41 results on '"Martin W. Lafleur"'
Search Results
2. T Cell Activation Depends on Extracellular Alanine
- Author
-
Noga Ron-Harel, Jonathan M. Ghergurovich, Giulia Notarangelo, Martin W. LaFleur, Yoshiki Tsubosaka, Arlene H. Sharpe, Joshua D. Rabinowitz, and Marcia C. Haigis
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: T cell stimulation is metabolically demanding. To exit quiescence, T cells rely on environmental nutrients, including glucose and the amino acids glutamine, leucine, serine, and arginine. The expression of transporters for these nutrients is tightly regulated and required for T cell activation. In contrast to these amino acids, which are essential or require multi-step biosynthesis, alanine can be made from pyruvate by a single transamination. Here, we show that extracellular alanine is nevertheless required for efficient exit from quiescence during naive T cell activation and memory T cell restimulation. Alanine deprivation leads to metabolic and functional impairments. Mechanistically, this vulnerability reflects the low expression of alanine aminotransferase, the enzyme required for interconverting pyruvate and alanine, whereas activated T cells instead induce alanine transporters. Stable isotope tracing reveals that alanine is not catabolized but instead supports protein synthesis. Thus, T cells depend on exogenous alanine for protein synthesis and normal activation. : In health, T lymphocytes are in a resting state. However, stimulation with their cognate antigen induces massive growth and proliferation. Ron-Harel et al. demonstrate that T cells rely on extracellular alanine for activation. Consumed alanine is used primarily for protein synthesis, and alanine deprivation inhibits T cell metabolism and effector functions. Keywords: T cells, T cell activation, protein synthesis, metabolism, alanine
- Published
- 2019
- Full Text
- View/download PDF
3. Targeting PD-L2–RGMb overcomes microbiome-related immunotherapy resistance
- Author
-
Joon Seok Park, Francesca S. Gazzaniga, Meng Wu, Amalia K. Luthens, Jacob Gillis, Wen Zheng, Martin W. LaFleur, Sarah B. Johnson, Golnaz Morad, Elizabeth M. Park, Yifan Zhou, Stephanie S. Watowich, Jennifer A. Wargo, Gordon J. Freeman, Dennis L. Kasper, and Arlene H. Sharpe
- Subjects
Multidisciplinary - Published
- 2023
4. Supplementary Figure and Table Legends from Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells
- Author
-
Arlene H. Sharpe, Marcia C. Haigis, Justin D. Trombley, Vikram R. Juneja, Kiran Kurmi, Cong-Hui Yao, Alison E. Ringel, Martin W. LaFleur, Isaac S. Harris, Emily F. Gaudiano, Thao H. Nguyen, Tara Muijlwijk, Jacob E. Gillis, and Jefte M. Drijvers
- Abstract
Supplementary Figure and Table Legends
- Published
- 2023
5. Supplementary Figures from Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells
- Author
-
Arlene H. Sharpe, Marcia C. Haigis, Justin D. Trombley, Vikram R. Juneja, Kiran Kurmi, Cong-Hui Yao, Alison E. Ringel, Martin W. LaFleur, Isaac S. Harris, Emily F. Gaudiano, Thao H. Nguyen, Tara Muijlwijk, Jacob E. Gillis, and Jefte M. Drijvers
- Abstract
Supplementary Figures
- Published
- 2023
6. Supplementary Table S1 from Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells
- Author
-
Arlene H. Sharpe, Marcia C. Haigis, Justin D. Trombley, Vikram R. Juneja, Kiran Kurmi, Cong-Hui Yao, Alison E. Ringel, Martin W. LaFleur, Isaac S. Harris, Emily F. Gaudiano, Thao H. Nguyen, Tara Muijlwijk, Jacob E. Gillis, and Jefte M. Drijvers
- Abstract
Supplementary Table S1
- Published
- 2023
7. Supplementary Data File 1 from Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells
- Author
-
Arlene H. Sharpe, Marcia C. Haigis, Justin D. Trombley, Vikram R. Juneja, Kiran Kurmi, Cong-Hui Yao, Alison E. Ringel, Martin W. LaFleur, Isaac S. Harris, Emily F. Gaudiano, Thao H. Nguyen, Tara Muijlwijk, Jacob E. Gillis, and Jefte M. Drijvers
- Abstract
All dose-response curves. Dose-response curves for all library compounds
- Published
- 2023
8. Data from Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells
- Author
-
Arlene H. Sharpe, Marcia C. Haigis, Justin D. Trombley, Vikram R. Juneja, Kiran Kurmi, Cong-Hui Yao, Alison E. Ringel, Martin W. LaFleur, Isaac S. Harris, Emily F. Gaudiano, Thao H. Nguyen, Tara Muijlwijk, Jacob E. Gillis, and Jefte M. Drijvers
- Abstract
Metabolic constraints in the tumor microenvironment constitute a barrier to effective antitumor immunity and similarities in the metabolic properties of T cells and cancer cells impede the specific therapeutic targeting of metabolism in either population. To identify distinct metabolic vulnerabilities of CD8+ T cells and cancer cells, we developed a high-throughput in vitro pharmacologic screening platform and used it to measure the cell type–specific sensitivities of activated CD8+ T cells and B16 melanoma cells to a wide array of metabolic perturbations during antigen-specific killing of cancer cells by CD8+ T cells. We illustrated the applicability of this screening platform by showing that CD8+ T cells were more sensitive to ferroptosis induction by inhibitors of glutathione peroxidase 4 (GPX4) than B16 and MC38 cancer cells. Overexpression of ferroptosis suppressor protein 1 (FSP1) or cytosolic GPX4 yielded ferroptosis-resistant CD8+ T cells without compromising their function, while genetic deletion of the ferroptosis sensitivity–promoting enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) protected CD8+ T cells from ferroptosis but impaired antitumor CD8+ T-cell responses. Our screen also revealed high T cell–specific vulnerabilities for compounds targeting NAD+ metabolism or autophagy and endoplasmic reticulum (ER) stress pathways. We focused the current screening effort on metabolic agents. However, this in vitro screening platform may also be valuable for rapid testing of other types of compounds to identify regulators of antitumor CD8+ T-cell function and potential therapeutic targets.
- Published
- 2023
9. CRISPR Screens to Identify Regulators of Tumor Immunity
- Author
-
Martin W. LaFleur and Arlene H. Sharpe
- Subjects
Cancer Research ,Oncology ,Cell Biology ,Article - Abstract
Cancer immunotherapies, such as immune checkpoint blockade (ICB), have been used in a wide range of tumor types with immense clinical benefit. However, ICB does not work in all patients, and attempts to combine ICB with other immune-based therapies have not lived up to their initial promise. Thus, there is a significant unmet need to discover new targets and combination therapies to extend the benefits of immunotherapy to more patients. Systems biology approaches are well suited for addressing this problem because these approaches enable evaluation of many gene targets simultaneously and ranking their relative importance for a phenotype of interest. As such, loss-of-function CRISPR screens are an emerging set of tools being used to prioritize gene targets for modulating pathways of interest in tumor and immune cells. This review describes the first screens performed to discover cancer immunotherapy targets and the technological advances that will enable next-generation screens.
- Published
- 2023
10. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells
- Author
-
Vikram R. Juneja, Arlene H. Sharpe, Isaac S. Harris, Jacob E. Gillis, Emily F. Gaudiano, Marcia C. Haigis, Thao H. Nguyen, Alison E. Ringel, Tara Muijlwijk, Jefte M. Drijvers, Martin W. LaFleur, Cong-Hui Yao, Kiran Kurmi, and Justin D. Trombley
- Subjects
0301 basic medicine ,Cancer Research ,education.field_of_study ,Tumor microenvironment ,Chemistry ,Endoplasmic reticulum ,Immunology ,Population ,Cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,medicine ,Cancer research ,Cytotoxic T cell ,NAD+ kinase ,education ,CD8 - Abstract
Metabolic constraints in the tumor microenvironment constitute a barrier to effective antitumor immunity and similarities in the metabolic properties of T cells and cancer cells impede the specific therapeutic targeting of metabolism in either population. To identify distinct metabolic vulnerabilities of CD8+ T cells and cancer cells, we developed a high-throughput in vitro pharmacologic screening platform and used it to measure the cell type–specific sensitivities of activated CD8+ T cells and B16 melanoma cells to a wide array of metabolic perturbations during antigen-specific killing of cancer cells by CD8+ T cells. We illustrated the applicability of this screening platform by showing that CD8+ T cells were more sensitive to ferroptosis induction by inhibitors of glutathione peroxidase 4 (GPX4) than B16 and MC38 cancer cells. Overexpression of ferroptosis suppressor protein 1 (FSP1) or cytosolic GPX4 yielded ferroptosis-resistant CD8+ T cells without compromising their function, while genetic deletion of the ferroptosis sensitivity–promoting enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) protected CD8+ T cells from ferroptosis but impaired antitumor CD8+ T-cell responses. Our screen also revealed high T cell–specific vulnerabilities for compounds targeting NAD+ metabolism or autophagy and endoplasmic reticulum (ER) stress pathways. We focused the current screening effort on metabolic agents. However, this in vitro screening platform may also be valuable for rapid testing of other types of compounds to identify regulators of antitumor CD8+ T-cell function and potential therapeutic targets.
