Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Commun ; 14(1): 5945, 2023 09 23.
Article in English | MEDLINE | ID: mdl-37741832

ABSTRACT

Microsatellite-stable colorectal cancer (MSS-CRC) is highly refractory to immunotherapy. Understanding tumor-intrinsic determinants of immunotherapy resistance is critical to improve MSS-CRC patient outcomes. Here, we demonstrate that high tumor expression of the core autophagy gene ATG16L1 is associated with poor clinical response to anti-PD-L1 therapy in KRAS-mutant tumors from IMblaze370 (NCT02788279), a large phase III clinical trial of atezolizumab (anti-PD-L1) in advanced metastatic MSS-CRC. Deletion of Atg16l1 in engineered murine colon cancer organoids inhibits tumor growth in primary (colon) and metastatic (liver and lung) niches in syngeneic female hosts, primarily due to increased sensitivity to IFN-γ-mediated immune pressure. ATG16L1 deficiency enhances programmed cell death of colon cancer organoids induced by IFN-γ and TNF, thus increasing their sensitivity to host immunity. In parallel, ATG16L1 deficiency reduces tumor stem-like populations in vivo independently of adaptive immune pressure. This work reveals autophagy as a clinically relevant mechanism of immune evasion and tumor fitness in MSS-CRC and provides a rationale for autophagy inhibition to boost immunotherapy responses in the clinic.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Animals , Female , Humans , Mice , Autophagy/genetics , Autophagy-Related Proteins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, Regulator , Liver , Clinical Trials, Phase III as Topic
2.
PLoS One ; 16(12): e0260800, 2021.
Article in English | MEDLINE | ID: mdl-34879110

ABSTRACT

The cancer-associated fibroblast (CAF) marker podoplanin (PDPN) is generally correlated with poor clinical outcomes in cancer patients and thus represents a promising therapeutic target. Despite its biomedical relevance, basic aspects of PDPN biology such as its cellular functions and cell surface ligands remain poorly uncharacterized, thus challenging drug development. Here, we utilize a high throughput platform to elucidate the PDPN cell surface interactome, and uncover the neutrophil protein CD177 as a new binding partner. Quantitative proteomics analysis of the CAF phosphoproteome reveals a role for PDPN in cell signaling, growth and actomyosin contractility, among other processes. Moreover, cellular assays demonstrate that CD177 is a functional antagonist, recapitulating the phenotype observed in PDPN-deficient CAFs. In sum, starting from the unbiased elucidation of the PDPN co-receptome, our work provides insights into PDPN functions and reveals the PDPN/CD177 axis as a possible modulator of fibroblast physiology in the tumor microenvironment.


Subject(s)
Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Isoantigens/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Tumor Microenvironment , Apoptosis , Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Isoantigens/genetics , Membrane Glycoproteins/genetics , Neutrophils/immunology , Neutrophils/metabolism , Prognosis , Receptors, Cell Surface/genetics , Survival Rate , Tumor Cells, Cultured
3.
Immunity ; 54(7): 1511-1526.e8, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260887

ABSTRACT

Myeloid cells encounter stromal cells and their matrix determinants on a continual basis during their residence in any given organ. Here, we examined the impact of the collagen receptor LAIR1 on myeloid cell homeostasis and function. LAIR1 was highly expressed in the myeloid lineage and enriched in non-classical monocytes. Proteomic definition of the LAIR1 interactome identified stromal factor Colec12 as a high-affinity LAIR1 ligand. Proteomic profiling of LAIR1 signaling triggered by Collagen1 and Colec12 highlighted pathways associated with survival, proliferation, and differentiation. Lair1-/- mice had reduced frequencies of Ly6C- monocytes, which were associated with altered proliferation and apoptosis of non-classical monocytes from bone marrow and altered heterogeneity of interstitial macrophages in lung. Myeloid-specific LAIR1 deficiency promoted metastatic growth in a melanoma model and LAIR1 expression associated with improved clinical outcomes in human metastatic melanoma. Thus, monocytes and macrophages rely on LAIR1 sensing of stromal determinants for fitness and function, with relevance in homeostasis and disease.


