84 results on '"Klebanoff CA"'
Search Results
2. The resting and ligand-bound states of the membrane-embedded human T-cell receptor-CD3 complex.
- Author
-
Notti RQ, Yi F, Heissel S, Bush MW, Molvi Z, Das P, Molina H, Klebanoff CA, and Walz T
- Abstract
The T-cell receptor (TCR) initiates T-lymphocyte activation, but mechanistic questions remain( 1-4 ). Here, we present cryogenic electron microscopy structures for the unliganded and human leukocyte antigen (HLA)-bound human TCR-CD3 complex in nanodiscs that provide a native-like lipid environment. Distinct from the "open and extended" conformation seen in detergent( 5-8 ), the unliganded TCR-CD3 in nanodiscs adopts two related "closed and compacted" conformations that represent its physiologic resting state in vivo . By contrast, the HLA-bound complex adopts the open and extended conformation, and conformation-locking disulfide mutants show that ectodomain opening is necessary for maximal ligand-dependent T-cell activation. Together, these results reveal allosteric conformational change during TCR activation and highlight the importance of native-like lipid environments for membrane protein structure determination.
- Published
- 2024
- Full Text
- View/download PDF
3. Engineered NKG2C + NK-like T cells exhibit superior antitumor efficacy while mitigating cytokine release syndrome.
- Author
-
Lupo KB, Panjwani MK, Shahid S, Sottile R, Lawry C, Kolk G, Kontopolous T, Daniyan AF, Chandran SS, Klebanoff CA, and Hsu KC
- Abstract
Engineered T and NK cell therapies have widely been used to treat hematologic malignancies and solid tumors, with promising clinical results. Current chimeric antigen receptor (CAR) T cell therapeutics have, however, been associated with treatment-related adverse events such as cytokine release syndrome (CRS) and are prone to immunologic exhaustion. CAR-NK therapeutics, while not associated with CRS, have limited in vivo persistence. We now demonstrate that an NK-like TCRαβ
+ CD8 T cell subset, identified and expanded ex vivo through its expression of the activating receptor NKG2C (NKG2C+ NK-like T cells), can be transduced to express a second-generation CD19 CAR (1928z), resulting in superior tumor clearance, longer persistence and decreased exhaustion compared to conventional 1928z CAR+ CD8 T cells and 1928z CAR+ NK cells. Moreover, CAR-modified NKG2C+ NK-like T cells resulted in significantly reduced CRS compared to conventional CAR+ CD8 T cells. Similarly, NKG2C+ NK-like T cells engineered with a TCR targeting the NY-ESO-1 antigen exhibit robust tumor control and minimal exhaustion compared to TCR-engineered conventional CD8 T cells. These data establish NKG2C+ NK-like T cells as a robust platform for cell engineering, and offer a safer, more durable alternative to conventional CAR-T and CAR-NK therapies.- Published
- 2024
- Full Text
- View/download PDF
4. Dynamic allostery in the peptide/MHC complex enables TCR neoantigen selectivity.
- Author
-
Ma J, Ayres CM, Brambley CA, Chandran SS, Rosales TJ, Corcelli SA, Kovrigin EL, Klebanoff CA, and Baker BM
- Abstract
The inherent cross-reactivity of the T cell receptor (TCR) is balanced by high specificity, which often manifests in confounding ways not easily interpretable from static structures. We show here that TCR discrimination between an HLA-A*03:01 (HLA-A3)-restricted public neoantigen derived from mutant PIK3CA and its wild-type (WT) counterpart emerges from motions within the HLA binding groove that vary with the identity of the peptide's first primary anchor. The motions form a dynamic gate that in the complex with the WT peptide impedes a large conformational change required for TCR binding. The more rigid neoantigen is insusceptible to this limiting dynamic, and with the gate open, is able to transit its central tryptophan residue underneath the peptide backbone to the contralateral side of the HLA-A3 peptide binding groove, facilitating TCR binding. Our findings reveal a novel mechanism driving TCR specificity for a cancer neoantigen that is rooted in the dynamic and allosteric nature of peptide/MHC-I complexes, with implications for resolving long-standing and often confounding questions about the determinants of T cell specificity., Competing Interests: Additional Declarations: Yes there is potential Competing Interest. C.A.K. and S.S.C. are inventors on patents related to the T cell receptor (TCR) sequences featured in this manuscript and are recipients of licensing revenue from Intima Bioscience shared according to Memorial Sloan Kettering Cancer Center (MSKCC) institutional policies. C.A.K. has consulted for or is on the scientific advisory boards for Achilles Therapeutics, Affini-T Therapeutics, Aleta BioTherapeutics, Bellicum Pharmaceuticals, Bristol Myers Squibb, Catamaran Bio, Cell Design Labs, Decheng Capital, G1 Therapeutics, Klus Pharma, Obsidian Therapeutics, PACT Pharma, Roche/Genentech, Royalty Pharma, and T-knife, and is a scientific co-founder and equity holder in Affini-T Therapeutics. S.S.C. is a scientific advisor and equity holder in Affini-T Therapeutics. B.M.B. is an inventor on patents relating to differences between mutant and self in identifying immunogenic neoantigens, and is on the scientitic advisory board or has consulted for Merck & Co., Pfizer, T-cure Bioscience, Eureka Therapeutics, and EnaraBio. Conflicts of interest statement C.A.K. and S.S.C. are inventors on patents related to the T cell receptor (TCR) sequences featured in this manuscript and are recipients of licensing revenue from Intima Bioscience shared according to Memorial Sloan Kettering Cancer Center (MSKCC) institutional policies. C.A.K. has consulted for or is on the scientific advisory boards for Achilles Therapeutics, Affini-T Therapeutics, Aleta BioTherapeutics, Bellicum Pharmaceuticals, Bristol Myers Squibb, Catamaran Bio, Cell Design Labs, Decheng Capital, G1 Therapeutics, Klus Pharma, Obsidian Therapeutics, PACT Pharma, Roche/Genentech, Royalty Pharma, and T-knife, and is a scientific co-founder and equity holder in Affini-T Therapeutics. S.S.C. is a scientific advisor and equity holder in Affini-T Therapeutics. B.M.B. is an inventor on patents relating to differences between mutant and self in identifying immunogenic neoantigens, and is on the scientific advisory board or has consulted for Merck & Co., Pfizer, T-cure Bioscience, Eureka Therapeutics, and EnaraBio.
- Published
- 2024
- Full Text
- View/download PDF
5. CAR-engineered lymphocyte persistence is governed by a FAS ligand/FAS auto-regulatory circuit.
- Author
-
Yi F, Cohen T, Zimmerman N, Dündar F, Zumbo P, Eltilib R, Brophy EJ, Arkin H, Feucht J, Gormally MV, Hackett CS, Kropp KN, Etxeberria I, Chandran SS, Park JH, Hsu KC, Sadelain M, Betel D, and Klebanoff CA
- Abstract
Chimeric antigen receptor (CAR)-engineered T and NK cells can cause durable remission of B-cell malignancies; however, limited persistence restrains the full potential of these therapies in many patients. The FAS ligand (FAS-L)/FAS pathway governs naturally-occurring lymphocyte homeostasis, yet knowledge of which cells express FAS-L in patients and whether these sources compromise CAR persistence remains incomplete. Here, we constructed a single-cell atlas of diverse cancer types to identify cellular subsets expressing FASLG , the gene encoding FAS-L. We discovered that FASLG is limited primarily to endogenous T cells, NK cells, and CAR-T cells while tumor and stromal cells express minimal FASLG . To establish whether CAR-T/NK cell survival is regulated through FAS-L, we performed competitive fitness assays using lymphocytes modified with or without a FAS dominant negative receptor (ΔFAS). Following adoptive transfer, ΔFAS-expressing CAR-T and CAR-NK cells became enriched across multiple tissues, a phenomenon that mechanistically was reverted through FASLG knockout. By contrast, FASLG was dispensable for CAR-mediated tumor killing. In multiple models, ΔFAS co-expression by CAR-T and CAR-NK enhanced antitumor efficacy compared with CAR cells alone. Together, these findings reveal that CAR-engineered lymphocyte persistence is governed by a FAS-L/FAS auto-regulatory circuit.
- Published
- 2024
- Full Text
- View/download PDF
6. Nivolumab plus ipilimumab in advanced salivary gland cancer: a phase 2 trial.
- Author
-
Vos JL, Burman B, Jain S, Fitzgerald CWR, Sherman EJ, Dunn LA, Fetten JV, Michel LS, Kriplani A, Ng KK, Eng J, Tchekmedyian V, Haque S, Katabi N, Kuo F, Han CY, Nadeem Z, Yang W, Makarov V, Srivastava RM, Ostrovnaya I, Prasad M, Zuur CL, Riaz N, Pfister DG, Klebanoff CA, Chan TA, Ho AL, and Morris LGT
- Subjects
- Humans, Nivolumab adverse effects, Ipilimumab therapeutic use, Receptors, Antigen, T-Cell, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Salivary Gland Neoplasms drug therapy, Salivary Gland Neoplasms genetics, Salivary Gland Neoplasms chemically induced
- Abstract
Salivary gland cancers (SGCs) are rare, aggressive cancers without effective treatments when metastasized. We conducted a phase 2 trial evaluating nivolumab (nivo, anti-PD-1) and ipilimumab (ipi, anti-CTLA-4) in 64 patients with metastatic SGC enrolled in two histology-based cohorts (32 patients each): adenoid cystic carcinoma (ACC; cohort 1) and other SGCs (cohort 2). The primary efficacy endpoint (≥4 objective responses) was met in cohort 2 (5/32, 16%) but not in cohort 1 (2/32, 6%). Treatment safety/tolerability and progression-free survival (PFS) were secondary endpoints. Treatment-related adverse events grade ≥3 occurred in 24 of 64 (38%) patients across both cohorts, and median PFS was 4.4 months (95% confidence interval (CI): 2.4, 8.3) and 2.2 months (95% CI: 1.8, 5.3) for cohorts 1 and 2, respectively. We present whole-exome, RNA and T cell receptor (TCR) sequencing data from pre-treatment and on-treatment tumors and immune cell flow cytometry and TCR sequencing from peripheral blood at serial timepoints. Responding tumors universally demonstrated clonal expansion of pre-existing T cells and mutational contraction. Responding ACCs harbored neoantigens, including fusion-derived neoepitopes, that induced T cell responses ex vivo. This study shows that nivo+ipi has limited efficacy in ACC, albeit with infrequent, exceptional responses, and that it could be promising for non-ACC SGCs, particularly salivary duct carcinomas. ClinicalTrials.gov identifier: NCT03172624 ., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
7. T cell receptor therapeutics: immunological targeting of the intracellular cancer proteome.
- Author
-
Klebanoff CA, Chandran SS, Baker BM, Quezada SA, and Ribas A
- Subjects
- Humans, CD8-Positive T-Lymphocytes metabolism, Antigens, Neoplasm, Receptors, Antigen, T-Cell, Proteome metabolism, Neoplasms drug therapy
- Abstract
The T cell receptor (TCR) complex is a naturally occurring antigen sensor that detects, amplifies and coordinates cellular immune responses to epitopes derived from cell surface and intracellular proteins. Thus, TCRs enable the targeting of proteins selectively expressed by cancer cells, including neoantigens, cancer germline antigens and viral oncoproteins. As such, TCRs have provided the basis for an emerging class of oncology therapeutics. Herein, we review the current cancer treatment landscape using TCRs and TCR-like molecules. This includes adoptive cell transfer of T cells expressing endogenous or engineered TCRs, TCR bispecific engagers and antibodies specific for human leukocyte antigen (HLA)-bound peptides (TCR mimics). We discuss the unique complexities associated with the clinical development of these therapeutics, such as HLA restriction, TCR retrieval, potency assessment and the potential for cross-reactivity. In addition, we highlight emerging clinical data that establish the antitumour potential of TCR-based therapies, including tumour-infiltrating lymphocytes, for the treatment of diverse human malignancies. Finally, we explore the future of TCR therapeutics, including emerging genome editing methods to safely enhance potency and strategies to streamline patient identification., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
8. Tumor-wide RNA splicing aberrations generate immunogenic public neoantigens.
- Author
-
Kwok DW, Stevers NO, Nejo T, Chen LH, Etxeberria I, Jung J, Okada K, Cove MC, Lakshmanachetty S, Gallus M, Barpanda A, Hong C, Chan GKL, Wu SH, Ramos E, Yamamichi A, Liu J, Watchmaker P, Ogino H, Saijo A, Du A, Grishanina N, Woo J, Diaz A, Chang SM, Phillips JJ, Wiita AP, Klebanoff CA, Costello JF, and Okada H
- Abstract
T-cell-mediated immunotherapies are limited by the extent to which cancer-specific antigens are homogenously expressed throughout a tumor. We reasoned that recurrent splicing aberrations in cancer represent a potential source of tumor-wide and public neoantigens, and to test this possibility, we developed a novel pipeline for identifying neojunctions expressed uniformly within a tumor across diverse cancer types. Our analyses revealed multiple neojunctions that recur across patients and either exhibited intratumor heterogeneity or, in some cases, were tumor-wide. We identified CD8+ T-cell clones specific for neoantigens derived from tumor-wide and conserved neojunctions in GNAS and RPL22 , respectively. TCR-engineered CD8
+ T-cells targeting these mutations conferred neoantigen-specific tumor cell eradication. Furthermore, we revealed that cancer-specific dysregulation in splicing factor expression leads to recurrent neojunction expression. Together, these data reveal that a subset of neojunctions are both intratumorally conserved and public, providing the molecular basis for novel T-cell-based immunotherapies that address intratumoral heterogeneity.- Published
- 2023
- Full Text
- View/download PDF
9. Perspectives in Immunotherapy: meeting report from Immunotherapy Bridge (Naples, November 30th-December 1st, 2022).
- Author
-
Ascierto PA, Avallone A, Bifulco C, Bracarda S, Brody JD, Emens LA, Ferris RL, Formenti SC, Hamid O, Johnson DB, Kirchhoff T, Klebanoff CA, Lesinski GB, Monette A, Neyns B, Odunsi K, Paulos CM, Powell DJ Jr, Rezvani K, Segal BH, Singh N, Sullivan RJ, Fox BA, and Puzanov I
- Subjects
- Humans, Immunotherapy, Immunotherapy, Adoptive, Italy, Tumor Microenvironment, Melanoma pathology
- Abstract
The discovery and development of novel treatments that harness the patient's immune system and prevent immune escape has dramatically improved outcomes for patients across cancer types. However, not all patients respond to immunotherapy, acquired resistance remains a challenge, and responses are poor in certain tumors which are considered to be immunologically cold. This has led to the need for new immunotherapy-based approaches, including adoptive cell transfer (ACT), therapeutic vaccines, and novel immune checkpoint inhibitors. These new approaches are focused on patients with an inadequate response to current treatments, with emerging evidence of improved responses in various cancers with new immunotherapy agents, often in combinations with existing agents. The use of cell therapies, drivers of immune response, and trends in immunotherapy were the focus of the Immunotherapy Bridge (November 30th-December 1st, 2022), organized by the Fondazione Melanoma Onlus, Naples, Italy, in collaboration with the Society for Immunotherapy of Cancer., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
10. The ectonucleotidase CD39 identifies tumor-reactive CD8 + T cells predictive of immune checkpoint blockade efficacy in human lung cancer.
- Author
-
Chow A, Uddin FZ, Liu M, Dobrin A, Nabet BY, Mangarin L, Lavin Y, Rizvi H, Tischfield SE, Quintanal-Villalonga A, Chan JM, Shah N, Allaj V, Manoj P, Mattar M, Meneses M, Landau R, Ward M, Kulick A, Kwong C, Wierzbicki M, Yavner J, Egger J, Chavan SS, Farillas A, Holland A, Sridhar H, Ciampricotti M, Hirschhorn D, Guan X, Richards AL, Heller G, Mansilla-Soto J, Sadelain M, Klebanoff CA, Hellmann MD, Sen T, de Stanchina E, Wolchok JD, Merghoub T, and Rudin CM
- Subjects
- Humans, Immune Checkpoint Inhibitors therapeutic use, CD8-Positive T-Lymphocytes, Immunotherapy, Lung Neoplasms genetics, Carcinoma, Non-Small-Cell Lung genetics
- Abstract
Improved identification of anti-tumor T cells is needed to advance cancer immunotherapies. CD39 expression is a promising surrogate of tumor-reactive CD8
+ T cells. Here, we comprehensively profiled CD39 expression in human lung cancer. CD39 expression enriched for CD8+ T cells with features of exhaustion, tumor reactivity, and clonal expansion. Flow cytometry of 440 lung cancer biospecimens revealed weak association between CD39+ CD8+ T cells and tumoral features, such as programmed death-ligand 1 (PD-L1), tumor mutation burden, and driver mutations. Immune checkpoint blockade (ICB), but not cytotoxic chemotherapy, increased intratumoral CD39+ CD8+ T cells. Higher baseline frequency of CD39+ CD8+ T cells conferred improved clinical outcomes from ICB therapy. Furthermore, a gene signature of CD39+ CD8+ T cells predicted benefit from ICB, but not chemotherapy, in a phase III clinical trial of non-small cell lung cancer. These findings highlight CD39 as a proxy of tumor-reactive CD8+ T cells in human lung cancer., Competing Interests: Declaration of interests C.A.K. received research funding support from Kite/Gilead and Intima Bioscience; is on the Scientific and/or Clinical Advisory Boards of Achilles Therapeutics, Aleta BioTherapeutics, Bellicum Pharmaceuticals, Catamaran Bio, Obsidian Therapeutics, and T-knife; and has performed consulting services for Bristol Myers Squibb, PACT Pharma, and Roche/Genentech. C.A.K. is a co-inventor on patent applications related to TCRs targeting public neoantigens unrelated to the current work. M.D.H. received a research grant from BMS; personal fees from Achilles, Arcus, AstraZeneca, Blueprint, BMS, Genentech/Roche, Genzyme, Immunai, Instil Bio, Janssen, Merck, Mirati, Natera, Nektar, Pact Pharma, Regeneron, Shattuck Labs, and Syndax; and equity options from Arcus, Factorial, Immunai, and Shattuck Labs. A patent filed by MSKCC related to the use of tumor mutational burden to predict response to immunotherapy (PCT/US2015/062208) is pending and licensed by PGDx. J.D.W. is a consultant for Amgen, Apricity, Ascentage Pharma, Astellas, AstraZeneca, Bicara Therapeutics, Boehringer Ingelheim, Bristol Myers Squibb, CellCarta, Chugai, Daiichi Sankyo, Dragonfly, Georgiamune, Idera, Imvaq, Larkspur, Maverick Therapeutics, Merck, Psioxus, Recepta, Tizona, Trishula, Sellas, Surface Oncology, and Werewolf Therapeutics. J.D.W. receives grant/research support from Bristol Myers Squibb and Sephora. J.D.W. has equity in Apricity, Arsenal IO, Ascentage, Beigene, Imvaq, Linneaus, Georgiamune, Maverick, Tizona Pharmaceuticals, and Trieza. J.D.W. is a co-inventor on the following patent application: xenogeneic (canine) DNA vaccines, myeloid-derived suppressor cell (MDSC) assay, anti-PD1 antibody, anti-CTLA4 antibodies, anti-GITR antibodies and methods of use thereof, Newcastle disease viruses for cancer therapy, and prediction of responsiveness to treatment with immunomodulatory therapeutics and method of monitoring abscopal effects during such treatment. J.D.W. and T.M. are co-inventors on patent applications related to CD40 and in situ vaccination (PCT/US2016/045970). T.M. is a consultant for Immunos Therapeutics and Pfizer. T.M. is a cofounder of and equity holder in IMVAQ Therapeutics. T.M. receives research funding from Bristol Myers Squibb, Surface Oncology, Kyn Therapeutics, Infinity Pharmaceuticals, Peregrine Pharmaceuticals, Adaptive Biotechnologies, Leap Therapeutics, and Aprea Therapeutics. T.M. is an inventor on patent applications related to work on oncolytic viral therapy, alpha virus-based vaccine, neoantigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4. C.M.R. has consulted regarding oncology drug development with AbbVie, Amgen, Ascentage, AstraZeneca, BMS, Celgene, Daiichi Sankyo, Genentech/Roche, Ipsen, Loxo, and PharmaMar and is on the scientific advisory boards of Elucida, Bridge, and Harpoon. B.Y.N. and X.G. are employees and stockholders of Genentech/Roche., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
11. Less, but better: A simplified design for T cell redirection and conditional payload delivery.
- Author
-
Kropp KN and Klebanoff CA
- Subjects
- Humans, Immunotherapy, T-Lymphocytes, Neoplasms therapy
- Abstract
Combining immune receptor engineering with conditional expression of accessory molecules holds great promise for advancing the field of cell-based immunotherapies. In this issue of Cancer Cell, Smole et al. introduce a modular single vector system to simultaneously redirect T cell specificity toward cancer antigens while achieving activation-gated delivery of customizable payloads., Competing Interests: Declaration of interests C.A.K. is on the scientific and/or clinical advisory boards for Achilles Therapeutics, Aleta BioTherapeutics, Bellicum Pharmaceuticals, Catamaran Bio, Obsidian Therapeutics, and T-knife; has consulted for Bristol Myers Squibb, Decheng Capital, PACT Pharma, and Roche/Genentech; and has patents broadly related to cell and gene therapy outside the scope of this work., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
12. Clinical implications of T cell exhaustion for cancer immunotherapy.
- Author
-
Chow A, Perica K, Klebanoff CA, and Wolchok JD
- Subjects
- Humans, Immune Checkpoint Inhibitors, Immunotherapy, Immunotherapy, Adoptive, Tumor Microenvironment, T-Lymphocytes, Neoplasms
- Abstract
Immunotherapy has been a remarkable clinical advancement in the treatment of cancer. T cells are pivotal to the efficacy of current cancer immunotherapies, including immune-checkpoint inhibitors and adoptive cell therapies. However, cancer is associated with T cell exhaustion, a hypofunctional state characterized by progressive loss of T cell effector functions and self-renewal capacity. The 'un-exhausting' of T cells in the tumour microenvironment is commonly regarded as a key mechanism of action for immune-checkpoint inhibitors, and T cell exhaustion is considered a pathway of resistance for cellular immunotherapies. Several elegant studies have provided important insights into the transcriptional and epigenetic programmes that govern T cell exhaustion. In this Review, we highlight recent discoveries related to the immunobiology of T cell exhaustion that offer a more nuanced perspective beyond this hypofunctional state being entirely undesirable. We review evidence that T cell exhaustion might be as much a reflection as it is the cause of poor tumour control. Furthermore, we hypothesize that, in certain contexts of chronic antigen stimulation, interruption of the exhaustion programme might impair T cell persistence. Therefore, the prioritization of interventions that mitigate the development of T cell exhaustion, including orthogonal cytoreduction therapies and novel cellular engineering strategies, might ultimately confer superior clinical outcomes and the greatest advances in cancer immunotherapy., (© 2022. Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
13. T-cell Receptor Gene Therapy Clinically Targeting a TP53 Public Neoantigen.
- Author
-
Klebanoff CA
- Subjects
- Antigens, Neoplasm immunology, Genes, T-Cell Receptor, Humans, Mutation, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms genetics, Neoplasms immunology, Neoplasms therapy
- Abstract
T-cell receptors (TCR) are an antigen receptor class that can uniquely respond to epitopes resulting from cytosolic and intranuclear proteins. In this issue, Kim and colleagues report the first successful application of TCR gene therapy targeting a shared, or public, neoantigen resulting from a TP53 hotspot mutation. These results establish clinical proof of concept that an off-the-shelf TCR targeting a recurrent mutation in a molecular driver of oncogenesis can benefit patients with metastatic cancer. See related article by Kim et al., p. 932 (4) ., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
14. Perspectives in Immunotherapy: meeting report from the Immunotherapy Bridge, December 1st-2nd, 2021.
- Author
-
Ascierto PA, Avallone A, Bhardwaj N, Bifulco C, Bracarda S, Brody JD, Buonaguro L, Demaria S, Emens LA, Ferris RL, Galon J, Khleif SN, Klebanoff CA, Laskowski T, Melero I, Paulos CM, Pignata S, Ruella M, Svane IM, Taube JM, Fox BA, Hwu P, and Puzanov I
- Subjects
- Humans, Immunologic Factors, Immunotherapy, Italy, Biomarkers, Tumor metabolism, Melanoma
- Abstract
Over the past decade, immunotherapy has become an increasingly fundamental modality in the treatment of cancer. The positive impact of immune checkpoint inhibition, especially anti-programmed death (PD)-1/PD-ligand (L)1 blockade, in patients with different cancers has focused attention on the potential for other immunotherapeutic approaches. These include inhibitors of additional immune checkpoints, adoptive cell transfer (ACT), and therapeutic vaccines. Patients with advanced cancers who previously had limited treatment options available may now benefit from immunotherapies that can offer durable responses and improved survival outcomes. However, despite this, a significant proportion of patients fail to respond to immunotherapy, especially those with less immunoresponsive cancer types, and there remains a need for new treatment strategies.The virtual Immunotherapy Bridge (December 1st-2nd, 2021), organized by the Fondazione Melanoma Onlus, Naples, Italy in collaboration with the Society for Immunotherapy of Cancer addressed several areas of current research in immunotherapy, including lessons learned from cell therapies, drivers of immune response, and trends in immunotherapy across different cancers, and these are summarised here., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
15. Immunogenicity and therapeutic targeting of a public neoantigen derived from mutated PIK3CA.
- Author
-
Chandran SS, Ma J, Klatt MG, Dündar F, Bandlamudi C, Razavi P, Wen HY, Weigelt B, Zumbo P, Fu SN, Banks LB, Yi F, Vercher E, Etxeberria I, Bestman WD, Da Cruz Paula A, Aricescu IS, Drilon A, Betel D, Scheinberg DA, Baker BM, and Klebanoff CA
- Subjects
- Animals, Class I Phosphatidylinositol 3-Kinases genetics, Humans, Mice, Mutation genetics, Receptors, Antigen, T-Cell, Antigens, Neoplasm genetics, Neoplasms genetics
- Abstract
Public neoantigens (NeoAgs) represent an elite class of shared cancer-specific epitopes derived from recurrently mutated driver genes. Here we describe a high-throughput platform combining single-cell transcriptomic and T cell receptor (TCR) sequencing to establish whether mutant PIK3CA, among the most frequently genomically altered driver oncogenes, generates an immunogenic public NeoAg. Using this strategy, we developed a panel of TCRs that recognize an endogenously processed neopeptide encompassing a common PIK3CA hotspot mutation restricted by the prevalent human leukocyte antigen (HLA)-A*03:01 allele. Mechanistically, immunogenicity to this public NeoAg arises from enhanced neopeptide/HLA complex stability caused by a preferred HLA anchor substitution. Structural studies indicated that the HLA-bound neopeptide presents a comparatively 'featureless' surface dominated by the peptide's backbone. To bind this epitope with high specificity and affinity, we discovered that a lead TCR clinical candidate engages the neopeptide through an extended interface facilitated by an unusually long CDR3β loop. In patients with diverse malignancies, we observed NeoAg clonal conservation and spontaneous immunogenicity to the neoepitope. Finally, adoptive transfer of TCR-engineered T cells led to tumor regression in vivo in mice bearing PIK3CA-mutant tumors but not wild-type PIK3CA tumors. Together, these findings establish the immunogenicity and therapeutic potential of a mutant PIK3CA-derived public NeoAg., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
16. Risk stratification of cardiac metastases using late gadolinium enhancement cardiovascular magnetic resonance: prognostic impact of hypo-enhancement evidenced tumor avascularity.
- Author
-
Chan AT, Dinsfriend W, Kim J, Yum B, Sultana R, Klebanoff CA, Plodkowski A, Perez Johnston R, Ginsberg MS, Liu J, Kim RJ, Steingart R, and Weinsaft JW
- Subjects
- Adult, Aged, Case-Control Studies, Female, Heart Neoplasms mortality, Heart Neoplasms secondary, Humans, Male, Middle Aged, Neoplasm Grading, Neoplasm Staging, New York City, Predictive Value of Tests, Prognosis, Risk Assessment, Risk Factors, Contrast Media, Heart Neoplasms blood supply, Heart Neoplasms diagnostic imaging, Magnetic Resonance Imaging, Cine, Meglumine, Neoplastic Cells, Circulating pathology, Organometallic Compounds
- Abstract
Background: Late gadolinium enhancement (LGE) cardiovascular magnetic resonance (CMR) is widely used to identify cardiac neoplasms, for which diagnosis is predicated on enhancement stemming from lesion vascularity: Impact of contrast-enhancement pattern on clinical outcomes is unknown. The objective of this study was to determine whether cardiac metastasis (C
MET ) enhancement pattern on LGE-CMR impacts prognosis, with focus on heterogeneous lesion enhancement as a marker of tumor avascularity., Methods: Advanced (stage IV) systemic cancer patients with and without CMET matched (1:1) by cancer etiology underwent a standardized CMR protocol. CMET was identified via established LGE-CMR criteria based on lesion enhancement; enhancement pattern was further classified as heterogeneous (enhancing and non-enhancing components) or diffuse and assessed via quantitative (contrast-to-noise ratio (CNR); signal-to-noise ratio (SNR)) analyses. Embolic events and mortality were tested in relation to lesion location and contrast-enhancement pattern., Results: 224 patients were studied, including 112 patients with CMET and unaffected (CMET -) controls matched for systemic cancer etiology/stage. CMET enhancement pattern varied (53% heterogeneous, 47% diffuse). Quantitative analyses were consistent with lesion classification; CNR was higher and SNR lower in heterogeneously enhancing CMET (p < 0.001)-paralleled by larger size based on linear dimensions (p < 0.05). Contrast-enhancement pattern did not vary based on lesion location (p = NS). Embolic events were similar between patients with diffuse and heterogeneous lesions (p = NS) but varied by location: Patients with right-sided lesions had threefold more pulmonary emboli (20% vs. 6%, p = 0.02); those with left-sided lesions had lower rates equivalent to controls (4% vs. 5%, p = 1.00). Mortality was higher among patients with CMET (hazard ratio [HR] = 1.64 [CI 1.17-2.29], p = 0.004) compared to controls, but varied by contrast-enhancement pattern: Diffusely enhancing CMET had equivalent mortality to controls (p = 0.21) whereas prognosis was worse with heterogeneous CMET (p = 0.005) and more strongly predicted by heterogeneous enhancement (HR = 1.97 [CI 1.23-3.15], p = 0.005) than lesion size (HR = 1.11 per 10 cm [CI 0.53-2.33], p = 0.79)., Conclusions: Contrast-enhancement pattern and location of CMET on CMR impacts prognosis. Embolic events vary by CMET location, with likelihood of PE greatest with right-sided lesions. Heterogeneous enhancement-a marker of tumor avascularity on LGE-CMR-is a novel marker of increased mortality risk.- Published
- 2021
- Full Text
- View/download PDF
17. Erasing iatrogenic neoantigens from in vivo CRISPR screens.
- Author
-
Yi F and Klebanoff CA
- Subjects
- Antigens, Humans, Iatrogenic Disease, Immunotherapy, CRISPR-Cas Systems genetics, Genetic Therapy
- Abstract
In vivo genetic screens using CRISPR-Cas9 are a powerful tool to resolve the molecular determinants of response and resistance to cancer immunotherapies; however, vector immunogenicity can introduce artifact. In this issue of Immunity, Dubrot et al. report a strategy to "erase" vector-associated neoantigens, enabling a more physiologic assessment of tumor-immune cell interactions in immunocompetent hosts., Competing Interests: Declaration of interests F.Y. and C.A.K. hold provisional patents related to cell and gene therapy unrelated to the current work. C.A.K. is a scientific advisor to the following immunotherapy-focused companies: Achilles Therapeutics, Aleta Biotherapeutics, Bellicum Pharmaceuticals, Catamaran Biotherapeutics, Obsidian Therapeutics, Roche/Genentech, Pact Biopharma, T-Knife., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. Immunomodulatory Activity of a Colony-stimulating Factor-1 Receptor Inhibitor in Patients with Advanced Refractory Breast or Prostate Cancer: A Phase I Study.
- Author
-
Autio KA, Klebanoff CA, Schaer D, Kauh JSW, Slovin SF, Adamow M, Blinder VS, Brahmachary M, Carlsen M, Comen E, Danila DC, Doman TN, Durack JC, Fox JJ, Gluskin JS, Hoffman DM, Kang S, Kang P, Landa J, McAndrew PF, Modi S, Morris MJ, Novosiadly R, Rathkopf DE, Sanford R, Chapman SC, Tate CM, Yu D, Wong P, and McArthur HL
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal adverse effects, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Proliferation drug effects, Drug-Related Side Effects and Adverse Reactions classification, Drug-Related Side Effects and Adverse Reactions epidemiology, Drug-Related Side Effects and Adverse Reactions pathology, Female, Humans, Immunologic Factors administration & dosage, Immunologic Factors adverse effects, Lipopolysaccharide Receptors genetics, Male, Middle Aged, Neoplasm Metastasis, Neoplasm Staging, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Receptor, Macrophage Colony-Stimulating Factor antagonists & inhibitors, Receptors, IgG genetics, Antibodies, Monoclonal administration & dosage, Breast Neoplasms drug therapy, Prostatic Neoplasms, Castration-Resistant drug therapy, Receptor, Macrophage Colony-Stimulating Factor genetics
- Abstract
Purpose: Tumor-associated macrophages correlate with increased invasiveness, growth, and immunosuppression. Activation of the colony-stimulating factor-1 receptor (CSF-1R) results in proliferation, differentiation, and migration of monocytes/macrophages. This phase I study evaluated the immunologic and clinical activity, and safety profile of CSF-1R inhibition with the mAb LY3022855., Patients and Methods: Patients with advanced refractory metastatic breast cancer (MBC) or metastatic castration-resistant prostate cancer (mCRPC) were treated with LY3022855 intravenously in 6-week cycles in cohorts: (A) 1.25 mg/kg every 2 weeks (Q2W); (B) 1.0 mg/kg on weeks 1, 2, 4, and 5; (C) 100 mg once weekly; (D)100 mg Q2W. mCRPC patients were enrolled in cohorts A and B; patients with MBC were enrolled in all cohorts. Efficacy was assessed by RECIST and Prostate Cancer Clinical Trials Working Group 2 criteria., Results: Thirty-four patients (22 MBC; 12 mCRPC) received ≥1 dose of LY3022855. At day 8, circulating CSF-1 levels increased and proinflammatory monocytes CD14
DIM CD16BRIGHT decreased. Best RECIST response was stable disease in five patients with MBC (23%; duration, 82-302 days) and three patients with mCRPC (25%; duration, 50-124 days). Two patients with MBC (cohort A) had durable stable disease >9 months and a third patient with MBC had palpable reduction in a nontarget neck mass. Immune-related gene activation in tumor biopsies posttreatment was observed. Common any grade treatment-related adverse events were fatigue, decreased appetite, nausea, asymptomatic increased lipase, and creatine phosphokinase., Conclusions: LY3022855 was well tolerated and showed evidence of immune modulation. Clinically meaningful stable disease >9 months was observed in two patients with MBC., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
