Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.133
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Cell ; 175(2): 313-326, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30290139

ABSTRACT

Harnessing an antitumor immune response has been a fundamental strategy in cancer immunotherapy. For over a century, efforts have primarily focused on amplifying immune activation mechanisms that are employed by humans to eliminate invaders such as viruses and bacteria. This "immune enhancement" strategy often results in rare objective responses and frequent immune-related adverse events (irAEs). However, in the last decade, cancer immunotherapies targeting the B7-H1/PD-1 pathway (anti-PD therapy), have achieved higher objective response rates in patients with much fewer irAEs. This more beneficial tumor response-to-toxicity profile stems from distinct mechanisms of action that restore tumor-induced immune deficiency selectively in the tumor microenvironment, here termed "immune normalization," which has led to its FDA approval in more than 10 cancer indications and facilitated its combination with different therapies. In this article, we wish to highlight the principles of immune normalization and learn from it, with the ultimate goal to guide better designs for future cancer immunotherapies.


Subject(s)
Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , B7-H1 Antigen/drug effects , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Combined Modality Therapy/methods , Humans , Immunotherapy/trends , Programmed Cell Death 1 Receptor/drug effects , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/drug effects
11.
Cell ; 175(2): 442-457.e23, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30290143

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) critically contribute to the efficacy of anti-tumor therapeutic antibodies. We report here an unexpected finding that macrophages after ADCP inhibit NK cell-mediated ADCC and T cell-mediated cytotoxicity in breast cancers and lymphomas. Mechanistically, AIM2 is recruited to the phagosomes by FcγR signaling following ADCP and activated by sensing the phagocytosed tumor DNAs through the disrupted phagosomal membrane, which subsequently upregulates PD-L1 and IDO and causes immunosuppression. Combined treatment with anti-HER2 antibody and inhibitors of PD-L1 and IDO enhances anti-tumor immunity and anti-HER2 therapeutic efficacy in mouse models. Furthermore, neoadjuvant trastuzumab therapy significantly upregulates PD-L1 and IDO in the tumor-associated macrophages (TAMs) of HER2+ breast cancer patients, correlating with poor trastuzumab response. Collectively, our findings unveil a deleterious role of ADCP macrophages in cancer immunosuppression and suggest that therapeutic antibody plus immune checkpoint blockade may provide synergistic effects in cancer treatment.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Cytophagocytosis/immunology , Macrophages/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity/physiology , B7-H1 Antigen/genetics , B7-H1 Antigen/physiology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cytophagocytosis/physiology , DNA-Binding Proteins/physiology , Disease Models, Animal , Female , Humans , Immunotherapy , Killer Cells, Natural/physiology , Lymphoma/immunology , Macrophages/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Phagocytosis/immunology , Phagocytosis/physiology , Phagosomes/physiology , Receptors, IgG/immunology
12.
Cell ; 173(2): 386-399.e12, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625054

ABSTRACT

The role of enhancers, a key class of non-coding regulatory DNA elements, in cancer development has increasingly been appreciated. Here, we present the detection and characterization of a large number of expressed enhancers in a genome-wide analysis of 8928 tumor samples across 33 cancer types using TCGA RNA-seq data. Compared with matched normal tissues, global enhancer activation was observed in most cancers. Across cancer types, global enhancer activity was positively associated with aneuploidy, but not mutation load, suggesting a hypothesis centered on "chromatin-state" to explain their interplay. Integrating eQTL, mRNA co-expression, and Hi-C data analysis, we developed a computational method to infer causal enhancer-gene interactions, revealing enhancers of clinically actionable genes. Having identified an enhancer ∼140 kb downstream of PD-L1, a major immunotherapy target, we validated it experimentally. This study provides a systematic view of enhancer activity in diverse tumor contexts and suggests the clinical implications of enhancers.