Subject(s)
Homeostasis/physiology , Lung/metabolism , Macrophages, Alveolar/metabolism , Monocytes/metabolism , Receptors, Immunologic/metabolism , Animals , Apoptosis/physiology , Bone Marrow/metabolism , Bone Marrow/pathology , COS Cells , Cell Differentiation/physiology , Cell Line , Cell Line, Tumor , Cell Lineage/physiology , Cell Proliferation/physiology , Chlorocebus aethiops , Female , Humans , Lung/pathology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Metastasis/pathology , Proteomics/methods , Signal Transduction/physiology
4.
Elife ; 102021 06 04.
Article in English | MEDLINE | ID: mdl-34085925

ABSTRACT

Defective autophagy is strongly associated with chronic inflammation. Loss-of-function of the core autophagy gene Atg16l1 increases risk for Crohn's disease in part by enhancing innate immunity through myeloid cells such as macrophages. However, autophagy is also recognized as a mechanism for clearance of certain intracellular pathogens. These divergent observations prompted a re-evaluation of ATG16L1 in innate antimicrobial immunity. In this study, we found that loss of Atg16l1 in myeloid cells enhanced the killing of virulent Shigella flexneri (S.flexneri), a clinically relevant enteric bacterium that resides within the cytosol by escaping from membrane-bound compartments. Quantitative multiplexed proteomics of murine bone marrow-derived macrophages revealed that ATG16L1 deficiency significantly upregulated proteins involved in the glutathione-mediated antioxidant response to compensate for elevated oxidative stress, which simultaneously promoted S.flexneri killing. Consistent with this, myeloid-specific deletion of Atg16l1 in mice accelerated bacterial clearance in vitro and in vivo. Pharmacological induction of oxidative stress through suppression of cysteine import enhanced microbial clearance by macrophages. Conversely, antioxidant treatment of macrophages permitted S.flexneri proliferation. These findings demonstrate that control of oxidative stress by ATG16L1 and autophagy regulates antimicrobial immunity against intracellular pathogens.


Subject(s)
Autophagy-Related Proteins/deficiency , Autophagy , Dysentery, Bacillary/microbiology , Immunity, Innate , Macrophages/microbiology , Oxidative Stress , Proteome , Proteomics , Shigella flexneri/pathogenicity , Animals , Autophagy-Related Proteins/genetics , Cells, Cultured , Disease Models, Animal , Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , Shigella flexneri/immunology , Shigella flexneri/metabolism , Virulence
5.
Nat Immunol ; 22(5): 571-585, 2021 05.
Article in English | MEDLINE | ID: mdl-33903764

ABSTRACT

Fibroblastic reticular cells (FRCs) are specialized stromal cells that define tissue architecture and regulate lymphocyte compartmentalization, homeostasis, and innate and adaptive immunity in secondary lymphoid organs (SLOs). In the present study, we used single-cell RNA sequencing (scRNA-seq) of human and mouse lymph nodes (LNs) to identify a subset of T cell-zone FRCs defined by the expression of Gremlin1 (Grem1) in both species. Grem1-CreERT2 knock-in mice enabled localization, multi-omics characterization and genetic depletion of Grem1+ FRCs. Grem1+ FRCs primarily localize at T-B cell junctions of SLOs, neighboring pre-dendritic cells and conventional dendritic cells (cDCs). As such, their depletion resulted in preferential loss and decreased homeostatic proliferation and survival of resident cDCs and compromised T cell immunity. Trajectory analysis of human LN scRNA-seq data revealed expression similarities to murine FRCs, with GREM1+ cells marking the endpoint of both trajectories. These findings illuminate a new Grem1+ fibroblastic niche in LNs that functions to maintain the homeostasis of lymphoid tissue-resident cDCs.


Subject(s)
Dendritic Cells, Follicular/immunology , Fibroblasts/immunology , Lymph Nodes/immunology , Stromal Cells/immunology , Aged , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Proliferation/genetics , Cell Survival/genetics , Cell Survival/immunology , Dendritic Cells, Follicular/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation/immunology , Gene Knock-In Techniques , Humans , Immunity, Cellular/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymph Nodes/cytology , Male , Mice , Mice, Transgenic , RNA-Seq , Single-Cell Analysis , Stromal Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Nature ; 591(7851): 652-658, 2021 03.
Article in English | MEDLINE | ID: mdl-33588426

ABSTRACT

Limiting metabolic competition in the tumour microenvironment may increase the effectiveness of immunotherapy. Owing to its crucial role in the glucose metabolism of activated T cells, CD28 signalling has been proposed as a metabolic biosensor of T cells1. By contrast, the engagement of CTLA-4 has been shown to downregulate T cell glycolysis1. Here we investigate the effect of CTLA-4 blockade on the metabolic fitness of intra-tumour T cells in relation to the glycolytic capacity of tumour cells. We found that CTLA-4 blockade promotes metabolic fitness and the infiltration of immune cells, especially in glycolysis-low tumours. Accordingly, treatment with anti-CTLA-4 antibodies improved the therapeutic outcomes of mice bearing glycolysis-defective tumours. Notably, tumour-specific CD8+ T cell responses correlated with phenotypic and functional destabilization of tumour-infiltrating regulatory T (Treg) cells towards IFNγ- and TNF-producing cells in glycolysis-defective tumours. By mimicking the highly and poorly glycolytic tumour microenvironments in vitro, we show that the effect of CTLA-4 blockade on the destabilization of Treg cells is dependent on Treg cell glycolysis and CD28 signalling. These findings indicate that decreasing tumour competition for glucose may facilitate the therapeutic activity of CTLA-4 blockade, thus supporting its combination with inhibitors of tumour glycolysis. Moreover, these results reveal a mechanism by which anti-CTLA-4 treatment interferes with Treg cell function in the presence of glucose.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Glycolysis , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
7.
Cell ; 182(2): 329-344.e19, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32589946

ABSTRACT

Cell surface receptors and their interactions play a central role in physiological and pathological signaling. Despite its clinical relevance, the immunoglobulin superfamily (IgSF) remains uncharacterized and underrepresented in databases. Here, we present a systematic extracellular protein map, the IgSF interactome. Using a high-throughput technology to interrogate most single transmembrane receptors for binding to 445 IgSF proteins, we identify over 500 interactions, 82% previously undocumented, and confirm more than 60 receptor-ligand pairs using orthogonal assays. Our study reveals a map of cell-type-specific interactions and the landscape of dysregulated receptor-ligand crosstalk in cancer, including selective loss of function for tumor-associated mutations. Furthermore, investigation of the IgSF interactome in a large cohort of cancer patients identifies interacting protein signatures associated with clinical outcome. The IgSF interactome represents an important resource to fuel biological discoveries and a framework for understanding the functional organization of the surfaceome during homeostasis and disease, ultimately informing therapeutic development.


Subject(s)
Immunoglobulins/metabolism , Neoplasms/pathology , Protein Interaction Maps , B7-H1 Antigen/metabolism , Carcinoembryonic Antigen/metabolism , Cell Communication , Cluster Analysis , Culture Media, Conditioned/chemistry , HEK293 Cells , Humans , Immunoglobulins/chemistry , Immunoglobulins/genetics , Ligands , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Cancer Discov ; 10(2): 232-253, 2020 02.
Article in English | MEDLINE | ID: mdl-31699795

ABSTRACT

With only a fraction of patients responding to cancer immunotherapy, a better understanding of the entire tumor microenvironment is needed. Using single-cell transcriptomics, we chart the fibroblastic landscape during pancreatic ductal adenocarcinoma (PDAC) progression in animal models. We identify a population of carcinoma-associated fibroblasts (CAF) that are programmed by TGFß and express the leucine-rich repeat containing 15 (LRRC15) protein. These LRRC15+ CAFs surround tumor islets and are absent from normal pancreatic tissue. The presence of LRRC15+ CAFs in human patients was confirmed in >80,000 single cells from 22 patients with PDAC as well as by using IHC on samples from 70 patients. Furthermore, immunotherapy clinical trials comprising more than 600 patients across six cancer types revealed elevated levels of the LRRC15+ CAF signature correlated with poor response to anti-PD-L1 therapy. This work has important implications for targeting nonimmune elements of the tumor microenvironment to boost responses of patients with cancer to immune checkpoint blockade therapy. SIGNIFICANCE: This study describes the single-cell landscape of CAFs in pancreatic cancer during in vivo tumor evolution. A TGFß-driven, LRRC15+ CAF lineage is associated with poor outcome in immunotherapy trial data comprising multiple solid-tumor entities and represents a target for combinatorial therapy.This article is highlighted in the In This Issue feature, p. 161.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm/genetics , Immune Checkpoint Inhibitors/pharmacology , Membrane Proteins/metabolism , Myofibroblasts/immunology , Pancreatic Neoplasms/drug therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Cell Lineage/genetics , Cell Lineage/immunology , Clinical Trials as Topic , Computational Biology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Mice , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , RNA-Seq , Single-Cell Analysis , Transforming Growth Factor beta/metabolism , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
9.
J Clin Endocrinol Metab ; 104(10): 4889-4899, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31237614