- Published
- 2021
11. Batf-mediated epigenetic control of effector CD8 + T cell differentiation
- Author
-
Hsiao-Wei Tsao, James Kaminski, Makoto Kurachi, R. Anthony Barnitz, Michael A. DiIorio, Martin W. LaFleur, Wataru Ise, Tomohiro Kurosaki, E. John Wherry, W. Nicholas Haining, and Nir Yosef
- Subjects
Immunology ,General Medicine - Abstract
The response of naive CD8 + T cells to their cognate antigen involves rapid and broad changes to gene expression that are coupled with extensive chromatin remodeling, but the mechanisms governing these changes are not fully understood. Here, we investigated how these changes depend on the basic leucine zipper ATF-like transcription factor Batf, which is essential for the early phases of the process. Through genome scale profiling, we characterized the role of Batf in chromatin organization at several levels, including the accessibility of key regulatory regions, the expression of their nearby genes, and the interactions that these regions form with each other and with key transcription factors. We identified a core network of transcription factors that cooperated with Batf, including Irf4, Runx3, and T-bet, as indicated by their colocalization with Batf and their binding in regions whose accessibility, interactions, and expression of nearby genes depend on Batf. We demonstrated the synergistic activity of this network by overexpressing the different combinations of these genes in fibroblasts. Batf and Irf4, but not Batf alone, were sufficient to increase accessibility and transcription of key loci, normally associated with T cell function. Addition of Runx3 and T-bet further contributed to fine-tuning of these changes and was essential for establishing chromatin loops characteristic of T cells. These data provide a resource for studying the epigenomic and transcriptomic landscape of effector differentiation of cytotoxic T cells and for investigating the interdependency between transcription factors and its effects on the epigenome and transcriptome of primary cells.
- Published
- 2022
12. Fibroblastic reticular cells enhance T cell metabolism and survival via epigenetic remodeling
- Author
-
Kevin Bi, Jernej Godec, Arlene H. Sharpe, Frank A. Schildberg, Illana A. Stanley, Debattama R. Sen, Flavian D. Brown, Veronika Lukacs-Kornek, Stéphane J. H. Ricoult, Varun N. Kapoor, Nika N. Danial, W. Nicholas Haining, Viviana Cremasco, Brendan D. Manning, Kathleen B. Yates, Shannon J. Turley, Justin D. Trombley, Martin W. LaFleur, and Hye-Jung Kim
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Cell Survival ,Cellular differentiation ,medicine.medical_treatment ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Nitric Oxide ,Article ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Reticular cell ,medicine ,Animals ,Immunology and Allergy ,Lymph node ,Cells, Cultured ,Cell Proliferation ,Mice, Knockout ,Interleukin-6 ,Cell growth ,Chemistry ,Cell Differentiation ,Fibroblasts ,Cellular Reprogramming ,Chromatin Assembly and Disassembly ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Gene Expression Regulation ,Lymph Nodes ,Immunologic Memory ,CD8 ,030215 immunology - Abstract
Lymph node fibroblastic reticular cells (FRCs) respond to signals from activated T cells by releasing nitric oxide, which inhibits T cell proliferation and restricts the size of the expanding T cell pool. Whether interactions with FRCs also support the function or differentiation of activated CD8(+) T cells is not known. Here we report that encounters with FRCs enhanced cytokine production and remodeled chromatin accessibility in newly activated CD8(+) T cells via interleukin-6. These epigenetic changes facilitated metabolic reprogramming and amplified the activity of pro-survival pathways through differential transcription factor activity. Accordingly, FRC conditioning significantly enhanced the persistence of virus-specific CD8+ T cells in vivo and augmented their differentiation into tissue-resident memory T cells. Our study demonstrates that FRCs play a role beyond restricting T cell expansion—they can also shape the fate and function of CD8(+) T cells.
- Published
- 2019
13. PTPN2 regulates the generation of exhausted CD8+ T cell subpopulations and restrains tumor immunity
- Author
-
Kathleen B. Yates, W. Nicholas Haining, Thao H. Nguyen, Rose Al Abosy, Debattama R. Sen, Arlene H. Sharpe, Gordon J. Freeman, Matthew A. Coxe, Jacob E. Gillis, Emily F. Gaudiano, Brian C. Miller, and Martin W. LaFleur
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T cell ,Immunology ,Biology ,Lymphocytic choriomeningitis ,medicine.disease ,3. Good health ,Immune tolerance ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,medicine ,Cancer research ,Immunology and Allergy ,Cytotoxic T cell ,Clone (B-cell biology) ,CD8 ,030215 immunology - Abstract
CD8+ T cell exhaustion is a state of dysfunction acquired in chronic viral infection and cancer, characterized by the formation of Slamf6+ progenitor exhausted and Tim-3+ terminally exhausted subpopulations through unknown mechanisms. Here we establish the phosphatase PTPN2 as a new regulator of the differentiation of the terminally exhausted subpopulation that functions by attenuating type 1 interferon signaling. Deletion of Ptpn2 in CD8+ T cells increased the generation, proliferative capacity and cytotoxicity of Tim-3+ cells without altering Slamf6+ numbers during lymphocytic choriomeningitis virus clone 13 infection. Likewise, Ptpn2 deletion in CD8+ T cells enhanced Tim-3+ anti-tumor responses and improved tumor control. Deletion of Ptpn2 throughout the immune system resulted in MC38 tumor clearance and improved programmed cell death-1 checkpoint blockade responses to B16 tumors. Our results indicate that increasing the number of cytotoxic Tim-3+CD8+ T cells can promote effective anti-tumor immunity and implicate PTPN2 in immune cells as an attractive cancer immunotherapy target.
- Published
- 2019
14. T Cell Activation Depends on Extracellular Alanine
- Author
-
Martin W. LaFleur, Marcia C. Haigis, Jonathan M. Ghergurovich, Yoshiki Tsubosaka, Arlene H. Sharpe, Noga Ron-Harel, Giulia Notarangelo, and Joshua D. Rabinowitz
- Subjects
Alanine ,chemistry.chemical_classification ,Chemistry ,Transamination ,T-Lymphocytes ,Metabolism ,Lymphocyte Activation ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell biology ,Amino acid ,Glutamine ,Serine ,chemistry.chemical_compound ,Mice ,Biosynthesis ,lcsh:Biology (General) ,Animals ,Leucine ,Immunologic Memory ,lcsh:QH301-705.5 - Abstract
Summary: T cell stimulation is metabolically demanding. To exit quiescence, T cells rely on environmental nutrients, including glucose and the amino acids glutamine, leucine, serine, and arginine. The expression of transporters for these nutrients is tightly regulated and required for T cell activation. In contrast to these amino acids, which are essential or require multi-step biosynthesis, alanine can be made from pyruvate by a single transamination. Here, we show that extracellular alanine is nevertheless required for efficient exit from quiescence during naive T cell activation and memory T cell restimulation. Alanine deprivation leads to metabolic and functional impairments. Mechanistically, this vulnerability reflects the low expression of alanine aminotransferase, the enzyme required for interconverting pyruvate and alanine, whereas activated T cells instead induce alanine transporters. Stable isotope tracing reveals that alanine is not catabolized but instead supports protein synthesis. Thus, T cells depend on exogenous alanine for protein synthesis and normal activation. : In health, T lymphocytes are in a resting state. However, stimulation with their cognate antigen induces massive growth and proliferation. Ron-Harel et al. demonstrate that T cells rely on extracellular alanine for activation. Consumed alanine is used primarily for protein synthesis, and alanine deprivation inhibits T cell metabolism and effector functions. Keywords: T cells, T cell activation, protein synthesis, metabolism, alanine
- Published
- 2019
15. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade
- Author
-
Scott J. Rodig, Girish S. Naik, Evisa Gjini, Arpit Panda, Kevin Bi, Seth Maleri, Gabriel K. Griffin, W. Nicholas Haining, Martin W. LaFleur, Sarah A. Weiss, Margaret D. Zimmer, Kristen Felt, Robert T. Manguso, Arlene H. Sharpe, F. Stephen Hodi, Yamini V. Virkud, Jeffrey J. Ishizuka, Jenna L. Collier, Ana Lako, Debattama R. Sen, Juhi R. Kuchroo, Michael Manos, Rose Al Abosy, Brian C. Miller, Kathleen B. Yates, Flavian D. Brown, and Dawn E. Comstock
- Subjects
0301 basic medicine ,Programmed Cell Death 1 Receptor ,Immunology ,Population ,Melanoma, Experimental ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Biology ,complex mixtures ,Article ,Mice, Congenic ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Lymphocytic choriomeningitis virus ,Immunology and Allergy ,Cytotoxic T cell ,Antibodies, Blocking ,Receptor ,education ,Progenitor ,education.field_of_study ,Melanoma ,hemic and immune systems ,medicine.disease ,Lymphocyte Subsets ,Blockade ,Mice, Inbred C57BL ,030104 developmental biology ,Cell culture ,Cancer research ,Female ,human activities ,CD8 ,030215 immunology - Abstract
T cell dysfunction is a hallmark of many cancers, but the basis for T cell dysfunction and the mechanisms by which antibody blockade of the inhibitory receptor PD-1 (anti-PD-1) reinvigorates T cells are not fully understood. Here we show that such therapy acts on a specific subpopulation of exhausted CD8(+) tumor-infiltrating lymphocytes (TILs). Dysfunctional CD8(+) TILs possess canonical epigenetic and transcriptional features of exhaustion that mirror those seen in chronic viral infection. Exhausted CD8(+) TILs include a subpopulation of ‘progenitor exhausted’ cells that retain polyfunctionality, persist long term and differentiate into ‘terminally exhausted’ TILs. Consequently, progenitor exhausted CD8(+) TILs are better able to control tumor growth than are terminally exhausted T cells. Progenitor exhausted TILs can respond to anti-PD-1 therapy, but terminally exhausted TILs cannot. Patients with melanoma who have a higher percentage of progenitor exhausted cells experience a longer duration of response to checkpoint-blockade therapy. Thus, approaches to expand the population of progenitor exhausted CD8(+) T cells might be an important component of improving the response to checkpoint blockade.
- Published
- 2019
16. FAP Delineates Heterogeneous and Functionally Divergent Stromal Cells in Immune-Excluded Breast Tumors
- Author
-
Stephen Santoro, Shilpa Keerthivasan, Viviana Cremasco, Kai W. Wucherpfennig, Lotte Spel, Matthew C. Woodruff, Jillian L. Astarita, Sara Cruz Migoni, Angelo Grauel, Michael P Wu, Martin W. LaFleur, Kenzie MacIsaac, Konstantin Knoblich, Tyler Laszewski, Michael C. Carroll, Shannon J. Turley, Ellen Puré, Anne L. Fletcher, Zohreh Amoozgar, and Glenn Dranoff
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,T-Lymphocytes ,Immunology ,Population ,Breast Neoplasms ,Biology ,Nitric Oxide ,Article ,03 medical and health sciences ,Immune system ,Cancer-Associated Fibroblasts ,Fibroblast activation protein, alpha ,Endopeptidases ,Tumor Microenvironment ,Animals ,Humans ,education ,PDPN ,Cell Proliferation ,Mice, Inbred BALB C ,Tumor microenvironment ,education.field_of_study ,Membrane Glycoproteins ,Serine Endopeptidases ,Mesenchymal stem cell ,Membrane Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Expression Regulation ,Podoplanin ,Gelatinases ,Cancer research ,Female ,Stromal Cells ,Pericytes - Abstract
Cancer-associated fibroblasts (CAFs) are generally associated with poor clinical outcome. CAFs support tumor growth in a variety of ways and can suppress antitumor immunity and response to immunotherapy. However, a precise understanding of CAF contributions to tumor growth and therapeutic response is lacking. Discrepancies in this field of study may stem from heterogeneity in the composition and function of fibroblasts in the tumor microenvironment. Furthermore, it remains unclear whether CAFs directly interact with and suppress T cells. Here, mouse and human breast tumors were used to examine stromal cells expressing fibroblast activation protein (FAP), a surface marker for CAFs. Two discrete populations of FAP+ mesenchymal cells were identified on the basis of podoplanin (PDPN) expression: a FAP+PDPN+ population of CAFs and a FAP+PDPN− population of cancer-associated pericytes (CAPs). Although both subsets expressed extracellular matrix molecules, the CAF transcriptome was enriched in genes associated with TGFβ signaling and fibrosis compared with CAPs. In addition, CAFs were enriched at the outer edge of the tumor, in close contact with T cells, whereas CAPs were localized around vessels. Finally, FAP+PDPN+ CAFs suppressed the proliferation of T cells in a nitric oxide–dependent manner, whereas FAP+PDPN− pericytes were not immunosuppressive. Collectively, these findings demonstrate that breast tumors contain multiple populations of FAP-expressing stromal cells of dichotomous function, phenotype, and location.
- Published
- 2018
17. Epitope spreading toward wild-type melanocyte-lineage antigens rescues suboptimal immune checkpoint blockade responses
- Author
-
Dieter Manstein, Martin W. LaFleur, Arlene H. Sharpe, Marcelo Pereira da Silva, Lajos Kemény, Mack Y. Su, Kristen E. Pauken, Masayoshi Kawakubo, David E. Fisher, S. Alireza Rabi, Tal Hadad Erlich, Catherine J. Wu, Rumya Raghavan, Genevieve M. Boland, Edward P. Browne, Mamunur Rashid, Yu Xu, Mai P. Hoang, Dennie T. Frederick, Jennifer A. Lo, Constance E. Brinckerhoff, Jennifer Allouche, David W. Mullins, David J. Adams, Anita A. J. van der Sande, Vikram R. Juneja, Mohsen Malehmir, Levi A. Garraway, David J. Lieb, Gordon J. Freeman, Keith T. Flaherty, Gyulnara G. Kasumova, Belinda Wang, Elisabeth Roider, Nir Hacohen, Whitney Silkworth, and Qing Yu Weng
- Subjects
business.industry ,Melanoma ,T cell ,Cancer ,Imiquimod ,General Medicine ,medicine.disease ,Article ,Immune checkpoint ,Epitope ,Epitopes ,Mice ,medicine.anatomical_structure ,Antigen ,Antigens, Neoplasm ,medicine ,Cancer research ,Animals ,Humans ,Melanocytes ,business ,Immune Checkpoint Inhibitors ,CD8 ,medicine.drug - Abstract
Although immune checkpoint inhibitors (ICIs), such as anti-programmed cell death protein 1 (PD-1), can deliver durable anti-tumor effects, most patients with cancer fail to respond. Recent studies suggest that ICI efficacy correlates with a higher load of tumor-specific neoantigens and development of vitiligo in patients with melanoma. Here, we report that patients with low melanoma neoantigen burdens who responded to ICIs had tumors with higher expression of pigmentation-related genes. Moreover, expansion of peripheral blood CD8(+) T cell populations specific for melanocyte antigens was observed only in patients who responded to anti-PD-1 therapy, suggesting that ICIs can promote breakdown of tolerance toward tumor-lineage self-antigens. In a mouse model of poorly immunogenic melanomas, skewing of epitope recognition toward wild type melanocyte antigens was associated with markedly improved anti-PD-1 efficacy in two independent approaches: introduction of neoantigens by ultraviolet (UV) B radiation mutagenesis, or the therapeutic combination of ablative fractional photothermolysis plus imiquimod. Complete responses against UV mutation-bearing tumors after anti-PD-1 resulted in protection from subsequent engraftment of melanomas lacking any shared neoantigens, as well as pancreatic adenocarcinomas forcibly overexpressing melanocyte-lineage antigens. Our data demonstrate that somatic mutations are sufficient to provoke strong anti-tumor responses after checkpoint blockade, but long-term responses are not restricted to these putative neoantigens. Epitope skewing toward T cell recognition of wild type tumor-lineage self-antigens represents a common pathway for successful response to ICIs, which can be evoked in neoantigen-deficient tumors by combination therapy with ablative fractional photothermolysis and imiquimod.
- Published
- 2021
18. Batf-mediated Epigenetic Control of Effector CD8+ T Cell Differentiation
- Author
-
Wataru Ise, James Kaminski, R. Anthony Barnitz, W. Nicholas Haining, Makoto Kurachi, Nir Yosef, E. John Wherry, Michael A. DiIorio, Martin W. LaFleur, Tomohiko Kurosaki, and Hsiao-Wei Tsao
- Subjects
Effector ,BATF ,Cytotoxic T cell ,Epigenetics ,Biology ,Transcription factor ,Chromatin remodeling ,Epigenomics ,Cell biology ,Chromatin - Abstract
SUMMARYThe response of cytotoxic T cells to their cognate antigen involves rapid and broad changes in gene expression that are coupled with extensive chromatin remodeling. Here, we study the mechanisms by which the basic leucine zipper ATF-like transcription factor Batf helps regulate this process. Through genome-scale profiling, we observe critical roles for Batf in inducing transcriptional changes in stimulated naive cells, while affecting the chromatin at several levels, namely binding of key transcription factors, accessibility, and long range contacts. We identify a critical network of transcription factors that cooperate with Batf, including its binding partner Irf4, as well as Runx3 and T-bet, and demonstrate its synergistic activity in initiating aspects of the effector T cells’ transcriptional and epigenetic program in ectopically-induced fibroblasts. Our results provide a comprehensive resource for studying the epigenomic and transcriptomic landscape of effector differentiation of cytotoxic T cells.