Subject(s)
Enhancer Elements, Genetic/genetics , Neoplasms/pathology , Aneuploidy , B7-H1 Antigen/genetics , Chromatin/genetics , Chromatin/metabolism , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/therapy , Sequence Analysis, RNA , Survival Rate
13.
Cell ; 174(3): 549-563.e19, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29937226

ABSTRACT

Chromatin regulators play a broad role in regulating gene expression and, when gone awry, can lead to cancer. Here, we demonstrate that ablation of the histone demethylase LSD1 in cancer cells increases repetitive element expression, including endogenous retroviral elements (ERVs), and decreases expression of RNA-induced silencing complex (RISC) components. Significantly, this leads to double-stranded RNA (dsRNA) stress and activation of type 1 interferon, which stimulates anti-tumor T cell immunity and restrains tumor growth. Furthermore, LSD1 depletion enhances tumor immunogenicity and T cell infiltration in poorly immunogenic tumors and elicits significant responses of checkpoint blockade-refractory mouse melanoma to anti-PD-1 therapy. Consistently, TCGA data analysis shows an inverse correlation between LSD1 expression and CD8+ T cell infiltration in various human cancers. Our study identifies LSD1 as a potent inhibitor of anti-tumor immunity and responsiveness to immunotherapy and suggests LSD1 inhibition combined with PD-(L)1 blockade as a novel cancer treatment strategy.


Subject(s)
Endogenous Retroviruses/genetics , Histone Demethylases/metabolism , RNA-Induced Silencing Complex/genetics , Animals , Cell Line, Tumor , Chromatin , Combined Modality Therapy , Gene Expression Regulation/genetics , Histone Demethylases/genetics , Humans , Immunity, Cellular , Immunotherapy , Interferon Type I , MCF-7 Cells , Mice , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , RNA, Double-Stranded/genetics , T-Lymphocytes
14.
Immunity ; 55(4): 656-670.e8, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35366396

ABSTRACT

Reinvigoration of exhausted CD8+ T (Tex) cells by checkpoint immunotherapy depends on the activation of precursors of exhausted T (Tpex) cells, but the local anatomical context of their maintenance, differentiation, and interplay with other cells is not well understood. Here, we identified transcriptionally distinct Tpex subpopulations, mapped their differentiation trajectories via transitory cellular states toward Tex cells, and localized these cell states to specific splenic niches. Conventional dendritic cells (cDCs) were critical for successful αPD-L1 therapy and were required to mediate viral control. cDC1s were dispensable for Tpex cell expansion but provided an essential niche to promote Tpex cell maintenance, preventing their overactivation and T-cell-mediated immunopathology. Mechanistically, cDC1s insulated Tpex cells via MHC-I-dependent interactions to prevent their activation within other inflammatory environments that further aggravated their exhaustion. Our findings reveal that cDC1s maintain and safeguard Tpex cells within distinct anatomical niches to balance viral control, exhaustion, and immunopathology.


Subject(s)
CD8-Positive T-Lymphocytes , Dendritic Cells , Cell Differentiation , Immunotherapy , Lymphocyte Count
15.
Immunity ; 54(8): 1883-1900.e5, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34331874

ABSTRACT

Mononuclear phagocytes (MNPs) encompass dendritic cells, monocytes, and macrophages (MoMac), which exhibit antimicrobial, homeostatic, and immunoregulatory functions. We integrated 178,651 MNPs from 13 tissues across 41 datasets to generate a MNP single-cell RNA compendium (MNP-VERSE), a publicly available tool to map MNPs and define conserved gene signatures of MNP populations. Next, we generated a MoMac-focused compendium that revealed an array of specialized cell subsets widely distributed across multiple tissues. Specific pathological forms were expanded in cancer and inflammation. All neoplastic tissues contained conserved tumor-associated macrophage populations. In particular, we focused on IL4I1+CD274(PD-L1)+IDO1+ macrophages, which accumulated in the tumor periphery in a T cell-dependent manner via interferon-γ (IFN-γ) and CD40/CD40L-induced maturation from IFN-primed monocytes. IL4I1_Macs exhibited immunosuppressive characteristics through tryptophan degradation and promoted the entry of regulatory T cell into tumors. This integrated analysis provides a robust online-available platform for uniform annotation and dissection of specific macrophage functions in healthy and pathological states.


Subject(s)
Dendritic Cells/immunology , Gene Expression/immunology , Monocytes/immunology , Transcriptome/genetics , Tumor-Associated Macrophages/immunology , Arthritis, Rheumatoid/immunology , COVID-19/immunology , Gene Expression/genetics , Gene Expression Profiling , Humans , Interferon-gamma/immunology , L-Amino Acid Oxidase/metabolism , Liver Cirrhosis/immunology , Macrophages/immunology , Neoplasms/immunology , RNA, Small Cytoplasmic/genetics , Single-Cell Analysis , T-Lymphocytes, Regulatory/immunology , Transcriptome/immunology
16.
Cell ; 162(6): 1242-56, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26359984

ABSTRACT

Therapeutic antibodies targeting programmed cell death 1 (PD-1) activate tumor-specific immunity and have shown remarkable efficacy in the treatment of melanoma. Yet, little is known about tumor cell-intrinsic PD-1 pathway effects. Here, we show that murine and human melanomas contain PD-1-expressing cancer subpopulations and demonstrate that melanoma cell-intrinsic PD-1 promotes tumorigenesis, even in mice lacking adaptive immunity. PD-1 inhibition on melanoma cells by RNAi, blocking antibodies, or mutagenesis of melanoma-PD-1 signaling motifs suppresses tumor growth in immunocompetent, immunocompromised, and PD-1-deficient tumor graft recipient mice. Conversely, melanoma-specific PD-1 overexpression enhances tumorigenicity, as does engagement of melanoma-PD-1 by its ligand, PD-L1, whereas melanoma-PD-L1 inhibition or knockout of host-PD-L1 attenuate growth of PD-1-positive melanomas. Mechanistically, the melanoma-PD-1 receptor modulates downstream effectors of mTOR signaling. Our results identify melanoma cell-intrinsic functions of the PD-1:PD-L1 axis in tumor growth and suggest that blocking melanoma-PD-1 might contribute to the striking clinical efficacy of anti-PD-1 therapy.


Subject(s)
Melanoma/genetics , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Animals , Antineoplastic Agents/administration & dosage , B7-H1 Antigen/genetics , Cell Line, Tumor , Cells, Cultured , Gene Knockdown Techniques , Heterografts , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation
17.
Immunity ; 53(3): 564-580.e9, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32750334

ABSTRACT

Tumor immune escape limits durable responses to T cell therapy. Here, we examined how regulation and function of gene products that provide the target epitopes for CD8+ T cell anti-tumor immunity influence therapeutic efficacy and resistance. We used a CRISPR-Cas9-based method (CRISPitope) in syngeneic melanoma models to fuse the same model CD8+ T cell epitope to the C-termini of different endogenous gene products. Targeting melanosomal proteins or oncogenic CDK4R24C (Cyclin-dependent kinase 4) by adoptive cell transfer (ACT) of the same epitope-specific CD8+ T cells revealed diverse genetic and non-genetic immune escape mechanisms. ACT directed against melanosomal proteins, but not CDK4R24C, promoted melanoma dedifferentiation, and increased myeloid cell infiltration. CDK4R24C antigen persistence was associated with an interferon-high and T-cell-rich tumor microenvironment, allowing for immune checkpoint inhibition as salvage therapy. Thus, the choice of target antigen determines the phenotype and immune contexture of recurrent melanomas, with implications to the design of cancer immunotherapies.