ABSTRACT

CONTEXT: Most papillary microcarcinomas (PMCs) are indolent and subclinical. However, as many as 10% can present with clinically significant nodal metastases. OBJECTIVE AND DESIGN: Characterization of the genomic and transcriptomic landscape of PMCs presenting with or without clinically important lymph node metastases. SUBJECTS AND SAMPLES: Formalin-fixed paraffin-embedded PMC samples from 40 patients with lateral neck nodal metastases (pN1b) and 71 patients with PMC with documented absence of nodal disease (pN0). OUTCOME MEASURES: To interrogate DNA alterations in 410 genes commonly mutated in cancer and test for differential gene expression using a custom NanoString panel of 248 genes selected primarily based on their association with tumor size and nodal disease in the papillary thyroid cancer TCGA project. RESULTS: The genomic landscapes of PMC with or without pN1b were similar. Mutations in TERT promoter (3%) and TP53 (1%) were exclusive to N1b cases. Transcriptomic analysis revealed differential expression of 43 genes in PMCs with pN1b compared with pN0. A random forest machine learning-based molecular classifier developed to predict regional lymph node metastasis demonstrated a negative predictive value of 0.98 and a positive predictive value of 0.72 at a prevalence of 10% pN1b disease. CONCLUSIONS: The genomic landscape of tumors with pN1b and pN0 disease was similar, whereas 43 genes selected primarily by mining the TCGA RNAseq data were differentially expressed. This bioinformatics-driven approach to the development of a custom transcriptomic assay provides a basis for a molecular classifier for pN1b risk stratification in PMC.


Subject(s)
Genomics/methods , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Adult , Aged , Case-Control Studies , Cohort Studies , Comparative Genomic Hybridization , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Neck , Transcriptome
11.
Cancer Cell ; 33(6): 1017-1032.e7, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894689

ABSTRACT

A significant proportion of cancer patients do not respond to immune checkpoint blockade. To better understand the molecular mechanisms underlying these treatments, we explored the role of CD4+Foxp3- T cells expressing PD-1 (4PD1hi) and observed that 4PD1hi accumulate intratumorally as a function of tumor burden. Interestingly, CTLA-4 blockade promotes intratumoral and peripheral 4PD1hi increases in a dose-dependent manner, while combination with PD-1 blockade mitigates this effect and improves anti-tumor activity. We found that lack of effective 4PD1hi reduction after anti-PD-1 correlates with poor prognosis. Mechanistically, we provide evidence that mouse and human circulating and intra-tumor 4PD1hi inhibit T cell functions in a PD-1/PD-L1 dependent fashion and resemble follicular helper T cell (TFH)-like cells. Accordingly, anti-CTLA-4 activity is improved in TFH deficient mice.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
12.
Nature ; 554(7693): 544-548, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29443960

