Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23.528
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 521-550, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38382538

ABSTRACT

Immune checkpoint blockade (ICB) induces a remarkable and durable response in a subset of cancer patients. However, most patients exhibit either primary or acquired resistance to ICB. This resistance arises from a complex interplay of diverse dynamic mechanisms within the tumor microenvironment (TME). These mechanisms include genetic, epigenetic, and metabolic alterations that prevent T cell trafficking to the tumor site, induce immune cell dysfunction, interfere with antigen presentation, drive heightened expression of coinhibitory molecules, and promote tumor survival after immune attack. The TME worsens ICB resistance through the formation of immunosuppressive networks via immune inhibition, regulatory metabolites, and abnormal resource consumption. Finally, patient lifestyle factors, including obesity and microbiome composition, influence ICB resistance. Understanding the heterogeneity of cellular, molecular, and environmental factors contributing to ICB resistance is crucial to develop targeted therapeutic interventions that enhance the clinical response. This comprehensive overview highlights key mechanisms of ICB resistance that may be clinically translatable.


Subject(s)
Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Neoplasms/etiology , Drug Resistance, Neoplasm/immunology , Animals , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Epigenesis, Genetic
2.
Annu Rev Immunol ; 40: 45-74, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35471840

ABSTRACT

The transformative success of antibodies targeting the PD-1 (programmed death 1)/B7-H1 (B7 homolog 1) pathway (anti-PD therapy) has revolutionized cancer treatment. However, only a fraction of patients with solid tumors and some hematopoietic malignancies respond to anti-PD therapy, and the reason for failure in other patients is less known. By dissecting the mechanisms underlying this resistance, current studies reveal that the tumor microenvironment is a major location for resistance to occur. Furthermore, the resistance mechanisms appear to be highly heterogeneous. Here, we discuss recent human cancer data identifying mechanisms of resistance to anti-PD therapy. We review evidence for immune-based resistance mechanisms such as loss of neoantigens, defects in antigen presentation and interferon signaling, immune inhibitory molecules, and exclusion of T cells. We also review the clinical evidence for emerging mechanisms of resistance to anti-PD therapy, such as alterations in metabolism, microbiota, and epigenetics. Finally, we discuss strategies to overcome anti-PD therapy resistance and emphasize the need to develop additional immunotherapies based on the concept of normalization cancer immunotherapy.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Animals , B7-H1 Antigen , Humans , Immunotherapy , Neoplasms/drug therapy , Neoplasms/metabolism , T-Lymphocytes , Tumor Microenvironment
3.
Annu Rev Immunol ; 37: 457-495, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30676822

ABSTRACT

Exhausted CD8 T (Tex) cells are a distinct cell lineage that arise during chronic infections and cancers in animal models and humans. Tex cells are characterized by progressive loss of effector functions, high and sustained inhibitory receptor expression, metabolic dysregulation, poor memory recall and homeostatic self-renewal, and distinct transcriptional and epigenetic programs. The ability to reinvigorate Tex cells through inhibitory receptor blockade, such as αPD-1, highlights the therapeutic potential of targeting this population. Emerging insights into the mechanisms of exhaustion are informing immunotherapies for cancer and chronic infections. However, like other immune cells, Tex cells are heterogeneous and include progenitor and terminal subsets with unique characteristics and responses to checkpoint blockade. Here, we review our current understanding of Tex cell biology, including the developmental paths, transcriptional and epigenetic features, and cell intrinsic and extrinsic factors contributing to exhaustion and how this knowledge may inform therapeutic targeting of Tex cells in chronic infections, autoimmunity, and cancer.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/metabolism , Immunotherapy/methods , Neoplasms/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/physiology , Virus Diseases/immunology , Animals , Cellular Senescence , Chronic Disease , Clonal Anergy , Epigenesis, Genetic , Humans , Neoplasms/therapy , Virus Diseases/therapy
4.
Cell ; 187(16): 4336-4354.e19, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121847

ABSTRACT

Exhausted CD8 T (Tex) cells in chronic viral infection and cancer have sustained co-expression of inhibitory receptors (IRs). Tex cells can be reinvigorated by blocking IRs, such as PD-1, but synergistic reinvigoration and enhanced disease control can be achieved by co-targeting multiple IRs including PD-1 and LAG-3. To dissect the molecular changes intrinsic when these IR pathways are disrupted, we investigated the impact of loss of PD-1 and/or LAG-3 on Tex cells during chronic infection. These analyses revealed distinct roles of PD-1 and LAG-3 in regulating Tex cell proliferation and effector functions, respectively. Moreover, these studies identified an essential role for LAG-3 in sustaining TOX and Tex cell durability as well as a LAG-3-dependent circuit that generated a CD94/NKG2+ subset of Tex cells with enhanced cytotoxicity mediated by recognition of the stress ligand Qa-1b, with similar observations in humans. These analyses disentangle the non-redundant mechanisms of PD-1 and LAG-3 and their synergy in regulating Tex cells.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class I , Lymphocyte Activation Gene 3 Protein , NK Cell Lectin-Like Receptor Subfamily D , Programmed Cell Death 1 Receptor , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Programmed Cell Death 1 Receptor/metabolism , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Mice, Inbred C57BL , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , Cytotoxicity, Immunologic , Cell Proliferation , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology
5.
Cell ; 187(11): 2690-2702.e17, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38723627

ABSTRACT

The quality and quantity of tumor-infiltrating lymphocytes, particularly CD8+ T cells, are important parameters for the control of tumor growth and response to immunotherapy. Here, we show in murine and human cancers that these parameters exhibit circadian oscillations, driven by both the endogenous circadian clock of leukocytes and rhythmic leukocyte infiltration, which depends on the circadian clock of endothelial cells in the tumor microenvironment. To harness these rhythms therapeutically, we demonstrate that efficacy of chimeric antigen receptor T cell therapy and immune checkpoint blockade can be improved by adjusting the time of treatment during the day. Furthermore, time-of-day-dependent T cell signatures in murine tumor models predict overall survival in patients with melanoma and correlate with response to anti-PD-1 therapy. Our data demonstrate the functional significance of circadian dynamics in the tumor microenvironment and suggest the importance of leveraging these features for improving future clinical trial design and patient care.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Lymphocytes, Tumor-Infiltrating , Mice, Inbred C57BL , Tumor Microenvironment , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Circadian Clocks , Circadian Rhythm , Endothelial Cells/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/therapy , Melanoma/pathology , Tumor Microenvironment/immunology
6.
Cell ; 187(13): 3373-3389.e16, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906102

ABSTRACT

The gut microbiota influences the clinical responses of cancer patients to immunecheckpoint inhibitors (ICIs). However, there is no consensus definition of detrimental dysbiosis. Based on metagenomics (MG) sequencing of 245 non-small cell lung cancer (NSCLC) patient feces, we constructed species-level co-abundance networks that were clustered into species-interacting groups (SIGs) correlating with overall survival. Thirty-seven and forty-five MG species (MGSs) were associated with resistance (SIG1) and response (SIG2) to ICIs, respectively. When combined with the quantification of Akkermansia species, this procedure allowed a person-based calculation of a topological score (TOPOSCORE) that was validated in an additional 254 NSCLC patients and in 216 genitourinary cancer patients. Finally, this TOPOSCORE was translated into a 21-bacterial probe set-based qPCR scoring that was validated in a prospective cohort of NSCLC patients as well as in colorectal and melanoma patients. This approach could represent a dynamic diagnosis tool for intestinal dysbiosis to guide personalized microbiota-centered interventions.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Gastrointestinal Microbiome , Immunotherapy , Lung Neoplasms , Neoplasms , Female , Humans , Male , Akkermansia , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Dysbiosis/microbiology , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Lung Neoplasms/microbiology , Lung Neoplasms/drug therapy , Metagenomics/methods , Neoplasms/microbiology , Treatment Outcome
7.
Cell ; 187(2): 375-389.e18, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242085

ABSTRACT

Immune checkpoint inhibition treatment using aPD-1 monoclonal antibodies is a promising cancer immunotherapy approach. However, its effect on tumor immunity is narrow, as most patients do not respond to the treatment or suffer from recurrence. We show that the crosstalk between conventional type I dendritic cells (cDC1) and T cells is essential for an effective aPD-1-mediated anti-tumor response. Accordingly, we developed a bispecific DC-T cell engager (BiCE), a reagent that facilitates physical interactions between PD-1+ T cells and cDC1. BiCE treatment promotes the formation of active dendritic/T cell crosstalk in the tumor and tumor-draining lymph nodes. In vivo, single-cell and physical interacting cell analysis demonstrates the distinct and superior immune reprogramming of the tumors and tumor-draining lymph nodes treated with BiCE as compared to conventional aPD-1 treatment. By bridging immune cells, BiCE potentiates cell circuits and communication pathways needed for effective anti-tumor immunity.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Antibodies, Bispecific/therapeutic use , Dendritic Cells/immunology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology
8.
Cell ; 187(4): 861-881.e32, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38301646

ABSTRACT

Genomic instability can trigger cancer-intrinsic innate immune responses that promote tumor rejection. However, cancer cells often evade these responses by overexpressing immune checkpoint regulators, such as PD-L1. Here, we identify the SNF2-family DNA translocase SMARCAL1 as a factor that favors tumor immune evasion by a dual mechanism involving both the suppression of innate immune signaling and the induction of PD-L1-mediated immune checkpoint responses. Mechanistically, SMARCAL1 limits endogenous DNA damage, thereby suppressing cGAS-STING-dependent signaling during cancer cell growth. Simultaneously, it cooperates with the AP-1 family member JUN to maintain chromatin accessibility at a PD-L1 transcriptional regulatory element, thereby promoting PD-L1 expression in cancer cells. SMARCAL1 loss hinders the ability of tumor cells to induce PD-L1 in response to genomic instability, enhances anti-tumor immune responses and sensitizes tumors to immune checkpoint blockade in a mouse melanoma model. Collectively, these studies uncover SMARCAL1 as a promising target for cancer immunotherapy.


Subject(s)
B7-H1 Antigen , DNA Helicases , Immunity, Innate , Melanoma , Tumor Escape , Animals , Mice , B7-H1 Antigen/metabolism , Genomic Instability , Melanoma/immunology , Melanoma/metabolism , DNA Helicases/metabolism
9.
Cell ; 187(16): 4355-4372.e22, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121848

ABSTRACT

Overcoming immune-mediated resistance to PD-1 blockade remains a major clinical challenge. Enhanced efficacy has been demonstrated in melanoma patients with combined nivolumab (anti-PD-1) and relatlimab (anti-LAG-3) treatment, the first in its class to be FDA approved. However, how these two inhibitory receptors synergize to hinder anti-tumor immunity remains unknown. Here, we show that CD8+ T cells deficient in both PD-1 and LAG-3, in contrast to CD8+ T cells lacking either receptor, mediate enhanced tumor clearance and long-term survival in mouse models of melanoma. PD-1- and LAG-3-deficient CD8+ T cells were transcriptionally distinct, with broad TCR clonality and enrichment of effector-like and interferon-responsive genes, resulting in enhanced IFN-γ release indicative of functionality. LAG-3 and PD-1 combined to drive T cell exhaustion, playing a dominant role in modulating TOX expression. Mechanistically, autocrine, cell-intrinsic IFN-γ signaling was required for PD-1- and LAG-3-deficient CD8+ T cells to enhance anti-tumor immunity, providing insight into how combinatorial targeting of LAG-3 and PD-1 enhances efficacy.


Subject(s)
Antigens, CD , CD8-Positive T-Lymphocytes , Interferon-gamma , Lymphocyte Activation Gene 3 Protein , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Programmed Cell Death 1 Receptor/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Interferon-gamma/metabolism , Mice , Antigens, CD/metabolism , Autocrine Communication , Humans , Melanoma/immunology , Melanoma/drug therapy , Female , Cell Line, Tumor , Melanoma, Experimental/immunology , T-Cell Exhaustion
10.
Cell ; 187(16): 4373-4388.e15, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121849