19. Optimization of T-cell Receptor-Modified T Cells for Cancer Therapy.
- Author
-
Drakes DJ, Rafiq S, Purdon TJ, Lopez AV, Chandran SS, Klebanoff CA, and Brentjens RJ
- Subjects
- Animals, Antigens, Neoplasm immunology, Apoptosis, CD8-Positive T-Lymphocytes metabolism, Cell Proliferation, Humans, Immunotherapy, Adoptive methods, Melanoma immunology, Melanoma metabolism, Melanoma pathology, Melanoma, Experimental immunology, Melanoma, Experimental metabolism, Melanoma, Experimental pathology, Melanoma, Experimental therapy, Mice, Mice, Knockout, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, CD8-Positive T-Lymphocytes immunology, Immunotherapy, Adoptive standards, Interleukin-12 metabolism, Interleukin-18 metabolism, Lymphocyte Activation immunology, Melanoma therapy, Receptors, Antigen, T-Cell immunology
- Abstract
T-cell receptor (TCR)-modified T-cell gene therapy can target a variety of extracellular and intracellular tumor-associated antigens, yet has had little clinical success. A potential explanation for limited antitumor efficacy is a lack of T-cell activation in vivo We postulated that expression of proinflammatory cytokines in TCR-modified T cells would activate T cells and enhance antitumor efficacy. We demonstrate that expression of interleukin 18 (IL18) in tumor-directed TCR-modified T cells provides a superior proinflammatory signal than expression of interleukin 12 (IL12). Tumor-targeted T cells secreting IL18 promote persistent and functional effector T cells and a proinflammatory tumor microenvironment. Together, these effects augmented overall survival of mice in the pmel-1 syngeneic tumor model. When combined with sublethal irradiation, IL18-secreting pmel-1 T cells were able to eradicate tumors, whereas IL12-secreting pmel-1 T cells caused toxicity in mice through excessive cytokine secretion. In another xenograft tumor model, IL18 secretion enhanced the persistence and antitumor efficacy of NY-ESO-1-reactive TCR-modified human T cells as well as overall survival of tumor-bearing mice. These results demonstrate a rationale for optimizing the efficacy of TCR-modified T-cell cancer therapy through expression of IL18. See related commentary by Wijewarnasuriya et al., p. 732 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
20. Identification of the Targets of T-cell Receptor Therapeutic Agents and Cells by Use of a High-Throughput Genetic Platform.
- Author
-
Gejman RS, Jones HF, Klatt MG, Chang AY, Oh CY, Chandran SS, Korontsvit T, Zakahleva V, Dao T, Klebanoff CA, and Scheinberg DA
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Histocompatibility Antigens Class I immunology, Histocompatibility Antigens Class I metabolism, Mice, Mice, Inbred C57BL, Neoplasms genetics, Neoplasms therapy, Receptors, Antigen, T-Cell immunology, Cross Reactions immunology, High-Throughput Screening Assays methods, Histocompatibility Antigens Class I genetics, Major Histocompatibility Complex immunology, Neoplasms immunology, Receptors, Antigen, T-Cell genetics, T-Lymphocytes immunology
- Abstract
T-cell receptor (TCR)-based therapeutic cells and agents have emerged as a new class of effective cancer therapies. These therapies work on cells that express intracellular cancer-associated proteins by targeting peptides displayed on MHC receptors. However, cross-reactivities of these agents to off-target cells and tissues have resulted in serious, sometimes fatal, adverse events. We have developed a high-throughput genetic platform (termed "PresentER") that encodes MHC-I peptide minigenes for functional immunologic assays and determines the reactivities of TCR-like therapeutic agents against large libraries of MHC-I ligands. In this article, we demonstrated that PresentER could be used to identify the on-and-off targets of T cells and TCR-mimic (TCRm) antibodies using in vitro coculture assays or binding assays. We found dozens of MHC-I ligands that were cross-reactive with two TCRm antibodies and two native TCRs and that were not easily predictable by other methods., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
21. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis.
- Author
-
Dafni U, Michielin O, Lluesma SM, Tsourti Z, Polydoropoulou V, Karlis D, Besser MJ, Haanen J, Svane IM, Ohashi PS, Kammula US, Orcurto A, Zimmermann S, Trueb L, Klebanoff CA, Lotze MT, Kandalaft LE, and Coukos G
- Subjects
- Combined Modality Therapy, Disease-Free Survival, Dose-Response Relationship, Drug, Humans, Interleukin-2 genetics, Melanoma immunology, Melanoma pathology, Remission Induction, Skin Neoplasms immunology, Skin Neoplasms pathology, Transplantation, Autologous, Melanoma, Cutaneous Malignant, Interleukin-2 therapeutic use, Lymphocytes, Tumor-Infiltrating transplantation, Melanoma therapy, Recombinant Proteins therapeutic use, Skin Neoplasms therapy
- Abstract
Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) has been tested in advanced melanoma patients at various centers. We conducted a systematic review and meta-analysis to assess its efficacy on previously treated advanced metastatic cutaneous melanoma. The PubMed electronic database was searched from inception to 17 December 2018 to identify studies administering TIL-ACT and recombinant interleukin-2 (IL-2) following non-myeloablative chemotherapy in previously treated metastatic melanoma patients. Objective response rate (ORR) was the primary end point. Secondary end points were complete response rate (CRR), overall survival (OS), duration of response (DOR) and toxicity. Pooled estimates were derived from fixed or random effect models, depending on the amount of heterogeneity detected. Analysis was carried out separately for high dose (HD) and low dose (LD) IL-2. Sensitivity analyses were carried out. Among 1211 records screened, 13 studies (published 1988 - 2016) were eligible for meta-analysis. Among 410 heavily pretreated patients (some with brain metastasis), 332 received HD-IL-2 and 78 LD-IL-2. The pooled overall ORR estimate was 41% [95% confidence interval (CI) 35% to 48%], and the overall CRR was 12% (95% CI 7% to 16%). For the HD-IL-2 group, the ORR was 43% (95% CI 36% to 50%), while for the LD-IL-2 it was 35% (95% CI 25% to 45%). Corresponding pooled estimates for CRR were 14% (95% CI 7% to 20%) and 7% (95% CI 1% to 12%). The majority of HD-IL-2 complete responders (27/28) remained in remission during the extent of follow-up after CR (median 40 months). Sensitivity analyses yielded similar results. Higher number of infused cells was associated with a favorable response. The ORR for HD-IL-2 compared favorably with the nivolumab/ipilimumab combination following anti-PD-1 failure. TIL-ACT therapy, especially when combined with HD-IL-2, achieves durable clinical benefit and warrants further investigation. We discuss the current position of TIL-ACT in the therapy of advanced melanoma, particularly in the era of immune checkpoint blockade therapy, and review future opportunities for improvement of this approach., (© The Author(s) 2019. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
22. TOX is a critical regulator of tumour-specific T cell differentiation.
- Author
-
Scott AC, Dündar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba M, Trivedi P, Menocal L, Appleby H, Camara S, Zamarin D, Walther T, Snyder A, Femia MR, Comen EA, Wen HY, Hellmann MD, Anandasabapathy N, Liu Y, Altorki NK, Lauer P, Levy O, Glickman MS, Kaye J, Betel D, Philip M, and Schietinger A
- Subjects
- Animals, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, High Mobility Group Proteins deficiency, High Mobility Group Proteins genetics, Homeodomain Proteins genetics, Humans, Immunologic Memory, Lymphocytes, Tumor-Infiltrating cytology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Mice, Neoplasms pathology, Phenotype, Receptors, Antigen, T-Cell immunology, Transcription, Genetic, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes pathology, Cell Differentiation immunology, High Mobility Group Proteins metabolism, Homeodomain Proteins metabolism, Neoplasms immunology
- Abstract
Tumour-specific CD8 T cell dysfunction is a differentiation state that is distinct from the functional effector or memory T cell states
1-6 . Here we identify the nuclear factor TOX as a crucial regulator of the differentiation of tumour-specific T (TST) cells. We show that TOX is highly expressed in dysfunctional TST cells from tumours and in exhausted T cells during chronic viral infection. Expression of TOX is driven by chronic T cell receptor stimulation and NFAT activation. Ectopic expression of TOX in effector T cells in vitro induced a transcriptional program associated with T cell exhaustion. Conversely, deletion of Tox in TST cells in tumours abrogated the exhaustion program: Tox-deleted TST cells did not upregulate genes for inhibitory receptors (such as Pdcd1, Entpd1, Havcr2, Cd244 and Tigit), the chromatin of which remained largely inaccessible, and retained high expression of transcription factors such as TCF-1. Despite their normal, 'non-exhausted' immunophenotype, Tox-deleted TST cells remained dysfunctional, which suggests that the regulation of expression of inhibitory receptors is uncoupled from the loss of effector function. Notably, although Tox-deleted CD8 T cells differentiated normally to effector and memory states in response to acute infection, Tox-deleted TST cells failed to persist in tumours. We hypothesize that the TOX-induced exhaustion program serves to prevent the overstimulation of T cells and activation-induced cell death in settings of chronic antigen stimulation such as cancer.- Published
- 2019
- Full Text
- View/download PDF
23. T cell receptor-based cancer immunotherapy: Emerging efficacy and pathways of resistance.
- Author
-
Chandran SS and Klebanoff CA
- Subjects
- Animals, Antigens, Neoplasm immunology, Clinical Studies as Topic, Genetic Engineering, Humans, Neoplasms metabolism, Receptors, Antigen, T-Cell genetics, Receptors, Chimeric Antigen genetics, T-Cell Antigen Receptor Specificity, Treatment Outcome, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Neoplasms immunology, Neoplasms therapy, Receptors, Antigen, T-Cell metabolism, Receptors, Chimeric Antigen metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Adoptive cell transfer (ACT) using chimeric antigen receptor (CAR)-modified T cells can induce durable remissions in patients with refractory B-lymphoid cancers. By contrast, results applying CAR-modified T cells to solid malignancies have been comparatively modest. Alternative strategies to redirect T cell specificity and cytolytic function are therefore necessary if ACT is to serve a greater role in human cancer treatments. T cell receptors (TCRs) are antigen recognition structures physiologically expressed by all T cells that have complementary, and in some cases superior, properties to CARs. Unlike CARs, TCRs confer recognition to epitopes derived from proteins residing within any subcellular compartment, including the membrane, cytoplasm and nucleus. This enables TCRs to detect a broad universe of targets, such as neoantigens, cancer germline antigens, and viral oncoproteins. Moreover, because TCRs have evolved to efficiently detect and amplify antigenic signals, these receptors respond to epitope densities many fold smaller than required for CAR-signaling. Herein, we summarize recent clinical data demonstrating that TCR-based immunotherapies can mediate regression of solid malignancies, including immune-checkpoint inhibitor refractory cancers. These trials simultaneously highlight emerging mechanisms of TCR resistance. We conclude by discussing how TCR-based immunotherapies can achieve broader dissemination through innovations in cell manufacturing and non-viral genome integration techniques., (© 2019 The Authors. Immunological Reviews Published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