- Published
- 2021
19. Control of gasdermin D oligomerization and pyroptosis by the Ragulator-Rag-mTORC1 pathway
- Author
-
Jasmin M. D’Andrea, Jay R. Thiagarajah, Pascal Devant, Elvira Boršić, Charles L. Evavold, Elsy M. Ngwa, Arlene H. Sharpe, Jonathan C. Kagan, Iva Hafner-Bratkovič, Martin W. LaFleur, and John G. Doench
- Subjects
Cell signaling ,Inflammasomes ,Cell Adhesion Molecules, Neuronal ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,Cleavage (embryo) ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,Protein Domains ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Pyroptosis ,Animals ,Humans ,Genetic Testing ,Nerve Growth Factors ,Amino Acids ,Caspase ,Adaptor Proteins, Signal Transducing ,Monomeric GTP-Binding Proteins ,Innate immune system ,Cell Death ,biology ,Effector ,Macrophages ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,Inflammasome ,Phosphate-Binding Proteins ,Cell biology ,Mitochondria ,Mice, Inbred C57BL ,Cytolysis ,biology.protein ,Protein Multimerization ,Reactive Oxygen Species ,medicine.drug ,RNA, Guide, Kinetoplastida ,Signal Transduction - Abstract
The process of pyroptosis is mediated by inflammasomes and a downstream effector known as gasdermin D (GSDMD). Upon cleavage by inflammasome-associated caspases, the N-terminal domain of GSDMD forms membrane pores that promote cytolysis. Numerous proteins promote GSDMD cleavage, but none are known to be required for pore formation after GSDMD cleavage. Herein, we report a forward genetic screen that identified the Ragulator-Rag complex as being necessary for GSDMD pore formation and pyroptosis in macrophages. Mechanistic analysis revealed that Ragulator-Rag is not required for GSDMD cleavage upon inflammasome activation, but rather promotes GSDMD oligomerization in the plasma membrane. Defects in GSDMD oligomerization and pore formation can be rescued by mitochondrial poisons that stimulate reactive oxygen species (ROS) production, and ROS modulation impacts the ability of inflammasome pathways to promote pore formation downstream of GSDMD cleavage. These findings reveal an unexpected link between key regulators of immunity (inflammasome-GSDMD) and metabolism (Ragulator-Rag).
- Published
- 2020
20. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8
- Author
-
Jefte M, Drijvers, Jacob E, Gillis, Tara, Muijlwijk, Thao H, Nguyen, Emily F, Gaudiano, Isaac S, Harris, Martin W, LaFleur, Alison E, Ringel, Cong-Hui, Yao, Kiran, Kurmi, Vikram R, Juneja, Justin D, Trombley, Marcia C, Haigis, and Arlene H, Sharpe
- Subjects
Antineoplastic Agents ,CD8-Positive T-Lymphocytes ,Endoplasmic Reticulum ,Article ,Mice, Inbred C57BL ,Mice ,Cell Line, Tumor ,Neoplasms ,Autophagy ,Tumor Cells, Cultured ,Animals ,Ferroptosis ,Humans ,Female - Abstract
Metabolic constraints in the tumor microenvironment constitute a barrier to effective anti-tumor immunity and similarities in the metabolic properties of T cells and cancer cells impede the specific therapeutic targeting of metabolism in either population. To identify distinct metabolic vulnerabilities of CD8(+) T cells and cancer cells, we developed a high-throughput in vitro pharmacologic screening platform and used it to measure the cell type–specific sensitivities of activated CD8(+) T cells and B16 melanoma cells to a wide array of metabolic perturbations during antigen-specific killing of cancer cells by CD8(+) T cells. We illustrated the applicability of this screening platform by showing that CD8(+) T cells were more sensitive to ferroptosis than B16 and MC38 cancer cells. Overexpression of ferroptosis suppressor protein 1 (FSP1) or cytosolic GPX4 yielded ferroptosis-resistant CD8(+) T cells without compromising their function, while genetic deletion of the ferroptosis sensitivity–promoting enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) protected CD8(+) T cells from ferroptosis, but impaired anti-tumor CD8(+) T cell responses. Our screen also revealed high T cell–specific vulnerabilities for compounds targeting NAD(+) metabolism or autophagy and ER stress pathways. We focused the current screening effort on metabolic agents. However, this in vitro screening platform may also be valuable for rapid testing of other types of compounds to identify regulators of anti-tumor CD8(+) T-cell function and potential therapeutic targets.
- Published
- 2020
21. Inhibitors of the PD-1 Pathway in Tumor Therapy
- Author
-
Yuki Muroyama, Arlene H. Sharpe, Charles G. Drake, and Martin W. LaFleur
- Subjects
0301 basic medicine ,T-Lymphocytes ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Immunology ,Drug Evaluation, Preclinical ,Drug resistance ,Article ,B7-H1 Antigen ,Immunomodulation ,Translational Research, Biomedical ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Neoplasms ,Biomarkers, Tumor ,Animals ,Humans ,Immunology and Allergy ,Combined Modality Therapy ,Medicine ,Neoplasm ,Molecular Targeted Therapy ,Adverse effect ,business.industry ,Clinical Studies as Topic ,Translation (biology) ,Prognosis ,medicine.disease ,Blockade ,Treatment Outcome ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,business ,Signal Transduction - Abstract
The programmed death 1 (PD-1) pathway delivers inhibitory signals that function as a brake for immune responses. This pathway limits the initiation and duration of immune responses, thereby protecting tissues from immune-mediated damage and autoimmune diseases. However, the PD-1 pathway also inhibits immune responses to tumors. The critical role of PD-1 in preventing antitumor immunity is demonstrated by the transformative effects of PD-1 pathway blockade in a broad range of cancers with the hallmark of durability of response. Despite this success, most patients do not respond to PD-1 monotherapy, and some patients experience adverse events. In this review, we discuss the functions of the PD-1 pathway and its translation to cancer immunotherapy. We also consider current challenges and opportunities for PD-1 cancer immunotherapy, including mechanisms of response and resistance, identification of biomarkers of response to PD-1 therapy, characterization and treatment of PD-1 therapy–related adverse events, and development of safe and effective combination therapies.
- Published
- 2018
22. Abstract NG04: Disrupting enhancers within the core epigenetic program of exhaustion improves CD8+ T cell responses and enhances tumor control
- Author
-
Debattama Sen, Sarah A. Weiss, Brian C. Miller, Kathleen B. Yates, Martin W. Lafleur, Arlene H. Sharpe, and W. Nicholas Haining
- Subjects
Cancer Research ,Oncology - Abstract
T cell exhaustion describes an acquired dysfunction common in settings of cancer and chronic viral infection. Despite clinical efforts to rescue exhaustion, the fundamental mechanisms specifying this state, and the potential for reprogramming exhausted T cells, remain poorly understood. We profiled accessible chromatin in chronic viral infection to show that exhausted CD8+ cells acquire a state-specific landscape of enhancers that profoundly differs from functional memory. By comparing antigen-specific T cells in several contexts of T cell dysfunction, we found that CD8+ tumor infiltrating lymphocytes share significant epigenetic and transcriptional features with chronic viral infection, highlighting that T cell exhaustion is a fundamental adaptation to settings of chronic stimulation. Critically, we identify a core epigenetic signature, independent of disease-specific milieu, that can act as a precise biomarker of the exhausted state. Comparison of mouse cells to those isolated from patients infected with HCV or HIV showed that the core epigenetic program of exhaustion is conserved across species. Importantly, curative therapy, which reduces viral antigen load, as well as checkpoint blockade immunotherapy, which reduces inhibitory T cell signaling, failed to reverse the exhausted epigenetic profile. T cell exhaustion is therefore a stable epigenetic state that is not rescued by common treatment modalities. We then sought new strategies to modulate T cell exhaustion. We identified a novel candidate enhancer near the PD-1 gene that is unique to exhausted CD8+ T cells and a component of the core epigenetic program. Using Cas9-mediated genome editing, we generated a novel mouse strain with germ-line deletion of this region to characterize the role of this enhancer in vivo. We observed 2-3-fold enrichment of PD-1 enhancer-null cells over control cells in chronic infection, suggesting that CD8+ T cells in these mice might be less prone to exhaustion. Importantly, PD-1 enhancer-null cells had increased persistence without any deficits in functionality as has been described with the full PD-1 gene knock-out. As a result, deletion of the PD-1 enhancer gave rise to significantly higher numbers of IFNg+ immunotherapy-responsive T cells compared to both WT and PD-1 gene ablation. These data suggest that deletion of a state-specific enhancer in immune cells can promote unique functional capacities from those observed with the full gene knock-out. Next, we wanted to understand the role of this enhancer in regulating CD8+ T cell responses in the tumor microenvironment. We found that PD-1 enhancer-null mice exhibit slower tumor growth and increased survival when challenged with either B16-ova melanoma or LLC-ova lung carcinoma. Moreover, PD-1 enhancer-null CD8+ T cells outcompete WT cells in the tumor and preferentially differentiate into functional effectors. The establishment of a core program of T cell exhaustion and increased insight into its epigenetic modulation has crucial implications for the future of immunotherapy. Furthermore, our work suggests that perturbing exhaustion-specific enhancers in T cells could be used to prevent sustained expression of inhibitory genes without damaging the gene locus itself in CAR-T based clinical trials, where there is intense interest in engineering against T cell exhaustion. Citation Format: Debattama Sen, Sarah A. Weiss, Brian C. Miller, Kathleen B. Yates, Martin W. Lafleur, Arlene H. Sharpe, W. Nicholas Haining. Disrupting enhancers within the core epigenetic program of exhaustion improves CD8+ T cell responses and enhances tumor control [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr NG04.
- Published
- 2021
23. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity
- Author
-
Kathleen A. McGuire, Arlene H. Sharpe, W. Nicholas Haining, Robert T. Manguso, Natalie B. Collins, Martin W. LaFleur, Vikram R. Juneja, and Gordon J. Freeman
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,Immunology ,Programmed Cell Death 1 Receptor ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,B7-H1 Antigen ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,PD-L1 ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Cytotoxicity ,Melanoma ,Research Articles ,Tumor microenvironment ,biology ,Chemistry ,Brief Definitive Report ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,030104 developmental biology ,Tumor Escape ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Colorectal Neoplasms ,CD8 - Abstract
Both tumor- and host-derived PD-L1 can play critical roles in immunosuppression; differences in tumor immunogenicity appear to underlie their relative contributions. Juneja et al. show that in immunogenic MC38 tumors, PD-L1 on tumor cells dominates in suppressing tumor immunity by inhibiting CD8 T cell cytotoxicity., It is unclear whether PD-L1 on tumor cells is sufficient for tumor immune evasion or simply correlates with an inflamed tumor microenvironment. We used three mouse tumor models sensitive to PD-1 blockade to evaluate the significance of PD-L1 on tumor versus nontumor cells. PD-L1 on nontumor cells is critical for inhibiting antitumor immunity in B16 melanoma and a genetically engineered melanoma. In contrast, PD-L1 on MC38 colorectal adenocarcinoma cells is sufficient to suppress antitumor immunity, as deletion of PD-L1 on highly immunogenic MC38 tumor cells allows effective antitumor immunity. MC38-derived PD-L1 potently inhibited CD8+ T cell cytotoxicity. Wild-type MC38 cells outcompeted PD-L1–deleted MC38 cells in vivo, demonstrating tumor PD-L1 confers a selective advantage. Thus, both tumor- and host-derived PD-L1 can play critical roles in immunosuppression. Differences in tumor immunogenicity appear to underlie their relative importance. Our findings establish reduced cytotoxicity as a key mechanism by which tumor PD-L1 suppresses antitumor immunity and demonstrate that tumor PD-L1 is not just a marker of suppressed antitumor immunity.
- Published
- 2017
24. Prevention of CAR-T-cell dysfunction
- Author
-
Arlene H. Sharpe, Martin W. LaFleur, and Brian C. Miller
- Subjects
0301 basic medicine ,business.industry ,medicine.medical_treatment ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Immunotherapy ,Computer Science Applications ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cancer immunotherapy ,Cell culture ,Cancer research ,medicine ,Car t cells ,business ,Tumor immunology ,Transcription factor ,030217 neurology & neurosurgery ,Biotechnology - Abstract
The overexpression of the transcription factor c-Jun improves chimeric-antigen-receptor T-cell functionality and enhances the killing of low-antigen-expressing liquid and solid cancers in mice.
- Published
- 2020
25. Obesity Shapes Metabolism in the Tumor Microenvironment to Suppress Anti-Tumor Immunity
- Author
-
Thao H. Nguyen, Vikram R. Juneja, Peter K. Sorger, Alison E. Ringel, Shakchhi Joshi, Martin W. LaFleur, Brandon M. Gassaway, Gregory J. Baker, Jefte M. Drijvers, Steven P. Gygi, Connor A. Jacobson, Zoltan Maliga, Marcia C. Haigis, Justin D. Trombley, Joshua D. Rabinowitz, Brian C. Miller, Peter T. Sage, Haejin Yoon, Alessia Catozzi, Juan Carlos García-Cañaveras, Cong-Hui Yao, and Arlene H. Sharpe
- Subjects
Proteomics ,Colorectal cancer ,medicine.medical_treatment ,Procollagen-Proline Dioxygenase ,Biology ,CD8-Positive T-Lymphocytes ,Diet, High-Fat ,General Biochemistry, Genetics and Molecular Biology ,Hypoxia-Inducible Factor-Proline Dioxygenases ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Cancer immunotherapy ,Immunity ,Cell Line, Tumor ,Neoplasms ,medicine ,Tumor Microenvironment ,Cytotoxic T cell ,Animals ,Humans ,Obesity ,030304 developmental biology ,Adiposity ,Cell Proliferation ,Mice, Knockout ,0303 health sciences ,Tumor microenvironment ,Principal Component Analysis ,Fatty Acids ,Metabolism ,medicine.disease ,CD8+ T cells, anti-tumor immunity, colorectal cancer, fat oxidation, metabolism, obesity, tumor microenvironment ,Mice, Inbred C57BL ,Kinetics ,HEK293 Cells ,Cancer research ,sense organs ,Infiltration (medical) ,Oxidation-Reduction ,030217 neurology & neurosurgery ,CD8 - Abstract
Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8(+) T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8(+) T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8(+) T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8(+) T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8(+) T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy.
- Published
- 2019
26. Immuno-PET identifies the myeloid compartment as a key contributor to the outcome of the antitumor response under PD-1 blockade
- Author
-
Xia Bu, Robert A. Weinberg, Mohammad Rashidian, Vincent L. Verschoor, Hidde L. Ploegh, Amir Reza Aref, Stephen C. Kolifrath, Anushka Dongre, Christie J. Lau, Arlene H. Sharpe, Martin W. LaFleur, Gordon J. Freeman, Thao H. Nguyen, M. Inmaculada Barrasa, Yun Zhang, and Cloud P. Paweletz
- Subjects
Myeloid ,medicine.medical_treatment ,Population ,Programmed Cell Death 1 Receptor ,Adenocarcinoma ,CD8-Positive T-Lymphocytes ,Mice ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,Distribution (pharmacology) ,Animals ,education ,education.field_of_study ,Multidisciplinary ,CD11b Antigen ,biology ,Chemistry ,RNA ,Immunotherapy ,Neoplasms, Experimental ,Neoplasm Proteins ,medicine.anatomical_structure ,Integrin alpha M ,PNAS Plus ,Positron-Emission Tomography ,Cancer research ,biology.protein ,Female ,Antibody ,Colorectal Neoplasms ,CD8 - Abstract
Immunotherapy using checkpoint-blocking antibodies against PD-1 has produced impressive results in a wide range of cancers. However, the response remains heterogeneous among patients. We used noninvasive immuno-positron emission tomography (PET), using (89)Zr-labeled PEGylated single-domain antibody fragments (nanobodies or VHHs), to explore the dynamics and distribution of intratumoral CD8(+) T cells and CD11b(+) myeloid cells in response to anti–PD-1 treatment in the MC38 colorectal mouse adenocarcinoma model. Responding and nonresponding tumors showed consistent differences in the distribution of CD8(+) and CD11b(+) cells. Anti–PD-1 treatment mobilized CD8(+) T cells from the tumor periphery to a more central location. Only those tumors fully infiltrated by CD8(+) T cells went on to complete resolution. All tumors contained CD11b(+) myeloid cells from the outset of treatment, with later recruitment of additional CD11b(+) cells. As tumors grew, the distribution of intratumoral CD11b(+) cells became more heterogeneous. Shrinkage of tumors in responders correlated with an increase in the CD11b(+) population in the center of the tumors. The changes in distribution of CD8(+) and CD11b(+) cells, as assessed by PET, served as biomarkers to gauge the efficacy of anti–PD-1 treatment. Single-cell RNA sequencing of RNA from intratumoral CD45(+) cells showed that CD11b(+) cells in responders and nonresponders were markedly different. The responders exhibited a dominant population of macrophages with an M1-like signature, while the CD45(+) population in the nonresponders displayed an M2-like transcriptional signature. Thus, by using immuno-PET and single-cell RNA sequencing, we show that anti–PD-1 treatment not only affects interactions of CD8(+) T cells with the tumor but also impacts the intratumoral myeloid compartment.
- Published
- 2019
27. PTPN2 regulates the generation of exhausted CD8
- Author
-
Martin W, LaFleur, Thao H, Nguyen, Matthew A, Coxe, Brian C, Miller, Kathleen B, Yates, Jacob E, Gillis, Debattama R, Sen, Emily F, Gaudiano, Rose, Al Abosy, Gordon J, Freeman, W Nicholas, Haining, and Arlene H, Sharpe
- Subjects
Cytotoxicity, Immunologic ,Male ,Protein Tyrosine Phosphatase, Non-Receptor Type 2 ,Skin Neoplasms ,Melanoma, Experimental ,Mice, Transgenic ,Adenocarcinoma ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Lymphoid Progenitor Cells ,Article ,Mice, Inbred C57BL ,Mice ,Signaling Lymphocytic Activation Molecule Family ,Colonic Neoplasms ,Interferon Type I ,Immune Tolerance ,Animals ,Lymphocytic choriomeningitis virus ,Female ,Immunotherapy ,Hepatitis A Virus Cellular Receptor 2 ,Melanoma ,Cellular Senescence ,Signal Transduction - Abstract
CD8+ T cell exhaustion is a state of dysfunction acquired in chronic viral infection and cancer, characterized by the formation of Slamf6+ progenitor exhausted and Tim-3+ terminally exhausted subpopulations through unknown mechanisms. Here we establish the phosphatase PTPN2 as a novel regulator of the differentiation of the terminally exhausted subpopulation that functions by attenuating type 1 interferon signaling. Deletion of Ptpn2 in CD8+ T cells increased the generation, proliferative capacity, and cytotoxicity of Tim-3+ cells without altering Slamf6+ numbers during LCMV Clone 13 infection. Likewise, Ptpn2-deletion in CD8+ T cells enhanced Tim-3+ anti-tumor responses and improved tumor control. Deletion of Ptpn2 throughout the immune system resulted in MC38 tumor clearance and improved PD-1 checkpoint blockade responses to B16 tumors. Our results indicate that increasing the number of cytotoxic Tim-3+ CD8+ T cells can promote effective anti-tumor immunity and implicate PTPN2 in immune cells as an attractive cancer immunotherapy target.
- Published
- 2019
28. Creating CRISPR-Cas9 Knockout Immune Cells using CHIME
- Author
-
W. Nicholas Haining, Thao H. Nguyen, Martin W. LaFleur, Matthew A. Coxe, and Arlene H. Sharpe
- Subjects
Immune system ,General Earth and Planetary Sciences ,CRISPR ,Biology ,General Environmental Science ,Cell biology - Published
- 2019
29. Producing sgRNA-expressing lentivirus for creating chimeras with CHIME
- Author
-
W. Nicholas Haining, Arlene H. Sharpe, Martin W. LaFleur, and Thao H. Nguyen
- Subjects
biology ,Lentivirus ,General Earth and Planetary Sciences ,biology.organism_classification ,Virology ,General Environmental Science ,Subgenomic mRNA - Published
- 2019
30. Abstract PR6: Disrupting enhancers within the core epigenetic program of exhaustion improves T-cell responses and enhances tumor control
- Author
-
Lauer Georg, David Wolski, Sarah A. Weiss, Pierre Tonnerre, W. Nicholas Haining, Kathleen B. Yates, Kevin Bi, Martin W. LaFleur, Debattama R. Sen, Arlene H. Sharpe, Rose Al Abosy, and Brian C. Miller
- Subjects
Cancer Research ,Core (anatomy) ,Chemistry ,Immunology ,Cytotoxic T cell ,Epigenetics ,Enhancer ,Tumor control ,Cell biology - Abstract
T-cell exhaustion describes an acquired dysfunction common in settings of cancer and chronic viral infection. Despite clinical efforts to rescue exhaustion, the fundamental mechanisms specifying this state, and the potential for reprogramming exhausted T cells, remain poorly understood. We profiled accessible chromatin in chronic viral infection to show that exhausted CD8+ cells acquire a state-specific landscape of enhancers that profoundly differs from functional memory. Critically, CD8+ tumor-infiltrating lymphocytes shared significant epigenetic and transcriptional features with chronic viral infection, suggesting that T-cell exhaustion is a fundamental adaptation to settings of chronic stimulation. Comparison of mouse cells to those isolated from patients infected with HCV or HIV showed that the core epigenetic program of exhaustion is conserved across species. Importantly, curative therapy, which reduces viral antigen load, as well as anti-PD-1 immunotherapy, which reduces inhibitory T-cell signaling, failed to reverse the exhausted epigenetic profile. T-cell exhaustion is therefore a stable epigenetic state that is not rescued by common treatment modalities. We then sought new strategies to modulate T-cell exhaustion. We used Cas9-mediated genome editing to generate a novel mouse strain with germline deletion of a core exhaustion-associated enhancer near PD-1. We observed 2- to 3-fold enrichment in vivo of PD-1 enhancer-null cells over control cells in chronic infection, suggesting that CD8+ T cells in these mice might be less prone to exhaustion. PD-1 enhancer-null mice also exhibited slower tumor growth and increased survival when challenged with B16-ova melanoma. The establishment of a core program of T-cell exhaustion and increased insight into its epigenetic modulation has crucial implications for the future of immunotherapy and rational engineering of T cells for clinical use. This abstract is also being presented as Poster A3. Citation Format: Debattama R. Sen, Sarah A. Weiss, Brian C. Miller, Pierre Tonnerre, Rose Al Abosy, Kathleen B. Yates, Kevin Bi, Martin W. Lafleur, David Wolski, Lauer Georg, Arlene H. Sharpe, W. Nicholas Haining. Disrupting enhancers within the core epigenetic program of exhaustion improves T-cell responses and enhances tumor control [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr PR6.
- Published
- 2020
31. Abstract PR9: Loss of PD-1 promotes antitumor immunity by improving functions of both PD-1+ and PD-1- CD8+ T cells in the tumor microenvironment
- Author
-
Vikram R. Juneja, Noga Ron-Harel, Gordon J. Freeman, Jaclyn M. Long, Alison E. Ringel, Martin W. LaFleur, Kelly P. Burke, Juhi R. Kuchroo, Marcia C. Haigis, Nicolas Chevrier, Kristen E. Pauken, Peter T. Sage, Jared H. Rowe, Seth Maleri, and Arlene H. Sharpe
- Subjects
Cancer Research ,Tumor microenvironment ,Antitumor immunity ,Chemistry ,Immunology ,Cancer research ,Cytotoxic T cell - Abstract
Although PD-1 pathway inhibitors are revolutionizing cancer treatment, the mechanisms by which PD-1 regulates antitumor immunity are not fully understood. To determine PD-1 functions on different cell types, we used PD-1 conditional knockout mice. Subcutaneous transplantation of MC38 adenocarcinoma tumor cells in mice with complete deletion of PD-1 selectively on CD8+ T cells improved CD8+ T functions in the tumor microenvironment (TME). CD8+ T cells were required for the protective effects of PD-1 deletion in this model. To assess whether loss of PD-1 on all cells was necessary for improved antitumor immunity, we restricted PD-1 deletion to only half of the T-cell population. We hypothesized that a cell-intrinsic loss of PD-1 was necessary for improved T-cell fitness and effector functions. Here, deletion of PD-1 indeed led to T cell-intrinsic boosts in function. Unexpectedly, however, there was also a bystander effect that improved functions of PD-1-expressing CD8+ T cells in the TME. These data suggest that complete loss of PD-1 is not necessary for optimal tumor immunity, a finding that has important implications for applying PD-1-based immunotherapies to cancer patients. This abstract is also being presented as Poster B82. Citation Format: Kristen E. Pauken, Vikram R. Juneja, Alison Ringel, Jared H. Rowe, Kelly P. Burke, Peter T. Sage, Martin W. LaFleur, Juhi R. Kuchroo, Noga Ron-Harel, Seth Maleri, Jaclyn M. Long, Gordon J. Freeman, Nicolas Chevrier, Marcia C. Haigis, Arlene H. Sharpe. Loss of PD-1 promotes antitumor immunity by improving functions of both PD-1+ and PD-1- CD8+ T cells in the tumor microenvironment [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr PR9.
- Published
- 2020
32. Abstract B47: CHIME screening identifies PTPN2 as a novel regulator of antitumor immunity
- Author
-
Matthew A. Coxe, Debattama R. Sen, Arlene H. Sharpe, Martin W. LaFleur, Thao H. Nguyen, Brian C. Miller, John G. Doench, Jacob E. Gillis, Kathleen B. Yates, and Nicholas Haining
- Subjects
Cancer Research ,Antitumor immunity ,Immunology ,Regulator ,Cancer research ,Biology - Abstract
Despite the significant clinical responses observed with checkpoint blockade, there is an urgent need to identify new therapeutic targets in immune cells for combination therapies. However, the use of functional genomic approaches in immune cells to discover immunotherapy targets is limited by inefficient vector delivery or perturbation of cell states. To circumvent these limitations, we developed CHIME: CHimeric IMmune Editing, a bone marrow chimeric CRISPR-Cas9 delivery system, to rapidly evaluate gene function in innate and adaptive immune cells in vivo without prior ex vivo manipulation. This approach enables efficient deletion of genes of interest in major immune lineages without altering immune development or function. To discover novel immunotherapy targets we performed an in vivo pooled genetic screen for negative regulators of CD8+ T-cell responses to LCMV Clone 13 viral infection. We found that deletion of the phosphatase Ptpn2 enhances CD8+ T-cell responses to chronic pathogens and cancer. In models of both chronic viral infection and transplantable tumors, Ptpn2 null CD8+ T cells expand more and show increased expression of effector genes compared to wild-type cells. Consistent with this, in the LCMV Clone 13 model, Ptpn2 deletion in CD8+ T cells affects the differentiation and expansion of exhausted subpopulations by increasing IFN-I signaling. Furthermore, deletion of Ptpn2 in the immune system induces a CD8+ T cell-dependent clearance of tumors and synergizes with PD-1 immune checkpoint blockade. Our results suggest that therapeutic inhibition of Ptpn2 in immune cells may enhance CD8+ T-cell effector function and mediate antitumor immunity to improve tumor control. More generally, these findings suggest that this genetic platform can enable rapid target discovery through pooled loss-of-function screening in immune cell lineages in vivo and presents a novel target for potential immune based therapies. Citation Format: Martin LaFleur, Thao Nguyen, Matthew Coxe, Brian Miller, Kathleen Yates, Jacob Gillis, Debattama Sen, John Doench, Nicholas Haining, Arlene Sharpe. CHIME screening identifies PTPN2 as a novel regulator of antitumor immunity [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr B47.
- Published
- 2020
33. Abstract A83: Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade
- Author
-
Arlene H. Sharpe, Jeffrey J. Ishizuka, Kathleen B. Yates, Michael Manos, Girish S. Naik, W. Nicholas Haining, Martin W. LaFleur, F. Stephen Hodi, Ana Lako, Yamini V. Virkud, Debattama R. Sen, Scott J. Rodig, Rose Al Abosy, Brian C. Miller, Evisa Gjini, Kristen Felt, and Kevin Bi
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,Melanoma ,medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,medicine.disease ,complex mixtures ,Blockade ,medicine ,Cancer research ,Cytotoxic T cell ,business ,CD8 ,Progenitor - Abstract
T-cell dysfunction in the tumor microenvironment (TME) is a hallmark of many cancers. Reinvigoration of T-cell function by PD-1 checkpoint blockade can result in striking clinical responses, but is only effective in a minority of patients. The mechanisms by which anti-PD-1 therapy acts on exhausted T cells are not fully understood. Here we show that anti-PD-1 therapy acts on a specific subpopulation of CD8+ tumor-infiltrating lymphocytes (TILs) in melanoma mouse models, which can also be found in patients with melanoma. Exhausted CD8+ TILs contain a subpopulation of “progenitor exhausted” T cells with critical functional attributes that are not shared by the majority “terminally exhausted” TILs: they retain more polyfunctionality, persist following transfer into tumor-bearing mice, and differentiate to repopulate terminally exhausted TILs in the TME. As a result, progenitor exhausted CD8+ TILs are better able to control tumor growth than terminally exhausted cells. Progenitor exhausted, but not terminally exhausted, CD8+ TILs can respond to anti-PD-1 therapy. Melanoma patients with a higher percentage of progenitor exhausted cells have a longer duration of response to checkpoint blockade therapy. Therefore, approaches to expand progenitor exhausted CD8+ T cells in the tumor microenvironment may be an important component of improving checkpoint blockade response. Citation Format: Brian C. Miller, Debattama R. Sen, Rose Al Abosy, Kevin Bi, Yamini Virkud, Martin W. LaFleur, Kathleen B. Yates, Ana Lako, Kristen Felt, Girish S. Naik, Michael Manos, Evisa Gjini, Jeffrey J. Ishizuka, F. Stephen Hodi, Scott J. Rodig, Arlene H. Sharpe, W. Nicholas Haining. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2019 Nov 17-20; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(3 Suppl):Abstract nr A83.
- Published
- 2020
34. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target
- Author
-
Sarah A. Weiss, Robert T. Manguso, John G. Doench, Arlene H. Sharpe, Natalie B. Collins, Eliezer M. Van Allen, David E. Fisher, Brian C. Miller, David E. Root, Diana Miao, Kathleen B. Yates, Margaret D. Zimmer, W. Nicholas Haining, Martin W. LaFleur, Flavian D. Brown, Jennifer A. Lo, Hans W. Pope, Kevin Bi, and Vikram R. Juneja
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T-Lymphocytes ,Antigen presentation ,Melanoma, Experimental ,Biology ,03 medical and health sciences ,Mice ,Immune system ,Cancer immunotherapy ,Interferon ,Loss of Function Mutation ,medicine ,Animals ,Humans ,Gene Editing ,Antigen Presentation ,Protein Tyrosine Phosphatase, Non-Receptor Type 2 ,Multidisciplinary ,Melanoma ,NF-kappa B ,Cancer ,Immunotherapy ,Genomics ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Immunology ,Cancer research ,Unfolded Protein Response ,Tumor Escape ,Interferons ,CRISPR-Cas Systems ,medicine.drug ,Genetic screen - Abstract
Immunotherapy with PD-1 checkpoint blockade is effective in only a minority of patients with cancer, suggesting that additional treatment strategies are needed. Here we use a pooled in vivo genetic screening approach using CRISPR-Cas9 genome editing in transplantable tumours in mice treated with immunotherapy to discover previously undescribed immunotherapy targets. We tested 2,368 genes expressed by melanoma cells to identify those that synergize with or cause resistance to checkpoint blockade. We recovered the known immune evasion molecules PD-L1 and CD47, and confirmed that defects in interferon-γ signalling caused resistance to immunotherapy. Tumours were sensitized to immunotherapy by deletion of genes involved in several diverse pathways, including NF-κB signalling, antigen presentation and the unfolded protein response. In addition, deletion of the protein tyrosine phosphatase PTPN2 in tumour cells increased the efficacy of immunotherapy by enhancing interferon-γ-mediated effects on antigen presentation and growth suppression. In vivo genetic screens in tumour models can identify new immunotherapy targets in unanticipated pathways.
- Published
- 2017
35. The epigenetic landscape of T cell exhaustion
- Author
-
Nir Yosef, W. Nicholas Haining, Debattama R. Sen, Damien C. Tully, Ulrike Gerdemann, Flavian D. Brown, Kathleen B. Yates, Pierre Tonnerre, James Kaminski, R. Anthony Barnitz, Nicole Frahm, Jernej Godec, Makoto Kurachi, Raymond T. Chung, Martin W. LaFleur, Georg M. Lauer, Todd M. Allen, Hsiao-Wei Tsao, and E. John Wherry
- Subjects
0301 basic medicine ,HIV Infections ,CD8-Positive T-Lymphocytes ,Inbred C57BL ,B7-H1 Antigen ,Mice ,0302 clinical medicine ,Gene expression ,Chronic ,Genetics ,Regulation of gene expression ,Gene Editing ,Multidisciplinary ,Hepatitis C ,Chromatin ,medicine.anatomical_structure ,Infectious Diseases ,030220 oncology & carcinogenesis ,Immunotherapy ,Infection ,Transcription ,Enhancer Elements ,General Science & Technology ,T cell ,1.1 Normal biological development and functioning ,Biology ,Lymphocytic Choriomeningitis ,complex mixtures ,Article ,03 medical and health sciences ,Genetic ,Underpinning research ,medicine ,Animals ,Humans ,Cell Lineage ,Epigenetics ,Enhancer ,Gene ,Animal ,SOXB1 Transcription Factors ,Prevention ,Human Genome ,030104 developmental biology ,Disease Models ,Chronic Disease ,T-Box Domain Proteins ,Immunologic Memory ,CD8 ,Epigenesis - Abstract
Exhausted T cells in cancer and chronic viral infection express distinctive patterns of genes, including sustained expression of programmed cell death protein 1 (PD-1). However, the regulation of gene expression in exhausted T cells is poorly understood. Here, we define the accessible chromatin landscape in exhausted CD8+ T cells and show that it is distinct from functional memory CD8+ T cells. Exhausted CD8+ T cells in humans and a mouse model of chronic viral infection acquire a state-specific epigenetic landscape organized into functional modules of enhancers. Genome editing shows that PD-1 expression is regulated in part by an exhaustion-specific enhancer that contains essential RAR, T-bet, and Sox3 motifs. Functional enhancer maps may offer targets for genome editing that alter gene expression preferentially in exhausted CD8+ T cells.
- Published
- 2016
36. Author Correction: Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade
- Author
-
Gabriel K. Griffin, Kathleen B. Yates, F. Stephen Hodi, Kevin Bi, Sarah A. Weiss, Seth Maleri, Robert T. Manguso, Arlene H. Sharpe, Yamini V. Virkud, Evisa Gjini, Kristen Felt, Scott J. Rodig, Debattama R. Sen, Flavian D. Brown, Dawn E. Comstock, Martin W. LaFleur, Brian C. Miller, Rose Al Abosy, Jeffrey J. Ishizuka, W. Nicholas Haining, Jenna L. Collier, Margaret D. Zimmer, Ana Lako, Arpit Panda, Girish S. Naik, Juhi R. Kuchroo, and Michael Manos
- Subjects
business.industry ,Immunology ,Cancer research ,Immunology and Allergy ,Medicine ,Cytotoxic T cell ,business ,Tumor control ,Blockade - Published
- 2019
37. Abstract 2701: Functionally specialized subsets of exhausted CD8+ T cells mediate tumor control and differentially respond to checkpoint blockade
- Author
-
Scott J. Rodig, Kathleen B. Yates, Arlene H. Sharpe, Debattama R. Sen, Kristen Felt, Martin W. LaFleur, Ana Lako, Girish S. Naik, Evisa Gjini, Kevin Bi, Brian C. Miller, Rose Al Abosy, W. Nicholas Haining, Yamini V. Virkud, F. Stephen Hodi, and Michael Manos
- Subjects
Cancer Research ,Tumor microenvironment ,Melanoma ,T cell ,Cancer ,Biology ,medicine.disease ,Blockade ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Cytotoxic T cell ,CD8 ,Progenitor - Abstract
T cell dysfunction in the tumor microenvironment (TME) is a hallmark of many cancers. Reinvigoration of T cell function by PD-1 checkpoint blockade can result in striking clinical responses, but is only effective in a minority of patients. The basis for T cell dysfunction in the TME, as well as the mechanisms by which anti-PD-1 therapy acts on dysfunctional T cells are not fully understood. Here we show that anti-PD-1 therapy acts on a specific subpopulation of CD8+ tumor-infiltrating lymphocytes (TILs) in melanoma mouse models, which can also be found in patients with melanoma. We find that dysfunctional CD8+ TILs possess canonical epigenetic and transcriptional features of T cell exhaustion, mirroring those seen in chronic viral infection. Similar to chronic viral infection, exhausted CD8+ TILs contain a subpopulation of “progenitor exhausted” T cells that have a distinct regulatory state. Progenitor exhausted TILs also have critical functional attributes that are not shared by the majority “terminally exhausted” TILs: they retain more polyfunctionality, persist following transfer into tumor-bearing mice, and differentiate to repopulate terminally exhausted TILs in the TME. As a result, progenitor exhausted CD8+ TILs are better able to control tumor growth than terminally exhausted cells. Progenitor exhausted, but not terminally exhausted, CD8+ TILs can respond to anti-PD-1 therapy but this occurs without reversion of their exhausted epigenetic state. Human melanomas contain CD8+ T cells with a progenitor exhausted phenotype and patients with a higher fraction of this subpopulation in their tumors have a significantly longer duration of response to combination checkpoint blockade therapy. Therefore, approaches to expand progenitor exhausted CD8+ T cells in the tumor microenvironment may be an important component of improving checkpoint blockade response. Citation Format: Brian C. Miller, Debattama R. Sen, Rose Al Abosy, Kevin Bi, Yamini V. Virkud, Martin W. LaFleur, Kathleen B. Yates, Ana Lako, Kristen Felt, Girish S. Naik, Michael Manos, Evisa Gjini, F. Stephen Hodi, Scott J. Rodig, Arlene H. Sharpe, W. Nicholas Haining. Functionally specialized subsets of exhausted CD8+ T cells mediate tumor control and differentially respond to checkpoint blockade [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2701.
- Published
- 2019
38. Abstract A216: Functionally specialized subsets of exhausted CD8+ T-cells mediate tumor control and response to checkpoint blockade
- Author
-
Michael Manos, Scott J. Rodig, Yamini V. Virkud, Arlene H. Sharpe, Girish S. Naik, Debattama R. Sen, Kathleen B. Yates, Kevin Bi, Rose Al Abosy, Martin W. LaFleur, Evisa Gjini, Kristen Felt, Brian C. Miller, Ana Lako, Stephen Hodi, and W. Nicholas Haining
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Melanoma ,Immunology ,Cancer ,medicine.disease ,Blockade ,Cancer immunotherapy ,medicine ,Cancer research ,Cytotoxic T cell ,Epigenetics ,business ,CD8 - Abstract
T-cell dysfunction in the tumor microenvironment (TME) is a hallmark of many cancers. Reinvigoration of T-cell function by PD-1 checkpoint blockade can result in striking clinical responses, but is effective only in a minority of patients. The basis for T-cell dysfunction in the TME, as well as the mechanisms by which anti-PD-1 therapy acts on dysfunctional T-cells are not fully understood. Here we show that anti-PD-1 therapy acts on a specific subpopulation of CD8+ tumor-infiltrating lymphocytes (TILs) in melanoma mouse models as well as patients with melanoma. We find that dysfunctional CD8+ TILs possess canonical epigenetic and transcriptional features of T-cell exhaustion, mirroring those seen in chronic viral infection. Similar to chronic viral infection, exhausted CD8+ TILs contain a subpopulation of “stem-like exhausted” T-cells that have a distinct regulatory state. Stem-like exhausted TILs also have critical functional attributes that are not shared by the majority “terminally exhausted” TILs: they retain more polyfunctionality, persist following transfer into tumor-bearing mice, and differentiate to repopulate terminally exhausted TILs in the TME. As a result, stem-like exhausted CD8+ TILs are better able to control tumor growth than terminally exhausted cells. Stem-like exhausted, but not terminally exhausted, CD8+ TILs can respond to anti-PD-1 therapy without reversion of their exhausted epigenetic state. CD8+ T-cells with a stem-like exhausted phenotype can be found in human melanoma samples and patients with a higher fraction of this subpopulation in their tumors have a significantly longer duration of response to combination checkpoint blockade therapy. Responsiveness to checkpoint blockade is therefore restricted to a subpopulation of exhausted TILs that retain specific functional properties which enable them to control tumors. Approaches to expand stem-like exhausted CD8+ T-cells in the tumor microenvironment may be an important component of improving checkpoint blockade response. Citation Format: Debattama R. Sen, Brian C. Miller, Rose Al Abosy, Kevin Bi, Martin W. LaFleur, Kathleen B. Yates, Ana Lako, Kristen D. Felt, Girish S. Naik, Michael Manos, Evisa Gjini, Yamini V. Virkud, Stephen Hodi, Scott J. Rodig, Arlene H. Sharpe, W. Nicholas Haining. Functionally specialized subsets of exhausted CD8+ T-cells mediate tumor control and response to checkpoint blockade [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A216.
- Published
- 2019
39. Enhancing the Efficacy of Checkpoint Blockade Through Combination Therapies
- Author
-
Vikram R. Juneja, Martin W. LaFleur, Arlene H. Sharpe, and Robert T. Manguso
- Subjects
Oncology ,medicine.medical_specialty ,Combination therapy ,business.industry ,medicine.medical_treatment ,Ipilimumab ,Pembrolizumab ,Immunotherapy ,Blockade ,Targeted therapy ,CTLA-4 ,Internal medicine ,Medicine ,Nivolumab ,business ,medicine.drug - Abstract
Antibodies targeting coinhibitory receptors on T cells (“checkpoint blockade”) have emerged as some of the most promising therapies for a broad range of malignancies, including melanoma, non-small cell lung cancer, renal cell carcinoma, Hodgkin’s lymphoma, and bladder cancer. These coinhibitory molecules include CTLA-4, PD-1, LAG-3, TIM-3, and others. The anti-CTLA-4 antibody ipilimumab was approved in 2011 and the anti-PD-1 antibodies pembrolizumab and nivolumab were approved in 2014 for patients with advanced melanoma. Single agent checkpoint blockade is associated with 20–40 % objective response rates in advanced melanoma with improved overall survival. The combination of anti-CTLA-4 and anti-PD-1 antibodies leads to an increased durable response rate compared to either antibody alone, supporting the concept that combination therapy may result in increased clinical benefit. An important goal in the field is to combine checkpoint blockade with other immunotherapies and other types of therapy (e.g., radiation, targeted therapy, chemotherapy, surgery) to increase the fraction of patients that have objective and durable responses. Here, we discuss the current understanding of the mechanisms underlying checkpoint blockade and the rationale for combination therapy. We then discuss potential immunotherapeutic and non-immunotherapeutic combination therapies. Finally, we discuss critical issues that need to be addressed in order to develop combination strategies to induce long-term clinical responses in patients with cancer.
- Published
- 2016
40. Abstract A19: PD-1 modulation promotes antitumor immunity by improving metabolic fitness of both PD-1+ and PD-1- CD8+ T cells in the tumor
- Author
-
Arlene H. Sharpe, Juhi R. Kuchroo, Vikram R. Juneja, Kristen E. Pauken, Nicolas Chevrier, Seth Maleri, Peter T. Sage, Noga Ron-Harel, Gordon J. Freeman, Marcia C. Haigis, Alison E. Ringel, and Martin W. LaFleur
- Subjects
Cancer Research ,Tumor microenvironment ,medicine.medical_treatment ,Immunology ,Priming (immunology) ,Immunotherapy ,Biology ,Transplantation ,Immunity ,Cancer research ,medicine ,Bystander effect ,Cytotoxic T cell ,CD8 - Abstract
Although PD-1 pathway inhibitors are revolutionizing cancer treatment, the mechanisms by which PD-1 regulates anti-tumor immunity are not fully understood. Following subcutaneous transplantation of MC38 adenocarcinoma tumor cells into mice, we show that complete loss of PD-1 selectively on CD8+ T cells improved metabolic activity and functions in the tumor microenvironment (TME). Since clinically PD-1 inhibitors likely act on T cells post-priming, we next deleted PD-1 after initial priming and restricted deletion to roughly 50% of cells. Loss of PD-1 led to T cell-intrinsic boosts in metabolism and CD8+ T cells that lost PD-1 after priming preferentially formed anti-tumor memory cells, suggesting PD-1 antagonizes memory formation. Unexpectedly, there was also a bystander effect that improved functions of PD-1 expressing CD8+ T cells in the TME. These data suggest that complete loss of PD-1 is not necessary for optimal tumor immunity, and that enhancing the functions of a subset of CD8+ T cells can promote an antitumor microenvironment and immunologic memory. Citation Format: Kristen E. Pauken, Vikram R. Juneja, Peter T. Sage, Martin W. LaFleur, Juhi R. Kuchroo, Alison Ringel, Noga Ron-Harel, Seth P. Maleri, Gordon J. Freeman, Nicolas Chevrier, Marcia C. Haigis, Arlene H. Sharpe. PD-1 modulation promotes antitumor immunity by improving metabolic fitness of both PD-1+ and PD-1- CD8+ T cells in the tumor [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A19.
- Published
- 2018
41. Blue Native PAGE and Mass Spectrometry as an Approach for the Investigation of Stable and Transient Protein-Protein Interactions
- Author
-
Melissa Butkiewicz, Costel C. Darie, Martin W. LaFleur, Rama R. Yakubu, Izabela Sokolowska, Christopher E. Talbot, and Alisa G. Woods
- Subjects
Chromatography ,Chemistry ,Transient (computer programming) ,Native page ,Mass spectrometry ,Protein–protein interaction - Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.