ABSTRACT

Therapeutic antibodies that block the programmed death-1 (PD-1)-programmed death-ligand 1 (PD-L1) pathway can induce robust and durable responses in patients with various cancers, including metastatic urothelial cancer. However, these responses only occur in a subset of patients. Elucidating the determinants of response and resistance is key to improving outcomes and developing new treatment strategies. Here we examined tumours from a large cohort of patients with metastatic urothelial cancer who were treated with an anti-PD-L1 agent (atezolizumab) and identified major determinants of clinical outcome. Response to treatment was associated with CD8+ T-effector cell phenotype and, to an even greater extent, high neoantigen or tumour mutation burden. Lack of response was associated with a signature of transforming growth factor ß (TGFß) signalling in fibroblasts. This occurred particularly in patients with tumours, which showed exclusion of CD8+ T cells from the tumour parenchyma that were instead found in the fibroblast- and collagen-rich peritumoural stroma; a common phenotype among patients with metastatic urothelial cancer. Using a mouse model that recapitulates this immune-excluded phenotype, we found that therapeutic co-administration of TGFß-blocking and anti-PD-L1 antibodies reduced TGFß signalling in stromal cells, facilitated T-cell penetration into the centre of tumours, and provoked vigorous anti-tumour immunity and tumour regression. Integration of these three independent biological features provides the best basis for understanding patient outcome in this setting and suggests that TGFß shapes the tumour microenvironment to restrain anti-tumour immunity by restricting T-cell infiltration.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , Transforming Growth Factor beta/metabolism , Urologic Neoplasms/drug therapy , Urologic Neoplasms/immunology , Urothelium/pathology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibodies/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, Neoplasm/analysis , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Checkpoints/drug effects , Cohort Studies , Collagen/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Fibroblasts/metabolism , Humans , Immunotherapy , Mice , Mutation , Neoplasm Metastasis , Phenotype , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Treatment Outcome , Tumor Microenvironment/immunology , Urologic Neoplasms/genetics , Urologic Neoplasms/pathology , Urothelium/drug effects , Urothelium/immunology
13.
Nat Immunol ; 19(3): 246-254, 2018 03.
Article in English | MEDLINE | ID: mdl-29358708

ABSTRACT

Defective autophagy is linked to diseases such as rheumatoid arthritis, lupus and inflammatory bowel disease (IBD). However, the mechanisms by which autophagy limits inflammation remain poorly understood. Here we found that loss of the autophagy-related gene Atg16l1 promoted accumulation of the adaptor TRIF and downstream signaling in macrophages. Multiplex proteomic profiling identified SQSTM1 and Tax1BP1 as selective autophagy-related receptors that mediated the turnover of TRIF. Knockdown of Tax1bp1 increased production of the cytokines IFN-ß and IL-1ß. Mice lacking Atg16l1 in myeloid cells succumbed to lipopolysaccharide-mediated sepsis but enhanced their clearance of intestinal Salmonella typhimurium in an interferon receptor-dependent manner. Human macrophages with the Crohn's disease-associated Atg16l1 variant T300A exhibited more production of IFN-ß and IL-1ß. An elevated interferon-response gene signature was observed in patients with IBD who were resistant to treatment with an antibody to the cytokine TNF. These findings identify selective autophagy as a key regulator of signaling via the innate immune system.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Autophagy/immunology , Immunity, Innate/immunology , Inflammation/immunology , Animals , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/immunology , Crohn Disease/immunology , Female , Humans , Macrophages/immunology , Male , Mice , Mice, Transgenic , Signal Transduction/immunology
14.
Nature ; 551(7681): 512-516, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29132146

ABSTRACT

Pancreatic ductal adenocarcinoma is a lethal cancer with fewer than 7% of patients surviving past 5 years. T-cell immunity has been linked to the exceptional outcome of the few long-term survivors, yet the relevant antigens remain unknown. Here we use genetic, immunohistochemical and transcriptional immunoprofiling, computational biophysics, and functional assays to identify T-cell antigens in long-term survivors of pancreatic cancer. Using whole-exome sequencing and in silico neoantigen prediction, we found that tumours with both the highest neoantigen number and the most abundant CD8+ T-cell infiltrates, but neither alone, stratified patients with the longest survival. Investigating the specific neoantigen qualities promoting T-cell activation in long-term survivors, we discovered that these individuals were enriched in neoantigen qualities defined by a fitness model, and neoantigens in the tumour antigen MUC16 (also known as CA125). A neoantigen quality fitness model conferring greater immunogenicity to neoantigens with differential presentation and homology to infectious disease-derived peptides identified long-term survivors in two independent datasets, whereas a neoantigen quantity model ascribing greater immunogenicity to increasing neoantigen number alone did not. We detected intratumoural and lasting circulating T-cell reactivity to both high-quality and MUC16 neoantigens in long-term survivors of pancreatic cancer, including clones with specificity to both high-quality neoantigens and predicted cross-reactive microbial epitopes, consistent with neoantigen molecular mimicry. Notably, we observed selective loss of high-quality and MUC16 neoantigenic clones on metastatic progression, suggesting neoantigen immunoediting. Our results identify neoantigens with unique qualities as T-cell targets in pancreatic ductal adenocarcinoma. More broadly, we identify neoantigen quality as a biomarker for immunogenic tumours that may guide the application of immunotherapies.