24. T cell stemness and dysfunction in tumors are triggered by a common mechanism.
- Author
-
Vodnala SK, Eil R, Kishton RJ, Sukumar M, Yamamoto TN, Ha NH, Lee PH, Shin M, Patel SJ, Yu Z, Palmer DC, Kruhlak MJ, Liu X, Locasale JW, Huang J, Roychoudhuri R, Finkel T, Klebanoff CA, and Restifo NP
- Subjects
- Acetyl Coenzyme A metabolism, Acetylation, Animals, Autophagy immunology, Caloric Restriction, Cell Differentiation genetics, Epigenesis, Genetic, Histones metabolism, Humans, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Tumor Microenvironment, CD8-Positive T-Lymphocytes immunology, Immune Tolerance, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms immunology, Potassium metabolism, Stem Cells immunology
- Abstract
A paradox of tumor immunology is that tumor-infiltrating lymphocytes are dysfunctional in situ, yet are capable of stem cell-like behavior including self-renewal, expansion, and multipotency, resulting in the eradication of large metastatic tumors. We find that the overabundance of potassium in the tumor microenvironment underlies this dichotomy, triggering suppression of T cell effector function while preserving stemness. High levels of extracellular potassium constrain T cell effector programs by limiting nutrient uptake, thereby inducing autophagy and reduction of histone acetylation at effector and exhaustion loci, which in turn produces CD8
+ T cells with improved in vivo persistence, multipotency, and tumor clearance. This mechanistic knowledge advances our understanding of T cell dysfunction and may lead to novel approaches that enable the development of enhanced T cell strategies for cancer immunotherapy., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
25. 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
26. T cells genetically engineered to overcome death signaling enhance adoptive cancer immunotherapy.
- Author
-
Yamamoto TN, Lee PH, Vodnala SK, Gurusamy D, Kishton RJ, Yu Z, Eidizadeh A, Eil R, Fioravanti J, Gattinoni L, Kochenderfer JN, Fry TJ, Aksoy BA, Hammerbacher JE, Cruz AC, Siegel RM, Restifo NP, and Klebanoff CA
- Subjects
- Animals, Fas Ligand Protein genetics, Fas Ligand Protein immunology, Fas-Associated Death Domain Protein genetics, Fas-Associated Death Domain Protein immunology, Female, Humans, Male, Mice, Mice, Transgenic, Neoplasms, Experimental genetics, Neoplasms, Experimental immunology, Neoplasms, Experimental pathology, Signal Transduction genetics, Tumor Microenvironment genetics, fas Receptor genetics, fas Receptor immunology, Adoptive Transfer, Genetic Engineering, Neoplasms, Experimental therapy, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen therapeutic use, Signal Transduction immunology, Tumor Microenvironment immunology
- Abstract
Across clinical trials, T cell expansion and persistence following adoptive cell transfer (ACT) have correlated with superior patient outcomes. Herein, we undertook a pan-cancer analysis to identify actionable ligand-receptor pairs capable of compromising T cell durability following ACT. We discovered that FASLG, the gene encoding the apoptosis-inducing ligand FasL, is overexpressed within the majority of human tumor microenvironments (TMEs). Further, we uncovered that Fas, the receptor for FasL, is highly expressed on patient-derived T cells used for clinical ACT. We hypothesized that a cognate Fas-FasL interaction within the TME might limit both T cell persistence and antitumor efficacy. We discovered that genetic engineering of Fas variants impaired in the ability to bind FADD functioned as dominant negative receptors (DNRs), preventing FasL-induced apoptosis in Fas-competent T cells. T cells coengineered with a Fas DNR and either a T cell receptor or chimeric antigen receptor exhibited enhanced persistence following ACT, resulting in superior antitumor efficacy against established solid and hematologic cancers. Despite increased longevity, Fas DNR-engineered T cells did not undergo aberrant expansion or mediate autoimmunity. Thus, T cell-intrinsic disruption of Fas signaling through genetic engineering represents a potentially universal strategy to enhance ACT efficacy across a broad range of human malignancies.
- Published
- 2019
- Full Text
- View/download PDF
27. Tumor mutational load predicts survival after immunotherapy across multiple cancer types.
- Author
-
Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, Barron DA, Zehir A, Jordan EJ, Omuro A, Kaley TJ, Kendall SM, Motzer RJ, Hakimi AA, Voss MH, Russo P, Rosenberg J, Iyer G, Bochner BH, Bajorin DF, Al-Ahmadie HA, Chaft JE, Rudin CM, Riely GJ, Baxi S, Ho AL, Wong RJ, Pfister DG, Wolchok JD, Barker CA, Gutin PH, Brennan CW, Tabar V, Mellinghoff IK, DeAngelis LM, Ariyan CE, Lee N, Tap WD, Gounder MM, D'Angelo SP, Saltz L, Stadler ZK, Scher HI, Baselga J, Razavi P, Klebanoff CA, Yaeger R, Segal NH, Ku GY, DeMatteo RP, Ladanyi M, Rizvi NA, Berger MF, Riaz N, Solit DB, Chan TA, and Morris LGT
- Subjects
- Antineoplastic Agents immunology, High-Throughput Nucleotide Sequencing methods, Humans, Immunotherapy methods, Neoplasms immunology, Tumor Burden genetics, Tumor Burden immunology, Mutation genetics, Neoplasms genetics, Neoplasms therapy
- Abstract
Immune checkpoint inhibitor (ICI) treatments benefit some patients with metastatic cancers, but predictive biomarkers are needed. Findings in selected cancer types suggest that tumor mutational burden (TMB) may predict clinical response to ICI. To examine this association more broadly, we analyzed the clinical and genomic data of 1,662 advanced cancer patients treated with ICI, and 5,371 non-ICI-treated patients, whose tumors underwent targeted next-generation sequencing (MSK-IMPACT). Among all patients, higher somatic TMB (highest 20% in each histology) was associated with better overall survival. For most cancer histologies, an association between higher TMB and improved survival was observed. The TMB cutpoints associated with improved survival varied markedly between cancer types. These data indicate that TMB is associated with improved survival in patients receiving ICI across a wide variety of cancer types, but that there may not be one universal definition of high TMB.
- Published
- 2019
- Full Text
- View/download PDF
28. Association of PI3K Pathway Mutations with Early Positron-Emission Tomography/CT Imaging Response after Radioembolization for Breast Cancer Liver Metastases: Results of a Single-Center Retrospective Pilot Study.
- Author
-
Deipolyi AR, Riedl CC, Bromberg J, Chandarlapaty S, Klebanoff CA, Sofocleous CT, Yarmohammadi H, Brody LA, Boas FE, and Ziv E
- Subjects
- Adult, Breast Neoplasms genetics, Breast Neoplasms pathology, Clinical Decision-Making, DNA Mutational Analysis, Embolization, Therapeutic adverse effects, Female, Gene Expression Profiling, Humans, Liver Neoplasms genetics, Liver Neoplasms secondary, Middle Aged, New York City, Patient Selection, Pilot Projects, Precision Medicine, Predictive Value of Tests, Preliminary Data, Radiopharmaceuticals adverse effects, Retrospective Studies, Risk Factors, Signal Transduction genetics, Time Factors, Treatment Outcome, Breast Neoplasms diagnostic imaging, Embolization, Therapeutic methods, Liver Neoplasms diagnostic imaging, Liver Neoplasms surgery, Mutation, Phosphatidylinositol 3-Kinases genetics, Positron Emission Tomography Computed Tomography, Radiopharmaceuticals administration & dosage
- Abstract
Purpose: To describe imaging response and survival after radioembolization for metastatic breast cancer and to delineate genetic predictors of imaging responses and outcomes., Materials and Methods: This retrospective study included 31 women (average age, 52 y) with liver metastasis from invasive ductal carcinoma who underwent resin and glass radioembolization (average cumulative dose, 2.0 GBq ± 1.8) between January 2011 and September 2017 after receiving ≥ 3 lines of chemotherapy. Twenty-four underwent genetic profiling with MSK-IMPACT or Sequenom; 26 had positron-emission tomography (PET)/CT imaging before and after treatment. Survival after the first radioembolization and 2-4-month PET/CT imaging response were assessed. Laboratory and imaging features were assessed to determine variables predictive of outcomes. Unpaired Student t tests and Fisher exact tests were used to compare responders and nonresponders categorized by changes in fluorodeoxyglucose avidity. Kaplan-Meier survival analysis was used to determine the impact of predictors on survival after radioembolization., Results: Median survival after radioembolization was 11 months (range, 1-49 mo). Most patients (18 of 26; 69%) had complete or partial response based on changes in fluorodeoxyglucose avidity. Imaging response was associated with longer survival (P = .005). Whereas 100% of patients with PI3K pathway mutations showed an imaging response, only 45% of wild-type patients showed a response (P = .01). Median survival did not differ between PI3K pathway wild-type (10.9 mo) and mutant (undefined) patients (P = .50)., Conclusions: These preliminary data suggest that genomic profiling may predict which patients with metastatic breast cancer benefit most from radioembolization. PI3K pathway mutations are associated with improved imaging response, which is associated with longer survival., (Copyright © 2018 SIR. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
29. Perspectives in immunotherapy: meeting report from the Immunotherapy Bridge (29-30 November, 2017, Naples, Italy).
- Author
-
Ascierto PA, Brugarolas J, Buonaguro L, Butterfield LH, Carbone D, Daniele B, Ferris R, Fox BA, Galon J, Gridelli C, Kaufman HL, Klebanoff CA, Melero I, Nathan P, Paulos CM, Ruella M, Sullivan R, Zarour H, and Puzanov I
- Subjects
- Animals, Antigen Presentation immunology, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor, CTLA-4 Antigen antagonists & inhibitors, Cancer Vaccines immunology, Humans, Immunomodulation drug effects, Liquid Biopsy, Mutation, Neoplasms diagnosis, Neoplasms metabolism, Programmed Cell Death 1 Receptor antagonists & inhibitors, T-Lymphocytes drug effects, T-Lymphocytes immunology, T-Lymphocytes metabolism, Translational Research, Biomedical, Tumor Microenvironment drug effects, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy
- Abstract
Immunotherapy represents the third important wave in the history of the systemic treatment of cancer after chemotherapy and targeted therapy and is now established as a potent and effective treatment option across several cancer types. The clinical success of anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4, first, and anti-programmed death (PD)-1/PD-ligand (L)1 agents in melanoma and other cancers a few years later, has encouraged increasing focus on the development of other immunotherapies (e.g. monoclonal antibodies with other immune targets, adoptive cell transfer, and vaccines), with over 3000 immuno-oncology trials ongoing, involving hundreds of research institutes across the globe. The potential use of these different immunotherapeutic options in various combinations with one another and with other treatment modalities is an area of particular promise. The third Immunotherapy Bridge meeting (29-30 November, 2017, Naples, Italy) focused on recent advances in immunotherapy across various cancer types and is summarised in this report.
- Published
- 2018
- Full Text
- View/download PDF
30. Beyond Cell Death: New Functions for TNF Family Cytokines in Autoimmunity and Tumor Immunotherapy.
- Author
-
Yi F, Frazzette N, Cruz AC, Klebanoff CA, and Siegel RM
- Subjects
- Animals, Apoptosis physiology, Cell Proliferation physiology, Humans, Immunotherapy methods, Neoplasms immunology, Autoimmunity physiology, Cell Death physiology, Cytokines metabolism, Neoplasms metabolism, Neoplasms therapy, Tumor Necrosis Factor-alpha metabolism
- Abstract
Originally discovered as an inducer of apoptosis, the TNF-family receptor Fas (CD95, APO-1, TNFRSF6) has more recently been found to have functions beyond cell death, including T cell co-stimulation and promoting terminal differentiation of CD4
+ and CD8+ T cells. Other TNF family members also discovered as apoptosis inducers, such as TRAIL (APO-2L, TNFSF10), can promote inflammation through caspase-8. Surprisingly, non-apoptotic signaling through Fas can protect from the autoimmunity seen in Fas deficiency independently from the cell death inducing functions of the receptor. Non-apoptotic Fas signaling can induce tumor cell growth and migration, and impair the efficacy of T cell adoptive immunotherapy. Blocking of non-apoptotic functions of these receptors may be a novel strategy to regulate autoimmunity and inflammation, and enhance antitumor immunity., (Copyright © 2018. Published by Elsevier Ltd.)- Published
- 2018
- Full Text
- View/download PDF
31. Silencing stemness in T cell differentiation.
- Author
-
Henning AN, Klebanoff CA, and Restifo NP
- Subjects
- Humans, Cell Differentiation, Neoplastic Stem Cells
- Published
- 2018
- Full Text
- View/download PDF
32. Shared cancer neoantigens: Making private matters public.
- Author
-
Klebanoff CA and Wolchok JD
- Subjects
- Cell- and Tissue-Based Therapy, Epitopes, T-Lymphocyte, Glioma, Humans, Histones, Mutation
- Abstract
In this issue of JEM, Chheda et al. (https://doi.org/10.1084/jem.20171046) report that a conserved hotspot mutation associated with an aggressive form of brain cancer generates an immunogenic T cell epitope restricted by a common HLA subtype, thereby creating a "public" neoantigen., (© 2018 Klebanoff and Wolchok.)
- Published
- 2018
- Full Text
- View/download PDF
33. Inhibition of AKT signaling uncouples T cell differentiation from expansion for receptor-engineered adoptive immunotherapy.
- Author
-
Klebanoff CA, Crompton JG, Leonardi AJ, Yamamoto TN, Chandran SS, Eil RL, Sukumar M, Vodnala SK, Hu J, Ji Y, Clever D, Black MA, Gurusamy D, Kruhlak MJ, Jin P, Stroncek DF, Gattinoni L, Feldman SA, and Restifo NP
- Subjects
- Animals, Cell Differentiation, Female, Forkhead Box Protein O1 metabolism, Gene Expression Regulation immunology, Humans, Immunologic Memory, L-Selectin metabolism, Lymphocyte Activation immunology, Mice, Inbred NOD, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Signal Transduction immunology, Transduction, Genetic methods, Xenograft Model Antitumor Assays, Immunotherapy, Adoptive methods, Proto-Oncogene Proteins c-akt immunology, Receptors, Chimeric Antigen immunology, T-Lymphocyte Subsets immunology, Tissue Engineering methods
- Abstract
Adoptive immunotherapies using T cells genetically redirected with a chimeric antigen receptor (CAR) or T cell receptor (TCR) are entering mainstream clinical practice. Despite encouraging results, some patients do not respond to current therapies. In part, this phenomenon has been associated with infusion of reduced numbers of early memory T cells. Herein, we report that AKT signaling inhibition is compatible with CAR and TCR retroviral transduction of human T cells while promoting a CD62L-expressing central memory phenotype. Critically, this intervention did not compromise cell yield. Mechanistically, disruption of AKT signaling preserved MAPK activation and promoted the intranuclear localization of FOXO1, a transcriptional regulator of T cell memory. Consequently, AKT signaling inhibition synchronized the transcriptional profile for FOXO1-dependent target genes across multiple donors. Expression of an AKT-resistant FOXO1 mutant phenocopied the influence of AKT signaling inhibition, while addition of AKT signaling inhibition to T cells expressing mutant FOXO1 failed to further augment the frequency of CD62L-expressing cells. Finally, treatment of established B cell acute lymphoblastic leukemia was superior using anti-CD19 CAR-modified T cells transduced and expanded in the presence of an AKT inhibitor compared with conventionally grown T cells. Thus, inhibition of signaling along the PI3K/AKT axis represents a generalizable strategy to generate large numbers of receptor-modified T cells with an early memory phenotype and superior antitumor efficacy.
- Published
- 2017
- Full Text
- View/download PDF
34. Treatment of Patients With Metastatic Cancer Using a Major Histocompatibility Complex Class II-Restricted T-Cell Receptor Targeting the Cancer Germline Antigen MAGE-A3.
- Author
-
Lu YC, Parker LL, Lu T, Zheng Z, Toomey MA, White DE, Yao X, Li YF, Robbins PF, Feldman SA, van der Bruggen P, Klebanoff CA, Goff SL, Sherry RM, Kammula US, Yang JC, and Rosenberg SA
- Subjects
- Administration, Intravenous, Aged, Antineoplastic Agents administration & dosage, CD4-Positive T-Lymphocytes immunology, Female, Genetic Therapy adverse effects, Humans, Immunotherapy, Adoptive adverse effects, Interleukin-2 administration & dosage, Male, Middle Aged, Neoplasm Metastasis, Neoplasms genetics, Neoplasms immunology, Neoplasms pathology, Receptors, Antigen, T-Cell genetics, Time Factors, Transplantation, Autologous, Treatment Outcome, Antigens, Neoplasm immunology, CD4-Positive T-Lymphocytes transplantation, Genetic Therapy methods, HLA-DP beta-Chains immunology, Immunotherapy, Adoptive methods, Neoplasm Proteins immunology, Neoplasms therapy, Receptors, Antigen, T-Cell immunology
- Abstract
Purpose Adoptive transfer of genetically modified T cells is being explored as a treatment for patients with metastatic cancer. Most current strategies use genes that encode major histocompatibility complex (MHC) class I-restricted T-cell receptors (TCRs) or chimeric antigen receptors to genetically modify CD8
+ T cells or bulk T cells for treatment. Here, we evaluated the safety and efficacy of an adoptive CD4+ T-cell therapy using an MHC class II-restricted, HLA-DPB1*0401-restricted TCR that recognized the cancer germline antigen, MAGE-A3 (melanoma-associated antigen-A3). Patients and Methods Patients received a lymphodepleting preparative regimen, followed by adoptive transfer of purified CD4+ T cells, retrovirally transduced with MAGE-A3 TCR plus systemic high-dose IL-2. A cell dose escalation was conducted, starting at 107 total cells and escalating at half-log increments to approximately 1011 cells. Nine patients were treated at the highest dose level (0.78 to 1.23 × 1011 cells). Results Seventeen patients were treated. During the cell dose-escalation phase, an objective complete response was observed in a patient with metastatic cervical cancer who received 2.7 × 109 cells (ongoing at ≥ 29 months). Among nine patients who were treated at the highest dose level, objective partial responses were observed in a patient with esophageal cancer (duration, 4 months), a patient with urothelial cancer (ongoing at ≥ 19 months), and a patient with osteosarcoma (duration, 4 months). Most patients experienced transient fevers and the expected hematologic toxicities from lymphodepletion pretreatment. Two patients experienced transient grade 3 and 4 transaminase elevations. There were no treatment-related deaths. Conclusion These results demonstrate the safety and efficacy of administering autologous CD4+ T cells that are genetically engineered to express an MHC class II-restricted antitumor TCR that targets MAGE-A3. This clinical trial extends the reach of TCR gene therapy for patients with metastatic cancer.- Published
- 2017
- Full Text
- View/download PDF
35. Normalized Synergy Predicts That CD8 Co-Receptor Contribution to T Cell Receptor (TCR) and pMHC Binding Decreases As TCR Affinity Increases in Human Viral-Specific T Cells.
- Author
-
Williams CM, Schonnesen AA, Zhang SQ, Ma KY, He C, Yamamoto T, Eckhardt SG, Klebanoff CA, and Jiang N
- Abstract
The discovery of naturally occurring T cell receptors (TCRs) that confer specific, high-affinity recognition of pathogen and cancer-associated antigens remains a major goal in cellular immunotherapies. The contribution of the CD8 co-receptor to the interaction between the TCR and peptide-bound major histocompatibility complex (pMHC) has previously been correlated with the activation and responsiveness of CD8
+ T cells. However, these studies have been limited to model systems of genetically engineered hybridoma TCRs or transgenic mouse TCRs against either a single epitope or an array of altered peptide ligands. CD8 contribution in a native human antigen-specific T cell response remains elusive. Here, using Hepatitis C Virus-specific precursor CTLs spanning a large range of TCR affinities, we discovered that the functional responsiveness of any given TCR correlated with the contribution of CD8 to TCR/pMHC binding. Furthermore, we found that CD8 contribution to TCR/pMHC binding in the two-dimensional (2D) system was more accurately reflected by normalized synergy (CD8 cooperation normalized by total TCR/pMHC bonds) rather than synergy (total CD8 cooperation) alone. While synergy showed an increasing trend with TCR affinity, normalized synergy was demonstrated to decrease with the increase of TCR affinity. Critically, normalized synergy was shown to correlate with CTL functionality and peptide sensitivity, corroborating three-dimensional (3D) analysis of CD8 contribution with respect to TCR affinity. In addition, we identified TCRs that were independent of CD8 for TCR/pMHC binding. Our results resolve the current discrepancy between 2D and 3D analysis on CD8 contribution to TCR/pMHC binding, and demonstrate that naturally occurring high-affinity TCRs are more capable of CD8-independent interactions that yield greater functional responsiveness even with CD8 blocking. Taken together, our data suggest that addition of the normalized synergy parameter to our previously established TCR discovery platform using 2D TCR affinity and sequence test would allow for selection of TCRs specific to any given antigen with the desirable attributes of high TCR affinity, CD8 co-receptor independence and functional superiority. Utilizing TCRs with less CD8 contribution could be beneficial for adoptive cell transfer immunotherapies using naturally occurring or genetically engineered T cells against viral or cancer-associated antigens.- Published
- 2017
- Full Text
- View/download PDF
36. Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated With High Serum Interleukin-15 Levels.
- Author
-
Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, Xue A, Goff SL, Yang JC, Sherry RM, Klebanoff CA, Kammula US, Sherman M, Perez A, Yuan CM, Feldman T, Friedberg JW, Roschewski MJ, Feldman SA, McIntyre L, Toomey MA, and Rosenberg SA
- Subjects
- Adult, Aged, Antigens, CD19 immunology, Cyclophosphamide administration & dosage, Humans, Interleukin-15 immunology, Lymphoma blood, Lymphoma drug therapy, Lymphoma immunology, Middle Aged, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology, Transplantation Conditioning methods, Vidarabine administration & dosage, Vidarabine analogs & derivatives, Immunotherapy, Adoptive methods, Interleukin-15 blood, Lymphoma therapy, T-Lymphocytes transplantation
- Abstract
Purpose T cells genetically modified to express chimeric antigen receptors (CARs) targeting CD19 (CAR-19) have potent activity against acute lymphoblastic leukemia, but fewer results supporting treatment of lymphoma with CAR-19 T cells have been published. Patients with lymphoma that is chemotherapy refractory or relapsed after autologous stem-cell transplantation have a grim prognosis, and new treatments for these patients are clearly needed. Chemotherapy administered before adoptive T-cell transfer has been shown to enhance the antimalignancy activity of adoptively transferred T cells. Patients and Methods We treated 22 patients with advanced-stage lymphoma in a clinical trial of CAR-19 T cells preceded by low-dose chemotherapy. Nineteen patients had diffuse large B-cell lymphoma, two patients had follicular lymphoma, and one patient had mantle cell lymphoma. Patients received a single dose of CAR-19 T cells 2 days after a low-dose chemotherapy conditioning regimen of cyclophosphamide plus fludarabine. Results The overall remission rate was 73% with 55% complete remissions and 18% partial remissions. Eleven of 12 complete remissions are ongoing. Fifty-five percent of patients had grade 3 or 4 neurologic toxicities that completely resolved. The low-dose chemotherapy conditioning regimen depleted blood lymphocytes and increased serum interleukin-15 (IL-15). Patients who achieved a remission had a median peak blood CAR
+ cell level of 98/μL and those who did not achieve a remission had a median peak blood CAR+ cell level of 15/μL ( P = .027). High serum IL-15 levels were associated with high peak blood CAR+ cell levels ( P = .001) and remissions of lymphoma ( P < .001). Conclusion CAR-19 T cells preceded by low-dose chemotherapy induced remission of advanced-stage lymphoma, and high serum IL-15 levels were associated with the effectiveness of this treatment regimen. CAR-19 T cells will likely become an important treatment for patients with relapsed lymphoma.- Published
- 2017
- Full Text
- View/download PDF
37. 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
38. Landscape of immunogenic tumor antigens in successful immunotherapy of virally induced epithelial cancer.
- Author
-
Stevanović S, Pasetto A, Helman SR, Gartner JJ, Prickett TD, Howie B, Robins HS, Robbins PF, Klebanoff CA, Rosenberg SA, and Hinrichs CS
- Subjects
- Antigens, Neoplasm genetics, Carcinoma, Squamous Cell virology, DNA-Binding Proteins genetics, Female, Human papillomavirus 16 immunology, Human papillomavirus 18 immunology, Humans, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating transplantation, Oncogene Proteins, Viral genetics, Papillomavirus Infections complications, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor metabolism, Repressor Proteins genetics, T-Lymphocytes immunology, T-Lymphocytes transplantation, Uterine Cervical Neoplasms virology, Antigens, Neoplasm immunology, Carcinoma, Squamous Cell therapy, DNA-Binding Proteins immunology, Immunotherapy, Adoptive methods, Oncogene Proteins, Viral immunology, Papillomavirus Infections therapy, Repressor Proteins immunology, Uterine Cervical Neoplasms therapy
- Abstract
Immunotherapy has clinical activity in certain virally associated cancers. However, the tumor antigens targeted in successful treatments remain poorly defined. We used a personalized immunogenomic approach to elucidate the global landscape of antitumor T cell responses in complete regression of human papillomavirus-associated metastatic cervical cancer after tumor-infiltrating adoptive T cell therapy. Remarkably, immunodominant T cell reactivities were directed against mutated neoantigens or a cancer germline antigen, rather than canonical viral antigens. T cells targeting viral tumor antigens did not display preferential in vivo expansion. Both viral and nonviral tumor antigen-specific T cells resided predominantly in the programmed cell death 1 (PD-1)-expressing T cell compartment, which suggests that PD-1 blockade may unleash diverse antitumor T cell reactivities. These findings suggest a new paradigm of targeting nonviral antigens in immunotherapy of virally associated cancers., (Copyright © 2017, American Association for the Advancement of Science.)
- Published
- 2017
- Full Text
- View/download PDF
39. Fas/CD95 prevents autoimmunity independently of lipid raft localization and efficient apoptosis induction.
- Author
-
Cruz AC, Ramaswamy M, Ouyang C, Klebanoff CA, Sengupta P, Yamamoto TN, Meylan F, Thomas SK, Richoz N, Eil R, Price S, Casellas R, Rao VK, Lippincott-Schwartz J, Restifo NP, and Siegel RM
- Subjects
- Animals, Apoptosis genetics, Autoimmunity genetics, Cell Differentiation genetics, Cell Differentiation immunology, Cells, Cultured, HEK293 Cells, Humans, Lipoylation immunology, Membrane Microdomains metabolism, Mice, Inbred C57BL, Mice, Knockout, Mutation, T-Lymphocytes immunology, T-Lymphocytes metabolism, fas Receptor genetics, fas Receptor metabolism, Apoptosis immunology, Autoimmunity immunology, Membrane Microdomains immunology, Mice, Transgenic, fas Receptor immunology
- Abstract
Mutations affecting the apoptosis-inducing function of the Fas/CD95 TNF-family receptor result in autoimmune and lymphoproliferative disease. However, Fas can also costimulate T-cell activation and promote tumour cell growth and metastasis. Palmitoylation at a membrane proximal cysteine residue enables Fas to localize to lipid raft microdomains and induce apoptosis in cell lines. Here, we show that a palmitoylation-defective Fas C194V mutant is defective in inducing apoptosis in primary mouse T cells, B cells and dendritic cells, while retaining the ability to enhance naive T-cell differentiation. Despite inability to efficiently induce cell death, the Fas C194V receptor prevents the lymphoaccumulation and autoimmunity that develops in Fas-deficient mice. These findings indicate that induction of apoptosis through Fas is dependent on receptor palmitoylation in primary immune cells, and Fas may prevent autoimmunity by mechanisms other than inducing apoptosis.
- Published
- 2016
- Full Text
- View/download PDF
40. Customizing Functionality and Payload Delivery for Receptor-Engineered T Cells.
- Author
-
Klebanoff CA and Restifo NP
- Subjects
- Humans, Neoplasms therapy, T-Lymphocytes, Immunotherapy, Adoptive, Receptors, Antigen, T-Cell
- Abstract
Adoptive immunotherapy using receptor engineering to achieve specific tumor targeting by T cells holds much promise for advancing cancer therapy. Here, two studies by Boice et al. and Roybal et al. provide distinct and potentially complimentary approaches to improve the efficacy and curb potential toxicities of this approach., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
41. Ionic immune suppression within the tumour microenvironment limits T cell effector function.
- Author
-
Eil R, Vodnala SK, Clever D, Klebanoff CA, Sukumar M, Pan JH, Palmer DC, Gros A, Yamamoto TN, Patel SJ, Guittard GC, Yu Z, Carbonaro V, Okkenhaug K, Schrump DS, Linehan WM, Roychoudhuri R, and Restifo NP
- Subjects
- Animals, Humans, Immune Tolerance immunology, Immunotherapy methods, Kv1.3 Potassium Channel metabolism, Male, Melanoma metabolism, Melanoma pathology, Melanoma therapy, Membrane Potentials, Mice, Necrosis, Proto-Oncogene Proteins c-akt metabolism, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism, Signal Transduction, Survival Analysis, T-Lymphocytes metabolism, TOR Serine-Threonine Kinases metabolism, Cations, Monovalent metabolism, Melanoma immunology, Potassium metabolism, T-Lymphocytes immunology, Tumor Escape immunology, Tumor Microenvironment immunology
- Abstract
Tumours progress despite being infiltrated by tumour-specific effector T cells. Tumours contain areas of cellular necrosis, which are associated with poor survival in a variety of cancers. Here, we show that necrosis releases intracellular potassium ions into the extracellular fluid of mouse and human tumours, causing profound suppression of T cell effector function. Elevation of the extracellular potassium concentration ([K
+ ]e ) impairs T cell receptor (TCR)-driven Akt-mTOR phosphorylation and effector programmes. Potassium-mediated suppression of Akt-mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A. Although the suppressive effect mediated by elevated [K+ ]e is independent of changes in plasma membrane potential (Vm ), it requires an increase in intracellular potassium ([K+ ]i ). Accordingly, augmenting potassium efflux in tumour-specific T cells by overexpressing the potassium channel Kv 1.3 lowers [K+ ]i and improves effector functions in vitro and in vivo and enhances tumour clearance and survival in melanoma-bearing mice. These results uncover an ionic checkpoint that blocks T cell function in tumours and identify potential new strategies for cancer immunotherapy., Competing Interests: The authors declare no competing financial interests.- Published
- 2016
- Full Text
- View/download PDF
42. Oxygen Sensing by T Cells Establishes an Immunologically Tolerant Metastatic Niche.
- Author
-
Clever D, Roychoudhuri R, Constantinides MG, Askenase MH, Sukumar M, Klebanoff CA, Eil RL, Hickman HD, Yu Z, Pan JH, Palmer DC, Phan AT, Goulding J, Gattinoni L, Goldrath AW, Belkaid Y, and Restifo NP
- Subjects
- Adoptive Transfer, Animals, CD8-Positive T-Lymphocytes enzymology, Glycolysis immunology, Interferon-gamma immunology, Lung pathology, Lung Neoplasms therapy, Lymphocyte Activation, Mice, Mice, Knockout, Neoplasm Metastasis, Neuropilin-1 metabolism, Prolyl Hydroxylases genetics, T-Lymphocytes, Regulatory enzymology, Th1 Cells enzymology, Th1 Cells immunology, CD8-Positive T-Lymphocytes immunology, Lung immunology, Lung Neoplasms immunology, Lung Neoplasms secondary, Oxygen metabolism, Prolyl Hydroxylases metabolism, T-Lymphocytes, Regulatory immunology
- Abstract
Cancer cells must evade immune responses at distant sites to establish metastases. The lung is a frequent site for metastasis. We hypothesized that lung-specific immunoregulatory mechanisms create an immunologically permissive environment for tumor colonization. We found that T-cell-intrinsic expression of the oxygen-sensing prolyl-hydroxylase (PHD) proteins is required to maintain local tolerance against innocuous antigens in the lung but powerfully licenses colonization by circulating tumor cells. PHD proteins limit pulmonary type helper (Th)-1 responses, promote CD4(+)-regulatory T (Treg) cell induction, and restrain CD8(+) T cell effector function. Tumor colonization is accompanied by PHD-protein-dependent induction of pulmonary Treg cells and suppression of IFN-γ-dependent tumor clearance. T-cell-intrinsic deletion or pharmacological inhibition of PHD proteins limits tumor colonization of the lung and improves the efficacy of adoptive cell transfer immunotherapy. Collectively, PHD proteins function in T cells to coordinate distinct immunoregulatory programs within the lung that are permissive to cancer metastasis. PAPERCLIP., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
43. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies.
- Author
-
Sabatino M, Hu J, Sommariva M, Gautam S, Fellowes V, Hocker JD, Dougherty S, Qin H, Klebanoff CA, Fry TJ, Gress RE, Kochenderfer JN, Stroncek DF, Ji Y, and Gattinoni L
- Subjects
- Animals, Antigens, CD19 genetics, B-Lymphocytes pathology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Hematologic Neoplasms genetics, Hematologic Neoplasms immunology, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Receptors, Antigen, T-Cell genetics, Xenograft Model Antitumor Assays, Adoptive Transfer, Antigens, CD19 immunology, B-Lymphocytes immunology, CD8-Positive T-Lymphocytes transplantation, Hematologic Neoplasms therapy, Immunologic Memory, Receptors, Antigen, T-Cell immunology
- Abstract
Long-lived, self-renewing, multipotent T memory stem cells (TSCM) can trigger profound and sustained tumor regression but their rareness poses a major hurdle to their clinical application. Presently, clinically compliant procedures to generate relevant numbers of this T-cell population are undefined. Here, we provide a strategy for deriving large numbers of clinical-grade tumor-redirected TSCM starting from naive precursors. CD8(+)CD62L(+)CD45RA(+) naive T cells enriched by streptamer-based serial-positive selection were activated by CD3/CD28 engagement in the presence of interleukin-7 (IL-7), IL-21, and the glycogen synthase-3β inhibitor TWS119, and genetically engineered to express a CD19-specific chimeric antigen receptor (CD19-CAR). These conditions enabled the generation of CD19-CAR-modified CD8(+) TSCM that were phenotypically, functionally, and transcriptomically equivalent to their naturally occurring counterpart. Compared with CD8(+) T cells generated with clinical protocols currently under investigation, CD19-CAR-modified CD8(+) TSCM exhibited enhanced metabolic fitness and mediated robust, long-lasting antitumor responses against systemic acute lymphoblastic leukemia xenografts. This clinical-grade platform provides the basis for a phase 1 trial evaluating the activity of CD19-CAR-modified CD8(+) TSCM in patients with B-cell malignancies refractory to prior allogeneic hematopoietic stem cell transplantation.