ABSTRACT

Relatlimab (rela; anti-LAG-3) plus nivolumab (nivo; anti-PD-1) is safe and effective for treatment of advanced melanoma. We designed a trial (NCT03743766) where advanced melanoma patients received rela, nivo, or rela+nivo to interrogate the immunologic mechanisms of rela+nivo. Analysis of biospecimens from this ongoing trial demonstrated that rela+nivo led to enhanced capacity for CD8+ T cell receptor signaling and altered CD8+ T cell differentiation, leading to heightened cytotoxicity despite the retention of an exhaustion profile. Co-expression of cytotoxic and exhaustion signatures was driven by PRDM1, BATF, ETV7, and TOX. Effector function was upregulated in clonally expanded CD8+ T cells that emerged after rela+nivo. A rela+nivo intratumoral CD8+ T cell signature was associated with a favorable prognosis. This intratumoral rela+nivo signature was validated in peripheral blood as an elevated frequency of CD38+TIM3+CD8+ T cells. Overall, we demonstrated that cytotoxicity can be enhanced despite the retention of exhaustion signatures, which will inform future therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation Gene 3 Protein , Melanoma , Programmed Cell Death 1 Receptor , Humans , Antigens, CD/metabolism , Antigens, CD/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cytotoxicity, Immunologic , High Mobility Group Proteins , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Lymphocyte Activation Gene 3 Protein/antagonists & inhibitors , Melanoma/immunology , Melanoma/drug therapy , Melanoma/genetics , Nivolumab/therapeutic use , Nivolumab/pharmacology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction
11.
Annu Rev Immunol ; 34: 539-73, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26927206

ABSTRACT

The immune system is capable of recognizing tumors and eliminates many early malignant cells. However, tumors evolve to evade immune attack, and the tumor microenvironment is immunosuppressive. Immune responses are regulated by a number of immunological checkpoints that promote protective immunity and maintain tolerance. T cell coinhibitory pathways restrict the strength and duration of immune responses, thereby limiting immune-mediated tissue damage, controlling resolution of inflammation, and maintaining tolerance to prevent autoimmunity. Tumors exploit these coinhibitory pathways to evade immune eradication. Blockade of the PD-1 and CTLA-4 checkpoints is proving to be an effective and durable cancer immunotherapy in a subset of patients with a variety of tumor types, and additional combinations are further improving response rates. In this review we discuss the immunoregulatory functions of coinhibitory pathways and their translation to effective immunotherapies for cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Immunotherapy/methods , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Animals , Humans , Immunotherapy/trends , Lymphocyte Activation/drug effects , Neoplasms/immunology , Tumor Escape , Tumor Microenvironment
12.
Cell ; 186(8): 1652-1669, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37059068

ABSTRACT

Immune checkpoint therapy (ICT) has dramatically altered clinical outcomes for cancer patients and conferred durable clinical benefits, including cure in a subset of patients. Varying response rates across tumor types and the need for predictive biomarkers to optimize patient selection to maximize efficacy and minimize toxicities prompted efforts to unravel immune and non-immune factors regulating the responses to ICT. This review highlights the biology of anti-tumor immunity underlying response and resistance to ICT, discusses efforts to address the current challenges with ICT, and outlines strategies to guide the development of subsequent clinical trials and combinatorial efforts with ICT.


Subject(s)
Immunotherapy , Neoplasms , Humans , B7-H1 Antigen , Neoplasms/drug therapy , Clinical Trials as Topic , Immune Checkpoint Inhibitors/administration & dosage
13.
Cell ; 186(7): 1448-1464.e20, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001504

ABSTRACT

Neutrophils accumulate in solid tumors, and their abundance correlates with poor prognosis. Neutrophils are not homogeneous, however, and could play different roles in cancer therapy. Here, we investigate the role of neutrophils in immunotherapy, leading to tumor control. We show that successful therapies acutely expanded tumor neutrophil numbers. This expansion could be attributed to a Sellhi state rather than to other neutrophils that accelerate tumor progression. Therapy-elicited neutrophils acquired an interferon gene signature, also seen in human patients, and appeared essential for successful therapy, as loss of the interferon-responsive transcription factor IRF1 in neutrophils led to failure of immunotherapy. The neutrophil response depended on key components of anti-tumor immunity, including BATF3-dependent DCs, IL-12, and IFNγ. In addition, we found that a therapy-elicited systemic neutrophil response positively correlated with disease outcome in lung cancer patients. Thus, we establish a crucial role of a neutrophil state in mediating effective cancer therapy.


Subject(s)
Lung Neoplasms , Neutrophils , Humans , Lung Neoplasms/genetics , Signal Transduction/genetics , Immunotherapy , Interferons
14.
Cell ; 186(2): 363-381.e19, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36669472

ABSTRACT

Advanced solid cancers are complex assemblies of tumor, immune, and stromal cells characterized by high intratumoral variation. We use highly multiplexed tissue imaging, 3D reconstruction, spatial statistics, and machine learning to identify cell types and states underlying morphological features of known diagnostic and prognostic significance in colorectal cancer. Quantitation of these features in high-plex marker space reveals recurrent transitions from one tumor morphology to the next, some of which are coincident with long-range gradients in the expression of oncogenes and epigenetic regulators. At the tumor invasive margin, where tumor, normal, and immune cells compete, T cell suppression involves multiple cell types and 3D imaging shows that seemingly localized 2D features such as tertiary lymphoid structures are commonly interconnected and have graded molecular properties. Thus, while cancer genetics emphasizes the importance of discrete changes in tumor state, whole-specimen imaging reveals large-scale morphological and molecular gradients analogous to those in developing tissues.


Subject(s)
Adenocarcinoma , Colorectal Neoplasms , Humans , Adenocarcinoma/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Image Processing, Computer-Assisted , Oncogenes , Tumor Microenvironment
15.
Cell ; 185(11): 1924-1942.e23, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35525247

ABSTRACT

For many solid malignancies, lymph node (LN) involvement represents a harbinger of distant metastatic disease and, therefore, an important prognostic factor. Beyond its utility as a biomarker, whether and how LN metastasis plays an active role in shaping distant metastasis remains an open question. Here, we develop a syngeneic melanoma mouse model of LN metastasis to investigate how tumors spread to LNs and whether LN colonization influences metastasis to distant tissues. We show that an epigenetically instilled tumor-intrinsic interferon response program confers enhanced LN metastatic potential by enabling the evasion of NK cells and promoting LN colonization. LN metastases resist T cell-mediated cytotoxicity, induce antigen-specific regulatory T cells, and generate tumor-specific immune tolerance that subsequently facilitates distant tumor colonization. These effects extend to human cancers and other murine cancer models, implicating a conserved systemic mechanism by which malignancies spread to distant organs.


Subject(s)
Lymph Nodes , Melanoma , Animals , Immune Tolerance , Immunotherapy , Lymphatic Metastasis/pathology , Melanoma/pathology , Mice
16.
Cell ; 184(15): 4032-4047.e31, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34171309

ABSTRACT

Although mutations in DNA are the best-studied source of neoantigens that determine response to immune checkpoint blockade, alterations in RNA splicing within cancer cells could similarly result in neoepitope production. However, the endogenous antigenicity and clinical potential of such splicing-derived epitopes have not been tested. Here, we demonstrate that pharmacologic modulation of splicing via specific drug classes generates bona fide neoantigens and elicits anti-tumor immunity, augmenting checkpoint immunotherapy. Splicing modulation inhibited tumor growth and enhanced checkpoint blockade in a manner dependent on host T cells and peptides presented on tumor MHC class I. Splicing modulation induced stereotyped splicing changes across tumor types, altering the MHC I-bound immunopeptidome to yield splicing-derived neoepitopes that trigger an anti-tumor T cell response in vivo. These data definitively identify splicing modulation as an untapped source of immunogenic peptides and provide a means to enhance response to checkpoint blockade that is readily translatable to the clinic.


Subject(s)
Neoplasms/genetics , Neoplasms/immunology , RNA Splicing/genetics , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Epitopes/immunology , Ethylenediamines/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Inflammation/pathology , Mice, Inbred C57BL , Peptides/metabolism , Protein Isoforms/metabolism , Pyrroles/pharmacology , RNA Splicing/drug effects , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
17.
Cell ; 181(7): 1612-1625.e13, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32497499

ABSTRACT

Responses to anti-PD-1 immunotherapy occur but are infrequent in bladder cancer. The specific T cells that mediate tumor rejection are unknown. T cells from human bladder tumors and non-malignant tissue were assessed with single-cell RNA and paired T cell receptor (TCR) sequencing of 30,604 T cells from 7 patients. We find that the states and repertoires of CD8+ T cells are not distinct in tumors compared with non-malignant tissues. In contrast, single-cell analysis of CD4+ T cells demonstrates several tumor-specific states, including multiple distinct states of regulatory T cells. Surprisingly, we also find multiple cytotoxic CD4+ T cell states that are clonally expanded. These CD4+ T cells can kill autologous tumors in an MHC class II-dependent fashion and are suppressed by regulatory T cells. Further, a gene signature of cytotoxic CD4+ T cells in tumors predicts a clinical response in 244 metastatic bladder cancer patients treated with anti-PD-L1.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Biomarkers, Pharmacological/analysis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, MHC Class II , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Lymphocytes, Tumor-Infiltrating , Programmed Cell Death 1 Receptor/genetics , Receptors, Antigen, T-Cell/genetics , Single-Cell Analysis/methods , T-Lymphocytes, Regulatory , Urinary Bladder Neoplasms/immunology
18.
Cell ; 183(2): 347-362.e24, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33064988

ABSTRACT

Neoantigens arise from mutations in cancer cells and are important targets of T cell-mediated anti-tumor immunity. Here, we report the first open-label, phase Ib clinical trial of a personalized neoantigen-based vaccine, NEO-PV-01, in combination with PD-1 blockade in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. This analysis of 82 patients demonstrated that the regimen was safe, with no treatment-related serious adverse events observed. De novo neoantigen-specific CD4+ and CD8+ T cell responses were observed post-vaccination in all of the patients. The vaccine-induced T cells had a cytotoxic phenotype and were capable of trafficking to the tumor and mediating cell killing. In addition, epitope spread to neoantigens not included in the vaccine was detected post-vaccination. These data support the safety and immunogenicity of this regimen in patients with advanced solid tumors (Clinicaltrials.gov: NCT02897765).


Subject(s)
Cancer Vaccines/immunology , Immunotherapy/methods , Precision Medicine/methods , Aged , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/drug therapy , Melanoma/immunology , Middle Aged , Mutation , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/immunology
19.
Cell ; 182(3): 655-671.e22, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32603654

ABSTRACT

Checkpoint blockade with antibodies specific for the PD-1 and CTLA-4 inhibitory receptors can induce durable responses in a wide range of human cancers. However, the immunological mechanisms responsible for severe inflammatory side effects remain poorly understood. Here we report a comprehensive single-cell analysis of immune cell populations in colitis, a common and severe side effect of checkpoint blockade. We observed a striking accumulation of CD8 T cells with highly cytotoxic and proliferative states and no evidence of regulatory T cell depletion. T cell receptor (TCR) sequence analysis demonstrated that a substantial fraction of colitis-associated CD8 T cells originated from tissue-resident populations, explaining the frequently early onset of colitis symptoms following treatment initiation. Our analysis also identified cytokines, chemokines, and surface receptors that could serve as therapeutic targets for colitis and potentially other inflammatory side effects of checkpoint blockade.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CTLA-4 Antigen/immunology , Colitis/metabolism , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Myeloid Cells/metabolism , Receptors, Chemokine/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/metabolism , Chemokines/metabolism , Colitis/drug therapy , Colitis/genetics , Colitis/immunology , Cytokines/metabolism , Flow Cytometry , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Multigene Family , Myeloid Cells/cytology , RNA-Seq , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Receptors, CXCR6/genetics , Receptors, CXCR6/metabolism , Receptors, Chemokine/genetics , Single-Cell Analysis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
20.
Cell ; 177(2): 414-427.e13, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30951669

ABSTRACT

PD-L1 on the surface of tumor cells binds its receptor PD-1 on effector T cells, thereby suppressing their activity. Antibody blockade of PD-L1 can activate an anti-tumor immune response leading to durable remissions in a subset of cancer patients. Here, we describe an alternative mechanism of PD-L1 activity involving its secretion in tumor-derived exosomes. Removal of exosomal PD-L1 inhibits tumor growth, even in models resistant to anti-PD-L1 antibodies. Exosomal PD-L1 from the tumor suppresses T cell activation in the draining lymph node. Systemically introduced exosomal PD-L1 rescues growth of tumors unable to secrete their own. Exposure to exosomal PD-L1-deficient tumor cells suppresses growth of wild-type tumor cells injected at a distant site, simultaneously or months later. Anti-PD-L1 antibodies work additively, not redundantly, with exosomal PD-L1 blockade to suppress tumor growth. Together, these findings show that exosomal PD-L1 represents an unexplored therapeutic target, which could overcome resistance to current antibody approaches.


Subject(s)
B7-H1 Antigen/metabolism , B7-H1 Antigen/physiology , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Exosomes/metabolism , Humans , Immunotherapy , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Tumor Microenvironment/physiology
SELECTION OF CITATIONS
SEARCH DETAIL