Subject(s)
Antigens, Neoplasm/immunology , Bacterial Proteins/immunology , Cancer Survivors , Cross Reactions/immunology , Pancreatic Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenocarcinoma/blood , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Antigens, Neoplasm/genetics , Bacterial Proteins/blood , Bacterial Proteins/genetics , CA-125 Antigen/genetics , CA-125 Antigen/immunology , Computer Simulation , Cross Reactions/genetics , Humans , Immunotherapy , Membrane Proteins/genetics , Membrane Proteins/immunology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/genetics , Prognosis , Survival Analysis , T-Lymphocytes, Cytotoxic/cytology , Exome Sequencing
16.
Genome Biol ; 17(1): 231, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27855702

ABSTRACT

BACKGROUND: Tumor-infiltrating immune cells have been linked to prognosis and response to immunotherapy; however, the levels of distinct immune cell subsets and the signals that draw them into a tumor, such as the expression of antigen presenting machinery genes, remain poorly characterized. Here, we employ a gene expression-based computational method to profile the infiltration levels of 24 immune cell populations in 19 cancer types. RESULTS: We compare cancer types using an immune infiltration score and a T cell infiltration score and find that clear cell renal cell carcinoma (ccRCC) is among the highest for both scores. Using immune infiltration profiles as well as transcriptomic and proteomic datasets, we characterize three groups of ccRCC tumors: T cell enriched, heterogeneously infiltrated, and non-infiltrated. We observe that the immunogenicity of ccRCC tumors cannot be explained by mutation load or neo-antigen load, but is highly correlated with MHC class I antigen presenting machinery expression (APM). We explore the prognostic value of distinct T cell subsets and show in two cohorts that Th17 cells and CD8+ T/Treg ratio are associated with improved survival, whereas Th2 cells and Tregs are associated with negative outcomes. Investigation of the association of immune infiltration patterns with the subclonal architecture of tumors shows that both APM and T cell levels are negatively associated with subclone number. CONCLUSIONS: Our analysis sheds light on the immune infiltration patterns of 19 human cancers and unravels mRNA signatures with prognostic utility and immunotherapeutic biomarker potential in ccRCC.


Subject(s)
Carcinoma, Renal Cell/immunology , Immunotherapy , Neoplasm Proteins/biosynthesis , Tumor Microenvironment/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Computer Simulation , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Nucleotide Motifs/genetics , Prognosis , Proteomics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Microenvironment/immunology
17.
JCI Insight ; 1(17): e89829, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27777979

ABSTRACT

Recent clinical trials have demonstrated a clear survival advantage in advanced head and neck squamous cell carcinoma (HNSCC) patients treated with immune checkpoint blockade. These emerging results reveal that HNSCC is one of the most promising frontiers for immunotherapy research. However, further progress in head and neck immuno-oncology will require a detailed understanding of the immune infiltrative landscape found in these tumors. We leveraged transcriptome data from 280 tumors profiled by The Cancer Genome Atlas (TCGA) to comprehensively characterize the immune landscape of HNSCC in order to develop a rationale for immunotherapeutic strategies in HNSCC and guide clinical investigation. We find that both HPV+ and HPV- HNSCC tumors are among the most highly immune-infiltrated cancer types. Strikingly, HNSCC had the highest median Treg/CD8+ T cell ratio and the highest levels of CD56dim NK cell infiltration, in our pan-cancer analysis of the most immune-infiltrated tumors. CD8+ T cell infiltration and CD56dim NK cell infiltration each correlated with superior survival in HNSCC. Tumors harboring genetic smoking signatures had lower immune infiltration and were associated with poorer survival, suggesting these patients may benefit from immune agonist therapy. These findings illuminate the immune landscape of HPV+ and HPV- HNSCC. Additionally, this landscape provides a potentially novel rationale for investigation of agents targeting modulators of Tregs (e.g., CTLA-4, GITR, ICOS, IDO, and VEGFA) and NK cells (e.g., KIR, TIGIT, and 4-1BB) as adjuncts to anti-PD-1 in the treatment of advanced HNSCC.


Subject(s)
Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , Immunotherapy , CD8-Positive T-Lymphocytes/cytology , Head and Neck Neoplasms/genetics , Humans , Killer Cells, Natural/cytology , Papillomaviridae , Smoking , T-Lymphocytes, Regulatory/cytology , Transcriptome , Tumor Microenvironment
18.
Cell Rep ; 14(10): 2476-89, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947078

ABSTRACT

On the basis of multidimensional and comprehensive molecular characterization (including DNA methalylation and copy number, RNA, and protein expression), we classified 894 renal cell carcinomas (RCCs) of various histologic types into nine major genomic subtypes. Site of origin within the nephron was one major determinant in the classification, reflecting differences among clear cell, chromophobe, and papillary RCC. Widespread molecular changes associated with TFE3 gene fusion or chromatin modifier genes were present within a specific subtype and spanned multiple subtypes. Differences in patient survival and in alteration of specific pathways (including hypoxia, metabolism, MAP kinase, NRF2-ARE, Hippo, immune checkpoint, and PI3K/AKT/mTOR) could further distinguish the subtypes. Immune checkpoint markers and molecular signatures of T cell infiltrates were both highest in the subtype associated with aggressive clear cell RCC. Differences between the genomic subtypes suggest that therapeutic strategies could be tailored to each RCC disease subset.


Subject(s)
Carcinoma, Renal Cell/pathology , Genomics , Kidney Neoplasms/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Chromatin/metabolism , Gene Expression Profiling , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , MicroRNAs/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction/genetics , Survival Rate , TOR Serine-Threonine Kinases/metabolism
19.
PLoS Comput Biol ; 12(2): e1004765, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26928298

ABSTRACT

Protein expression and post-translational modification levels are tightly regulated in neoplastic cells to maintain cellular processes known as 'cancer hallmarks'. The first Pan-Cancer initiative of The Cancer Genome Atlas (TCGA) Research Network has aggregated protein expression profiles for 3,467 patient samples from 11 tumor types using the antibody based reverse phase protein array (RPPA) technology. The resultant proteomic data can be utilized to computationally infer protein-protein interaction (PPI) networks and to study the commonalities and differences across tumor types. In this study, we compare the performance of 13 established network inference methods in their capacity to retrieve the curated Pathway Commons interactions from RPPA data. We observe that no single method has the best performance in all tumor types, but a group of six methods, including diverse techniques such as correlation, mutual information, and regression, consistently rank highly among the tested methods. We utilize the high performing methods to obtain a consensus network; and identify four robust and densely connected modules that reveal biological processes as well as suggest antibody-related technical biases. Mapping the consensus network interactions to Reactome gene lists confirms the pan-cancer importance of signal transduction pathways, innate and adaptive immune signaling, cell cycle, metabolism, and DNA repair; and also suggests several biological processes that may be specific to a subset of tumor types. Our results illustrate the utility of the RPPA platform as a tool to study proteomic networks in cancer.


Subject(s)
Neoplasm Proteins/metabolism , Neoplasms/metabolism , Protein Interaction Maps/physiology , Proteomics/methods , Software , Cluster Analysis , Databases, Protein , Gene Expression Profiling , Humans , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Neoplasms/genetics , Principal Component Analysis
20.
Elife ; 52016 Feb 22.
Article in English | MEDLINE | ID: mdl-26901439

ABSTRACT

Mutations, deletions, and changes in copy number of mitochondrial DNA (mtDNA), are observed throughout cancers. Here, we survey mtDNA copy number variation across 22 tumor types profiled by The Cancer Genome Atlas project. We observe a tendency for some cancers, especially of the bladder, breast, and kidney, to be depleted of mtDNA, relative to matched normal tissue. Analysis of genetic context reveals an association between incidence of several somatic alterations, including IDH1 mutations in gliomas, and mtDNA content. In some but not all cancer types, mtDNA content is correlated with the expression of respiratory genes, and anti-correlated to the expression of immune response and cell-cycle genes. In tandem with immunohistochemical evidence, we find that some tumors may compensate for mtDNA depletion to sustain levels of respiratory proteins. Our results highlight the extent of mtDNA copy number variation in tumors and point to related therapeutic opportunities.


Subject(s)
DNA Copy Number Variations , DNA, Mitochondrial/genetics , Neoplasms/pathology , Humans , Immunohistochemistry , Pathology, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL
...