- Published
- 2016
- Full Text
- View/download PDF
44. 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
45. BACH2 regulates CD8(+) T cell differentiation by controlling access of AP-1 factors to enhancers.
- Author
-
Roychoudhuri R, Clever D, Li P, Wakabayashi Y, Quinn KM, Klebanoff CA, Ji Y, Sukumar M, Eil RL, Yu Z, Spolski R, Palmer DC, Pan JH, Patel SJ, Macallan DC, Fabozzi G, Shih HY, Kanno Y, Muto A, Zhu J, Gattinoni L, O'Shea JJ, Okkenhaug K, Igarashi K, Leonard WJ, and Restifo NP
- Subjects
- Adaptive Immunity, Animals, Basic-Leucine Zipper Transcription Factors genetics, CD8-Positive T-Lymphocytes virology, Cell Differentiation genetics, Cells, Cultured, Enhancer Elements, Genetic genetics, Gene Expression Regulation, Immunologic Memory genetics, Lymphocyte Activation genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Oncogene Protein p65(gag-jun), Signal Transduction genetics, Transcription Factor AP-1 genetics, Basic-Leucine Zipper Transcription Factors metabolism, CD8-Positive T-Lymphocytes physiology, Transcription Factor AP-1 metabolism, Vaccinia immunology, Vaccinia virus immunology
- Abstract
T cell antigen receptor (TCR) signaling drives distinct responses depending on the differentiation state and context of CD8(+) T cells. We hypothesized that access of signal-dependent transcription factors (TFs) to enhancers is dynamically regulated to shape transcriptional responses to TCR signaling. We found that the TF BACH2 restrains terminal differentiation to enable generation of long-lived memory cells and protective immunity after viral infection. BACH2 was recruited to enhancers, where it limited expression of TCR-driven genes by attenuating the availability of activator protein-1 (AP-1) sites to Jun family signal-dependent TFs. In naive cells, this prevented TCR-driven induction of genes associated with terminal differentiation. Upon effector differentiation, reduced expression of BACH2 and its phosphorylation enabled unrestrained induction of TCR-driven effector programs., Competing Interests: The authors declare no competing financial interests.
- Published
- 2016
- Full Text
- View/download PDF
46. Lineage relationship of CD8(+) T cell subsets is revealed by progressive changes in the epigenetic landscape.
- Author
-
Crompton JG, Narayanan M, Cuddapah S, Roychoudhuri R, Ji Y, Yang W, Patel SJ, Sukumar M, Palmer DC, Peng W, Wang E, Marincola FM, Klebanoff CA, Zhao K, Tsang JS, Gattinoni L, and Restifo NP
- Subjects
- Animals, Chromatin Assembly and Disassembly genetics, Enhancer Elements, Genetic genetics, Gene Expression Profiling, Histones metabolism, Immunologic Memory genetics, Lymphocyte Subsets metabolism, Methylation, Mice, Inbred C57BL, Promoter Regions, Genetic genetics, Protein Processing, Post-Translational, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Cell Lineage genetics, Cell Lineage immunology, Epigenesis, Genetic
- Abstract
To better elucidate epigenetic mechanisms that correlate with the dynamic gene expression program observed upon T-cell differentiation, we investigated the genomic landscape of histone modifications in naive and memory CD8(+) T cells. Using a ChIP-Seq approach coupled with global gene expression profiling, we generated genome-wide histone H3 lysine 4 (H3K4me3) and H3 lysine 27 (H3K27me3) trimethylation maps in naive, T memory stem cells, central memory cells, and effector memory cells in order to gain insight into how histone architecture is remodeled during T cell differentiation. We show that H3K4me3 histone modifications are associated with activation of genes, while H3K27me3 is negatively correlated with gene expression at canonical loci and enhancers associated with T-cell metabolism, effector function, and memory. Our results also reveal histone modifications and gene expression signatures that distinguish the recently identified T memory stem cells from other CD8(+) T-cell subsets. Taken together, our results suggest that CD8(+) lymphocytes undergo chromatin remodeling in a progressive fashion. These findings have major implications for our understanding of peripheral T-cell ontogeny and the formation of immunological memory.
- Published
- 2016
- Full Text
- View/download PDF
47. The transcription factor BACH2 promotes tumor immunosuppression.
- Author
-
Roychoudhuri R, Eil RL, Clever D, Klebanoff CA, Sukumar M, Grant FM, Yu Z, Mehta G, Liu H, Jin P, Ji Y, Palmer DC, Pan JH, Chichura A, Crompton JG, Patel SJ, Stroncek D, Wang E, Marincola FM, Okkenhaug K, Gattinoni L, and Restifo NP
- Subjects
- Animals, Basic-Leucine Zipper Transcription Factors genetics, CD8-Positive T-Lymphocytes pathology, Interferon-gamma genetics, Interferon-gamma immunology, Mice, Mice, Knockout, Neoplasms genetics, Neoplasms pathology, T-Lymphocytes, Regulatory pathology, Adaptive Immunity, Basic-Leucine Zipper Transcription Factors immunology, CD8-Positive T-Lymphocytes immunology, Immunity, Innate, Neoplasms immunology, T-Lymphocytes, Regulatory immunology
- Abstract
The immune system has a powerful ability to recognize and kill cancer cells, but its function is often suppressed within tumors, preventing clearance of disease. Functionally diverse innate and adaptive cellular lineages either drive or constrain immune reactions within tumors. The transcription factor (TF) BACH2 regulates the differentiation of multiple innate and adaptive cellular lineages, but its role in controlling tumor immunity has not been elucidated. Here, we demonstrate that BACH2 is required to establish immunosuppression within tumors. Tumor growth was markedly impaired in Bach2-deficient mice and coincided with intratumoral activation of both innate and adaptive immunity. However, augmented tumor clearance in the absence of Bach2 was dependent upon the adaptive immune system. Analysis of tumor-infiltrating lymphocytes from Bach2-deficient mice revealed high frequencies of rapidly proliferating effector CD4+ and CD8+ T cells that expressed the inflammatory cytokine IFN-γ. Effector T cell activation coincided with a reduction in the frequency of intratumoral Foxp3+ Tregs. Mechanistically, BACH2 promoted tumor immunosuppression through Treg-mediated inhibition of intratumoral CD8+ T cells and IFN-γ. These findings demonstrate that BACH2 is a key component of the molecular program of tumor immunosuppression and identify therapeutic targets for the reversal of immunosuppression in cancer.
- Published
- 2016
- Full Text
- View/download PDF
48. Mitochondrial Membrane Potential Identifies Cells with Enhanced Stemness for Cellular Therapy.
- Author
-
Sukumar M, Liu J, Mehta GU, Patel SJ, Roychoudhuri R, Crompton JG, Klebanoff CA, Ji Y, Li P, Yu Z, Whitehill GD, Clever D, Eil RL, Palmer DC, Mitra S, Rao M, Keyvanfar K, Schrump DS, Wang E, Marincola FM, Gattinoni L, Leonard WJ, Muranski P, Finkel T, and Restifo NP
- Subjects
- Animals, CD8-Positive T-Lymphocytes physiology, Cell Line, Tumor, Cytokines physiology, Hematopoietic Stem Cells physiology, Humans, Lymphoid Progenitor Cells transplantation, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Transplantation, Oxidative Stress, Stem Cell Transplantation, T-Lymphocyte Subsets transplantation, Transcriptome, Lymphoid Progenitor Cells physiology, Melanoma, Experimental therapy, Membrane Potential, Mitochondrial, T-Lymphocyte Subsets physiology
- Abstract
Long-term survival and antitumor immunity of adoptively transferred CD8(+) T cells is dependent on their metabolic fitness, but approaches to isolate therapeutic T cells based on metabolic features are not well established. Here we utilized a lipophilic cationic dye tetramethylrhodamine methyl ester (TMRM) to identify and isolate metabolically robust T cells based on their mitochondrial membrane potential (ΔΨm). Comprehensive metabolomic and gene expression profiling demonstrated global features of improved metabolic fitness in low-ΔΨm-sorted CD8(+) T cells. Transfer of these low-ΔΨm T cells was associated with superior long-term in vivo persistence and an enhanced capacity to eradicate established tumors compared with high-ΔΨm cells. Use of ΔΨm-based sorting to enrich for cells with superior metabolic features was observed in CD8(+), CD4(+) T cell subsets, and long-term hematopoietic stem cells. This metabolism-based approach to cell selection may be broadly applicable to therapies involving the transfer of HSC or lymphocytes for the treatment of viral-associated illnesses and cancer., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
49. Prospects for gene-engineered T cell immunotherapy for solid cancers.
- Author
-
Klebanoff CA, Rosenberg SA, and Restifo NP
- Subjects
- Adoptive Transfer, Antigens, Neoplasm immunology, Humans, Neoplasms immunology, Receptors, Antigen, T-Cell immunology, Genetic Engineering methods, Immunotherapy, Adoptive methods, Neoplasms therapy, Receptors, Antigen, T-Cell genetics, T-Lymphocytes
- Abstract
Adoptive transfer of receptor-engineered T cells has produced impressive results in treating patients with B cell leukemias and lymphomas. This success has captured public imagination and driven academic and industrial researchers to develop similar 'off-the-shelf' receptors targeting shared antigens on epithelial cancers, the leading cause of cancer-related deaths. However, the successful treatment of large numbers of people with solid cancers using this strategy is unlikely to be straightforward. Receptor-engineered T cells have the potential to cause lethal toxicity from on-target recognition of normal tissues, and there is a paucity of truly tumor-specific antigens shared across tumor types. Here we offer our perspective on how expanding the use of genetically redirected T cells to treat the majority of patients with solid cancers will require major technical, manufacturing and regulatory innovations centered around the development of autologous gene therapies targeting private somatic mutations.
- Published
- 2016
- Full Text
- View/download PDF
50. Memory T cell-driven differentiation of naive cells impairs adoptive immunotherapy.
- Author
-
Klebanoff CA, Scott CD, Leonardi AJ, Yamamoto TN, Cruz AC, Ouyang C, Ramaswamy M, Roychoudhuri R, Ji Y, Eil RL, Sukumar M, Crompton JG, Palmer DC, Borman ZA, Clever D, Thomas SK, Patel S, Yu Z, Muranski P, Liu H, Wang E, Marincola FM, Gros A, Gattinoni L, Rosenberg SA, Siegel RM, and Restifo NP
- Subjects
- Animals, Cell Differentiation, Fas Ligand Protein physiology, Female, Melanoma, Experimental therapy, Mice, Mice, Inbred C57BL, Proto-Oncogene Proteins c-akt physiology, T-Lymphocytes cytology, fas Receptor physiology, Immunologic Memory, Immunotherapy, Adoptive, T-Lymphocytes immunology
- Abstract
Adoptive cell transfer (ACT) of purified naive, stem cell memory, and central memory T cell subsets results in superior persistence and antitumor immunity compared with ACT of populations containing more-differentiated effector memory and effector T cells. Despite a clear advantage of the less-differentiated populations, the majority of ACT trials utilize unfractionated T cell subsets. Here, we have challenged the notion that the mere presence of less-differentiated T cells in starting populations used to generate therapeutic T cells is sufficient to convey their desirable attributes. Using both mouse and human cells, we identified a T cell-T cell interaction whereby antigen-experienced subsets directly promote the phenotypic, functional, and metabolic differentiation of naive T cells. This process led to the loss of less-differentiated T cell subsets and resulted in impaired cellular persistence and tumor regression in mouse models following ACT. The T memory-induced conversion of naive T cells was mediated by a nonapoptotic Fas signal, resulting in Akt-driven cellular differentiation. Thus, induction of Fas signaling enhanced T cell differentiation and impaired antitumor immunity, while Fas signaling blockade preserved the antitumor efficacy of naive cells within mixed populations. These findings reveal that T cell subsets can synchronize their differentiation state in a process similar to quorum sensing in unicellular organisms and suggest that disruption of this quorum-like behavior among T cells has potential to enhance T cell-based immunotherapies.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.