Subject(s)
Adoptive Transfer/methods , CD8-Positive T-Lymphocytes/transplantation , Epitopes, T-Lymphocyte/immunology , Melanoma/immunology , Melanoma/therapy , Tumor Escape/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell- and Tissue-Based Therapy/methods , Epitopes, T-Lymphocyte/genetics , Gene Knockout Techniques , Immune Checkpoint Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/immunology , Tumor Microenvironment/immunology
18.
Mol Cell ; 81(11): 2317-2331.e6, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33909988

ABSTRACT

Aberrant energy status contributes to multiple metabolic diseases, including obesity, diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4 immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on Ser283, thereby disrupting its interaction with CMTM4 and subsequently triggering PD-L1 degradation. In addition, AMPK phosphorylates EZH2, which disrupts PRC2 function, leading to enhanced IFNs and antigen presentation gene expression. Through these mechanisms, AMPK agonists or ketogenic diets enhance the efficacy of anti-CTLA-4 immunotherapy and improve the overall survival rate in syngeneic mouse tumor models. Our findings reveal a pivotal role for AMPK in regulating the immune response to immune-checkpoint blockade and advocate for combining ketogenic diets or AMPK agonists with anti-CTLA4 immunotherapy to combat cancer.


Subject(s)
AMP-Activated Protein Kinases/genetics , B7-H1 Antigen/genetics , Breast Neoplasms/genetics , CTLA-4 Antigen/genetics , Colorectal Neoplasms/genetics , Immune Checkpoint Inhibitors , AMP-Activated Protein Kinases/immunology , Allografts , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , B7-H1 Antigen/immunology , Biphenyl Compounds/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/therapy , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Diet, Ketogenic/methods , Energy Metabolism/drug effects , Energy Metabolism/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Nude , Pyrones/pharmacology , Signal Transduction , Survival Analysis , Thiophenes/pharmacology
19.
Mol Cell ; 81(6): 1216-1230.e9, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33606996

ABSTRACT

Interferon-γ (IFN-γ)-mediated adaptive resistance is one major barrier to improving immunotherapy in solid tumors. However, the mechanisms are not completely understood. Here, we report that IFN-γ promotes nuclear translocation and phase separation of YAP after anti-PD-1 therapy in tumor cells. Hydrophobic interactions of the YAP coiled-coil domain mediate droplet initiation, and weak interactions of the intrinsically disordered region in the C terminus promote droplet formation. YAP partitions with the transcription factor TEAD4, the histone acetyltransferase EP300, and Mediator1 and forms transcriptional hubs for maximizing target gene transcriptions, independent of the canonical STAT1-IRF1 transcription program. Disruption of YAP phase separation reduced tumor growth, enhanced immune response, and sensitized tumor cells to anti-PD-1 therapy. YAP activity is negatively correlated with patient outcome. Our study indicates that YAP mediates the IFN-γ pro-tumor effect through its nuclear phase separation and suggests that YAP can be used as a predictive biomarker and target of anti-PD-1 combination therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Interferon-gamma/metabolism , Neoplasms, Experimental , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Transcription Factors/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , HEK293 Cells , Humans , Interferon-gamma/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Transcription Factors/genetics , YAP-Signaling Proteins
20.
Genes Dev ; 35(9-10): 602-618, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33888558

ABSTRACT

The DNA damage response (DDR) fulfils essential roles to preserve genome integrity. Targeting the DDR in tumors has had remarkable success over the last decade, exemplified by the licensing of PARP inhibitors for cancer therapy. Recent studies suggest that the application of DDR inhibitors impacts on cellular innate and adaptive immune responses, wherein key DNA repair factors have roles in limiting chronic inflammatory signaling. Antitumor immunity plays an emerging part in cancer therapy, and extensive efforts have led to the development of immune checkpoint inhibitors overcoming immune suppressive signals in tumors. Here, we review the current understanding of the molecular mechanisms underlying DNA damage-triggered immune responses, including cytosolic DNA sensing via the cGAS/STING pathway. We highlight the implications of DDR components for therapeutic outcomes of immune checkpoint inhibitors or their use as biomarkers. Finally, we discuss the rationale for novel combinations of DDR inhibitors with antagonists of immune checkpoints and current hindrances limiting their broader therapeutic applications.


Subject(s)
DNA Repair/physiology , Immunity, Cellular/genetics , Immunotherapy , Neoplasms/therapy , Adaptive Immunity/genetics , DNA Damage/immunology , Discoidin Domain Receptors/antagonists & inhibitors , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL