Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Cell ; 184(18): 4734-4752.e20, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34450029

ABSTRACT

Immune responses to cancer are highly variable, with mismatch repair-deficient (MMRd) tumors exhibiting more anti-tumor immunity than mismatch repair-proficient (MMRp) tumors. To understand the rules governing these varied responses, we transcriptionally profiled 371,223 cells from colorectal tumors and adjacent normal tissues of 28 MMRp and 34 MMRd individuals. Analysis of 88 cell subsets and their 204 associated gene expression programs revealed extensive transcriptional and spatial remodeling across tumors. To discover hubs of interacting malignant and immune cells, we identified expression programs in different cell types that co-varied across tumors from affected individuals and used spatial profiling to localize coordinated programs. We discovered a myeloid cell-attracting hub at the tumor-luminal interface associated with tissue damage and an MMRd-enriched immune hub within the tumor, with activated T cells together with malignant and myeloid cells expressing T cell-attracting chemokines. By identifying interacting cellular programs, we reveal the logic underlying spatially organized immune-malignant cell networks.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Bone Morphogenetic Proteins/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Compartmentation , Cell Line, Tumor , Chemokines/metabolism , Cohort Studies , Colorectal Neoplasms/genetics , DNA Mismatch Repair/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunity , Inflammation/pathology , Monocytes/pathology , Myeloid Cells/pathology , Neutrophils/pathology , Stromal Cells/metabolism , T-Lymphocytes/metabolism , Transcription, Genetic
2.
Cancer Cell ; 39(5): 649-661.e5, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33711272

ABSTRACT

Immune checkpoint blockade (ICB) results in durable disease control in a subset of patients with advanced renal cell carcinoma (RCC), but mechanisms driving resistance are poorly understood. We characterize the single-cell transcriptomes of cancer and immune cells from metastatic RCC patients before or after ICB exposure. In responders, subsets of cytotoxic T cells express higher levels of co-inhibitory receptors and effector molecules. Macrophages from treated biopsies shift toward pro-inflammatory states in response to an interferon-rich microenvironment but also upregulate immunosuppressive markers. In cancer cells, we identify bifurcation into two subpopulations differing in angiogenic signaling and upregulation of immunosuppressive programs after ICB. Expression signatures for cancer cell subpopulations and immune evasion are associated with PBRM1 mutation and survival in primary and ICB-treated advanced RCC. Our findings demonstrate that ICB remodels the RCC microenvironment and modifies the interplay between cancer and immune cell populations critical for understanding response and resistance to ICB.


Subject(s)
Carcinoma, Renal Cell/therapy , Immunologic Factors/immunology , Immunotherapy , Kidney Neoplasms/therapy , Tumor Microenvironment/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , DNA-Binding Proteins/immunology , Humans , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Transcription Factors/immunology
3.
Nat Med ; 27(3): 426-433, 2021 03.
Article in English | MEDLINE | ID: mdl-33664492

ABSTRACT

Metastatic castration-resistant prostate cancer is typically lethal, exhibiting intrinsic or acquired resistance to second-generation androgen-targeting therapies and minimal response to immune checkpoint inhibitors1. Cellular programs driving resistance in both cancer and immune cells remain poorly understood. We present single-cell transcriptomes from 14 patients with advanced prostate cancer, spanning all common metastatic sites. Irrespective of treatment exposure, adenocarcinoma cells pervasively coexpressed multiple androgen receptor isoforms, including truncated isoforms hypothesized to mediate resistance to androgen-targeting therapies2,3. Resistance to enzalutamide was associated with cancer cell-intrinsic epithelial-mesenchymal transition and transforming growth factor-ß signaling. Small cell carcinoma cells exhibited divergent expression programs driven by transcriptional regulators promoting lineage plasticity and HOXB5, HOXB6 and NR1D2 (refs. 4-6). Additionally, a subset of patients had high expression of dysfunction markers on cytotoxic CD8+ T cells undergoing clonal expansion following enzalutamide treatment. Collectively, the transcriptional characterization of cancer and immune cells from human metastatic castration-resistant prostate cancer provides a basis for the development of therapeutic approaches complementing androgen signaling inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Prostatic Neoplasms, Castration-Resistant/therapy , Transcription, Genetic/drug effects , Biopsy , CD8-Positive T-Lymphocytes/immunology , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Male , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/metabolism
4.
Nat Med ; 26(8): 1271-1279, 2020 08.
Article in English | MEDLINE | ID: mdl-32572264

ABSTRACT

Malignant abdominal fluid (ascites) frequently develops in women with advanced high-grade serous ovarian cancer (HGSOC) and is associated with drug resistance and a poor prognosis1. To comprehensively characterize the HGSOC ascites ecosystem, we used single-cell RNA sequencing to profile ~11,000 cells from 22 ascites specimens from 11 patients with HGSOC. We found significant inter-patient variability in the composition and functional programs of ascites cells, including immunomodulatory fibroblast sub-populations and dichotomous macrophage populations. We found that the previously described immunoreactive and mesenchymal subtypes of HGSOC, which have prognostic implications, reflect the abundance of immune infiltrates and fibroblasts rather than distinct subsets of malignant cells2. Malignant cell variability was partly explained by heterogeneous copy number alteration patterns or expression of a stemness program. Malignant cells shared expression of inflammatory programs that were largely recapitulated in single-cell RNA sequencing of ~35,000 cells from additionally collected samples, including three ascites, two primary HGSOC tumors and three patient ascites-derived xenograft models. Inhibition of the JAK/STAT pathway, which was expressed in both malignant cells and cancer-associated fibroblasts, had potent anti-tumor activity in primary short-term cultures and patient-derived xenograft models. Our work contributes to resolving the HSGOC landscape3-5 and provides a resource for the development of novel therapeutic approaches.


Subject(s)
Ascites/genetics , Cystadenoma, Serous/genetics , Ovarian Neoplasms/genetics , Single-Cell Analysis , Ascites/pathology , Cell Line, Tumor , Cystadenoma, Serous/pathology , DNA Copy Number Variations/genetics , Drug Resistance, Neoplasm/genetics , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Janus Kinase 1/genetics , Neoplasm Grading , Neoplasm Proteins/genetics , Ovarian Neoplasms/pathology , Prognosis , STAT Transcription Factors/genetics , Sequence Analysis, RNA , Signal Transduction/genetics
5.
Cell ; 175(4): 984-997.e24, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388455

ABSTRACT

Immune checkpoint inhibitors (ICIs) produce durable responses in some melanoma patients, but many patients derive no clinical benefit, and the molecular underpinnings of such resistance remain elusive. Here, we leveraged single-cell RNA sequencing (scRNA-seq) from 33 melanoma tumors and computational analyses to interrogate malignant cell states that promote immune evasion. We identified a resistance program expressed by malignant cells that is associated with T cell exclusion and immune evasion. The program is expressed prior to immunotherapy, characterizes cold niches in situ, and predicts clinical responses to anti-PD-1 therapy in an independent cohort of 112 melanoma patients. CDK4/6-inhibition represses this program in individual malignant cells, induces senescence, and reduces melanoma tumor outgrowth in mouse models in vivo when given in combination with immunotherapy. Our study provides a high-resolution landscape of ICI-resistant cell states, identifies clinically predictive signatures, and suggests new therapeutic strategies to overcome immunotherapy resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Melanoma/immunology , Protein Kinase Inhibitors/therapeutic use , T-Lymphocytes/immunology , Tumor Escape , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Immunotherapy/methods , Male , Melanoma/drug therapy , Melanoma/therapy , Mice , Mice, Inbred C57BL , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology
6.
Nucleic Acids Res ; 43(22): 10655-72, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26578579

ABSTRACT

To understand how transposon landscapes (TLs) vary across animal genomes, we describe a new method called the Transposon Insertion and Depletion AnaLyzer (TIDAL) and a database of >300 TLs in Drosophila melanogaster (TIDAL-Fly). Our analysis reveals pervasive TL diversity across cell lines and fly strains, even for identically named sub-strains from different laboratories such as the ISO1 strain used for the reference genome sequence. On average, >500 novel insertions exist in every lab strain, inbred strains of the Drosophila Genetic Reference Panel (DGRP), and fly isolates in the Drosophila Genome Nexus (DGN). A minority (<25%) of transposon families comprise the majority (>70%) of TL diversity across fly strains. A sharp contrast between insertion and depletion patterns indicates that many transposons are unique to the ISO1 reference genome sequence. Although TL diversity from fly strains reaches asymptotic limits with increasing sequencing depth, rampant TL diversity causes unsaturated detection of TLs in pools of flies. Finally, we show novel transposon insertions negatively correlate with Piwi-interacting RNA (piRNA) levels for most transposon families, except for the highly-abundant roo retrotransposon. Our study provides a useful resource for Drosophila geneticists to understand how transposons create extensive genomic diversity in fly cell lines and strains.


Subject(s)
DNA Transposable Elements , Drosophila melanogaster/genetics , Genomics/methods , Retroelements , Animals , Cell Line , Databases, Nucleic Acid , Genetic Variation , Genome, Insect , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL