukin-12 eradicate established cancers in lymphodepleted hosts.
- Author
-
Kerkar SP, Muranski P, Kaiser A, Boni A, Sanchez-Perez L, Yu Z, Palmer DC, Reger RN, Borman ZA, Zhang L, Morgan RA, Gattinoni L, Rosenberg SA, Trinchieri G, and Restifo NP
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Female, Flow Cytometry, Interleukin-12 biosynthesis, Interleukin-12 genetics, Lymphocytes, Tumor-Infiltrating metabolism, Melanoma, Experimental immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Protein Engineering methods, Retroviridae genetics, Transduction, Genetic, CD8-Positive T-Lymphocytes immunology, Immunotherapy, Adoptive methods, Interleukin-12 immunology, Lymphocytes, Tumor-Infiltrating immunology, Melanoma, Experimental therapy
- Abstract
T-cell-based immunotherapies can be effective in the treatment of large vascularized tumors, but they rely on adoptive transfer of substantial numbers ( approximately 20 million) of tumor-specific T cells administered together with vaccination and high-dose interleukin (IL)-2. In this study, we report that approximately 10,000 T cells gene-engineered to express a single-chain IL-12 molecule can be therapeutically effective against established tumors in the absence of exogenous IL-2 and vaccine. Although IL-12-engineered cells did not perist long-term in hosts, they exhibited enhanced functionality and were detected in higher numbers intratumorally along with increased numbers of endogenous natural killer and CD8(+) T cells just before regression. Importantly, transferred T cells isolated from tumors stably overproduced supraphysiologic amounts of IL-12, and the therapeutic effect of IL-12 produced within the tumor microenvironment could not be mimicked with high doses of exogenously provided IL-12. Furthermore, antitumor effects could be recapitulated by engineering wild-type open-repertoire splenocytes to express both the single-chain IL-12 and a recombinant tumor-specific T-cell receptor (TCR), but only when individual cells expressed both the TCR and IL-12, indicating that arrested migration of T cells at the tumor site was required for their activities. Successful tumor eradication was dependent on a lymphodepleting preconditioning regimen that reduced the number of intratumoral CD4(+) Foxp3(+) T regulatory cells. Our findings reveal an approach to genetically modify T cells to reduce the cell number needed, eliminate the need for vaccines or systemic IL-2, and improve immunotherapy efficacy based on adoptive transfer of gene-engineered T cells.
- Published
- 2010
- Full Text
- View/download PDF
44. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer.
- Author
-
Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, and Rosenberg SA
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Bevacizumab, Combined Modality Therapy, Epitopes, T-Lymphocyte immunology, Melanoma, Experimental immunology, Membrane Glycoproteins immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Skin Neoplasms immunology, T-Lymphocytes drug effects, T-Lymphocytes immunology, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor A immunology, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, Vascular Endothelial Growth Factor Receptor-2 immunology, Whole-Body Irradiation, gp100 Melanoma Antigen, Angiogenesis Inhibitors pharmacology, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Melanoma, Experimental therapy, Skin Neoplasms therapy
- Abstract
Adoptive cell transfer (ACT)-based immunotherapies can mediate objective cancer regression in animal models and in up to 70% of patients with metastatic melanoma; however, it remains unclear whether the tumor vasculature impedes the egress of tumor-specific T cells, thus hindering this immunotherapy. Disruption of the proangiogenic interaction of vascular endothelial growth factor (VEGF) with its receptor (VEGFR-2) has been reported to "normalize" tumor vasculature, enhancing the efficacy of chemotherapeutic agents by increasing their delivery to the tumor intersitium. We thus sought to determine whether disrupting VEGF/VEGFR-2 signaling could enhance the effectiveness of ACT in a murine cancer model. The administration of an antibody against mouse VEGF synergized with ACT to enhance inhibition of established, vascularized, B16 melanoma (P = 0.009) and improve survival (P = 0.003). Additive effects of an antibody against VEGFR-2 in conjunction with ACT were seen in this model (P = 0.013). Anti-VEGF, but not anti-VEGFR-2, antibody significantly increased infiltration of transferred cells into the tumor. Thus, normalization of tumor vasculature through disruption of the VEGF/VEGFR-2 axis can increase extravasation of adoptively transferred T cells into the tumor and improve ACT-based immunotherapy. These studies provide a rationale for the exploration of combining antiangiogenic agents with ACT for the treatment of patients with cancer.
- Published
- 2010
- Full Text
- View/download PDF
45. Enrichment of CD8+ cells from melanoma tumor-infiltrating lymphocyte cultures reveals tumor reactivity for use in adoptive cell therapy.
- Author
-
Prieto PA, Durflinger KH, Wunderlich JR, Rosenberg SA, and Dudley ME
- Subjects
- Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes pathology, CD8-Positive T-Lymphocytes transplantation, Cell Line, Tumor, Cell Proliferation, Flow Cytometry, HLA-A2 Antigen metabolism, Humans, Immunomagnetic Separation, Interferon-gamma metabolism, Isoantigens immunology, Lymphocyte Activation, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Lymphocytes, Tumor-Infiltrating transplantation, Melanoma, CD8-Positive T-Lymphocytes metabolism, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating metabolism
- Abstract
Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TIL) for metastatic melanoma has shown objective response rates as high as 72%. The successful application of this therapy requires the selection of unique tumor-reactive lymphocyte cultures for each patient. This is a technically and logistically difficult undertaking, and patients who do not have tumor-reactive TIL are not considered eligible for treatment. To simplify the methods of TIL generation and extend TIL-based immunotherapy to additional patients, methods were developed to use unselected, minimally cultured ("young") TIL. Young TIL cultures contain a variable number of CD8(+), CD4(+), and CD3(-)CD56(+) natural killer cells. In this study we retrospectively investigated a role for these subsets in the clinical outcome of patients treated with TIL derived from selected microcultures. This analysis demonstrated a suggestive but nonsignificant association between the number of CD8(+) cells administered and tumor regression. We therefore investigated the feasibility of selecting CD8(+) cells from young TIL cultures for ACT therapy. The available methods for clinical scale CD8(+) enrichment proved inadequate for TIL, so an optimized CD8(+) enrichment method was developed and is reported here. We observed that CD8 (+)enrichment of some TIL cultures revealed in vitro tumor recognition that was not evident in bulk culture, and an improved in vitro recognition of tumor in other TIL cultures. In addition, the enriched CD8(+) young TIL expanded more reliably and predictably in rapid expansions than the bulk TIL. Thus, optimized CD8(+) selection combines the benefits of antigen-selected TIL and young TIL for generating lymphocyte cultures for ACT, and should be evaluated in cell transfer therapy protocols.
- Published
- 2010
- Full Text
- View/download PDF
46. Liver resection for metastatic melanoma with postoperative tumor-infiltrating lymphocyte therapy.
- Author
-
Ripley RT, Davis JL, Klapper JA, Mathur A, Kammula U, Royal RE, Yang JC, Sherry RM, Hughes MS, Libutti SK, White DE, Steinberg SM, Dudley ME, Rosenberg SA, and Avital I
- Subjects
- Adolescent, Adult, Aged, Female, Humans, Liver Neoplasms immunology, Liver Neoplasms secondary, Male, Melanoma immunology, Melanoma secondary, Middle Aged, Neoplasm Staging, Postoperative Period, Prognosis, Prospective Studies, Retrospective Studies, Skin Neoplasms immunology, Skin Neoplasms pathology, Survival Rate, Treatment Outcome, Young Adult, Hepatectomy, Immunotherapy, Adoptive, Liver Neoplasms therapy, Lymphocytes, Tumor-Infiltrating, Melanoma therapy, Skin Neoplasms therapy
- Abstract
Background: Patients with metastatic melanoma to the liver (MML) have a median survival of 4 to 6 months. This study evaluated patients who underwent liver resection with intent to receive postoperative tumor-infiltrating lymphocyte (TIL) therapy., Methods: Retrospective analysis of a prospective database identified patients with MML who underwent liver resection from 1980 to 2008., Results: A total of 539 patients had MML, and 39% (204 of 539) had tumor collected for TIL. A total of 17% (35 of 204) underwent liver resection for TIL. The 3-year overall survival was 53%. Lack of extrahepatic disease (P = .026), negative margin (P = .056), and single hepatic metastasis (P = .04) predicted survival after univariate analysis. Only lack of extrahepatic disease remained a significant predictor of survival after multivariate analysis (P = .043). A total of 31% (11 of 35) underwent complete resection without TIL, and 69% (24 of 35) underwent resection with synchronous intrahepatic and extrahepatic disease with intent to receive TIL. For 9 of 11 patients (2 of 11 excluded for gene therapy), 3-year survival was 80%. A total of 4 (44%) of 9 experienced recurrence, with a median disease-free survival of 1.2 years. For 24 patients (69%) with residual disease, 3-year survival was 51% (2 of 24 excluded for gene therapy). A total of 63% (15 of 24) received postoperative TIL (3-year survival 65%), and 29% (7 of 24) did not. A total of 40% (6 of 15) had disease that partially responded to TIL; the disease of 67% (4 of 6) had not progressed at median follow-up of 55 months (range, 42-197+ months). The seven patients who did not receive TIL had a median survival of 4.6 months., Conclusions: Resection of MML with TIL should be considered because it can result in prolonged survival in a highly selected group of patients.
- Published
- 2010
- Full Text
- View/download PDF
47. Impact of a recombinant fowlpox vaccine on the efficacy of adoptive cell therapy with tumor infiltrating lymphocytes in a patient with metastatic melanoma.
- Author
-
Smith FO, Klapper JA, Wunderlich JR, Rosenberg SA, and Dudley ME
- Subjects
- Blood Transfusion, Autologous, Cell Proliferation, Disease Progression, Epitopes, T-Lymphocyte genetics, Epitopes, T-Lymphocyte metabolism, Facial Neoplasms pathology, Humans, Lymphocytes, Tumor-Infiltrating pathology, Male, Melanoma pathology, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Middle Aged, Neoplasm Metastasis, Viral Vaccines genetics, gp100 Melanoma Antigen, Facial Neoplasms immunology, Facial Neoplasms therapy, Fowlpox virus, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating metabolism, Melanoma immunology, Melanoma therapy
- Abstract
A patient with metastatic melanoma who had progressive disease after prior surgical resections, high dose interleukin-2, and anti-cytotoxic T lymphocyte antigen-4 antibody received sequential treatments with autologous tumor infiltrating lymphocytes that recognized the gp100 melanocyte differentiation antigen. Although no clinical response was seen when cells were administered alone, an objective clinical response to therapy was seen with tumor infiltrating lymphocytes administered together with a highly immunogenic fowlpox vaccine expressing a gp100: 209-217 (210M) epitope. Persistence of the transferred antigen-specific lymphocytes in the peripheral blood was observed only after adoptive cell therapy plus administration of vaccine. Cell proliferation in vitro was further stimulated by additional vaccine and interleukin-2. The patient has an ongoing partial response at 10 months after the last treatment.
- Published
- 2009
- Full Text
- View/download PDF
48. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired.
- Author
-
Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, and Rosenberg SA
- Subjects
- Adolescent, Adult, Aged, Antigens, Neoplasm metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes pathology, Child, Cytotoxicity, Immunologic physiology, Female, Humans, Lymphocyte Activation physiology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, MART-1 Antigen, Male, Melanoma blood, Melanoma metabolism, Melanoma physiopathology, Middle Aged, Neoplasm Proteins metabolism, Programmed Cell Death 1 Receptor, Tumor Escape immunology, Up-Regulation, Young Adult, Antigens, CD metabolism, Apoptosis Regulatory Proteins metabolism, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes physiology, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating physiology, Melanoma immunology
- Abstract
Tumor antigen-specific T cells are found within melanomas, yet tumors continue to grow. Although the tumor microenvironment is thought to influence the suppression of tumor-reactive T cells, the underlying mechanisms for this T-cell dysfunction are not clear. Here, we report that the majority of tumor infiltrating T lymphocytes (TIL), including MART-1/Melan-A melanoma antigen-specific CD8 T cells, predominantly expressed PD-1, in contrast to T cells in normal tissues and peripheral blood T lymphocytes (PBL). PD-1(+) TIL expressed CTLA-4 and Ki-67, markers that were not expressed by PD-1(-) TIL and T cells in the normal tissues and PBL. Moreover, PD-1(+) TIL were primarily HLA-DR(+) and CD127(-), in contrast to PD-1(-) TIL. Effector cytokine production by PD-1(+) TIL was impaired compared with PD-1(-) TIL and PBL. Collectively, the phenotypic and functional characterizations of TIL revealed a significantly higher frequency and level of PD-1 expression on TIL compared with normal tissue T-cell infiltrates and PBL, and PD-1 expression correlated with an exhausted phenotype and impaired effector function. These findings suggest that the tumor microenvironment can lead to up-regulation of PD-1 on tumor-reactive T cells and contribute to impaired antitumor immune responses.
- Published
- 2009
- Full Text
- View/download PDF
49. Single-pass, closed-system rapid expansion of lymphocyte cultures for adoptive cell therapy.
- Author
-
Klapper JA, Thomasian AA, Smith DM, Gorgas GC, Wunderlich JR, Smith FO, Hampson BS, Rosenberg SA, and Dudley ME
- Subjects
- Cell Survival, Cells, Cultured, Culture Media, Humans, Perfusion, Phenotype, Bioreactors, Cell Culture Techniques methods, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating cytology
- Abstract
Adoptive cell therapy (ACT) for metastatic melanoma involves the ex vivo expansion and reinfusion of tumor infiltrating lymphocytes (TIL) obtained from resected specimens. With an overall objective response rate of 56%, this T-cell immunotherapy provides an appealing alternative to other therapies, including conventional therapies with lower response rates. However, there are significant regulatory and logistical concerns associated with the ex vivo activation and large-scale expansion of these cells. The best current practice uses a rapid expansion protocol (REP) consisting of an ex vivo process that occurs in tissue culture flasks (T-flasks) and gas-permeable bags, utilizes OKT3 (anti-CD3 monoclonal antibody), recombinant human interleukin-2, and irradiated peripheral blood mononuclear cells to initiate rapid lymphocyte growth. A major limitation to the widespread delivery of therapy to large numbers of melanoma patients is the open system in which a REP is initiated. To address this problem, we have investigated the initiation, expansion and harvest at clinical scale of TIL in a closed-system continuous perfusion bioreactor. Each cell product met all safety criteria for patient treatment and by head-to-head comparison had a similar potency and phenotype as cells grown in control T-flasks and gas-permeable bags. However, the currently available bioreactor cassettes were limited in the total cell numbers that could be generated. This bioreactor may simplify the process of the rapid expansion of TIL under stringent regulatory conditions thereby enabling other institutions to pursue this form of ACT.
- Published
- 2009
- Full Text
- View/download PDF
50. Lentiviral vector design for optimal T cell receptor gene expression in the transduction of peripheral blood lymphocytes and tumor-infiltrating lymphocytes.
- Author
-
Jones S, Peng PD, Yang S, Hsu C, Cohen CJ, Zhao Y, Abad J, Zheng Z, Rosenberg SA, and Morgan RA
- Subjects
- Antigens immunology, Cell Line, Green Fluorescent Proteins metabolism, Humans, Transgenes, Tumor Suppressor Protein p53 metabolism, Gene Expression Regulation, Genetic Vectors genetics, Lentivirus genetics, Lymphocyte Subsets metabolism, Lymphocytes, Tumor-Infiltrating metabolism, Receptors, Antigen, T-Cell genetics, Transduction, Genetic
- Abstract
Lentiviral vectors containing promoters of distinct origins, that is, strong viral promoters (cytomegalovirus [CMV] and murine stem cell virus [MSCV]), a cellular promoter (phosphoglycerate kinase [PGK]), and two composite promoters (CAG [a composite promoter sequence comprised of the CMV enhancer and portions of the chicken beta-actin promoter and the rabbit beta-globin gene] and SV40/CD43), were used to evaluate green fluorescent protein (GFP) reporter gene expression in human primary peripheral blood lymphocytes (PBLs) and tumor-infiltrating lymphocytes (TILs). In PBLs, vectors containing the MSCV promoter were found to be optimal for expression in both minimally stimulated and highly activated lymphocytes. The stability of gene expression was monitored for up to 7 weeks in culture and the MSCV promoter-containing vector was found to be comparable to the cellular PGK promoter-containing vector. The MSCV promoter-containing lentiviral vector was also the most active in transduced TILs and these cells retained biological activity as measured by antimelanoma antigen reactivity. Using the knowledge gained in comparing individual promoters, a series of two-gene-containing lentiviral vectors was constructed in an attempt to produce the alpha and beta chains of antitumor antigen T cell receptors (TCRs). Dual-promoter or internal ribosome entry site (IRES)-containing vector designs were evaluated and found to be unable to produce both chains of the TCR in amounts that led to significant biological activity. In contrast, if the alpha and beta chains were linked by a 2A ribosomal skip peptide, both proper TCR chain pairing and biologically activity were observed. This paper emphasizes the need to optimize both promoter function and protein synthesis in constructs that require stoichiometric production of multiple protein subunits.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.