Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
bioRxiv ; 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38260392

ABSTRACT

Neuroblastoma is a pediatric cancer arising from the developing sympathoadrenal lineage with complex inter- and intra-tumoral heterogeneity. To chart this complexity, we generated a comprehensive cell atlas of 55 neuroblastoma patient tumors, collected from two pediatric cancer institutions, spanning a range of clinical, genetic, and histologic features. Our atlas combines single-cell/nucleus RNA-seq (sc/scRNA-seq), bulk RNA-seq, whole exome sequencing, DNA methylation profiling, spatial transcriptomics, and two spatial proteomic methods. Sc/snRNA-seq revealed three malignant cell states with features of sympathoadrenal lineage development. All of the neuroblastomas had malignant cells that resembled sympathoblasts and the more differentiated adrenergic cells. A subset of tumors had malignant cells in a mesenchymal cell state with molecular features of Schwann cell precursors. DNA methylation profiles defined four groupings of patients, which differ in the degree of malignant cell heterogeneity and clinical outcomes. Using spatial proteomics, we found that neuroblastomas are spatially compartmentalized, with malignant tumor cells sequestered away from immune cells. Finally, we identify spatially restricted signaling patterns in immune cells from spatial transcriptomics. To facilitate the visualization and analysis of our atlas as a resource for further research in neuroblastoma, single cell, and spatial-omics, all data are shared through the Human Tumor Atlas Network Data Commons at www.humantumoratlas.org.

2.
bioRxiv ; 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-37425718

ABSTRACT

TP53 is the most frequently mutated gene across many cancers and is associated with shorter survival in lung adenocarcinoma (LUAD). To define how TP53 mutations affect the LUAD tumor microenvironment (TME), we constructed a multi-omic cellular and spatial tumor atlas of 23 treatment-naïve human lung tumors. We found that TP53 -mutant ( TP53 mut ) malignant cells lose alveolar identity and upregulate highly proliferative and entropic gene expression programs consistently across resectable LUAD patient tumors, genetically engineered mouse models, and cell lines harboring a wide spectrum of TP53 mutations. We further identified a multicellular tumor niche composed of SPP1 + macrophages and collagen-expressing fibroblasts that coincides with hypoxic, pro-metastatic expression programs in TP53 mut tumors. Spatially correlated angiostatic and immune checkpoint interactions, including CD274 - PDCD1 and PVR - TIGIT , are also enriched in TP53 mut LUAD tumors, which may influence response to checkpoint blockade therapy. Our methodology can be further applied to investigate mutation-specific TME changes in other cancers.

3.
Sci Adv ; 9(39): eadd9668, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37756410

ABSTRACT

Neuroendocrine tumors (NETs) are rare cancers that most often arise in the gastrointestinal tract and pancreas. The fundamental mechanisms driving gastroenteropancreatic (GEP)-NET growth remain incompletely elucidated; however, the heterogeneous clinical behavior of GEP-NETs suggests that both cellular lineage dynamics and tumor microenvironment influence tumor pathophysiology. Here, we investigated the single-cell transcriptomes of tumor and immune cells from patients with gastroenteropancreatic NETs. Malignant GEP-NET cells expressed genes and regulons associated with normal, gastrointestinal endocrine cell differentiation, and fate determination stages. Tumor and lymphoid compartments sparsely expressed immunosuppressive targets commonly investigated in clinical trials, such as the programmed cell death protein-1/programmed death ligand-1 axis. However, infiltrating myeloid cell types within both primary and metastatic GEP-NETs were enriched for genes encoding other immune checkpoints, including VSIR (VISTA), HAVCR2 (TIM3), LGALS9 (Gal-9), and SIGLEC10. Our findings highlight the transcriptomic heterogeneity that distinguishes the cellular landscapes of GEP-NET anatomic subtypes and reveal potential avenues for future precision medicine therapeutics.


Subject(s)
Intestinal Neoplasms , Neuroendocrine Tumors , Pancreatic Neoplasms , Stomach Neoplasms , Humans , Neuroendocrine Tumors/genetics , Intestinal Neoplasms/genetics , Stomach Neoplasms/genetics , Pancreatic Neoplasms/genetics , Tumor Microenvironment/genetics
4.
Cell ; 184(25): 6119-6137.e26, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34890551

ABSTRACT

Prognostically relevant RNA expression states exist in pancreatic ductal adenocarcinoma (PDAC), but our understanding of their drivers, stability, and relationship to therapeutic response is limited. To examine these attributes systematically, we profiled metastatic biopsies and matched organoid models at single-cell resolution. In vivo, we identify a new intermediate PDAC transcriptional cell state and uncover distinct site- and state-specific tumor microenvironments (TMEs). Benchmarking models against this reference map, we reveal strong culture-specific biases in cancer cell transcriptional state representation driven by altered TME signals. We restore expression state heterogeneity by adding back in vivo-relevant factors and show plasticity in culture models. Further, we prove that non-genetic modulation of cell state can strongly influence drug responses, uncovering state-specific vulnerabilities. This work provides a broadly applicable framework for aligning cell states across in vivo and ex vivo settings, identifying drivers of transcriptional plasticity and manipulating cell state to target associated vulnerabilities.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Tumor Microenvironment , Adult , Aged , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Single-Cell Analysis
5.
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
6.
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
7.
Nat Genet ; 53(3): 332-341, 2021 03.
Article in English | MEDLINE | ID: mdl-33649592

ABSTRACT

Resistance to immune checkpoint inhibitors (ICIs) is a key challenge in cancer therapy. To elucidate underlying mechanisms, we developed Perturb-CITE-sequencing (Perturb-CITE-seq), enabling pooled clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 perturbations with single-cell transcriptome and protein readouts. In patient-derived melanoma cells and autologous tumor-infiltrating lymphocyte (TIL) co-cultures, we profiled transcriptomes and 20 proteins in ~218,000 cells under ~750 perturbations associated with cancer cell-intrinsic ICI resistance (ICR). We recover known mechanisms of resistance, including defects in the interferon-γ (IFN-γ)-JAK/STAT and antigen-presentation pathways in RNA, protein and perturbation space, and new ones, including loss/downregulation of CD58. Loss of CD58 conferred immune evasion in multiple co-culture models and was downregulated in tumors of melanoma patients with ICR. CD58 protein expression was not induced by IFN-γ signaling, and CD58 loss conferred immune evasion without compromising major histocompatibility complex (MHC) expression, suggesting that it acts orthogonally to known mechanisms of ICR. This work provides a framework for the deciphering of complex mechanisms by large-scale perturbation screens with multimodal, single-cell readouts, and discovers potentially clinically relevant mechanisms of immune evasion.


Subject(s)
CD58 Antigens/immunology , Drug Resistance, Neoplasm/immunology , Melanoma/pathology , Single-Cell Analysis/methods , Tumor Escape , CD58 Antigens/genetics , CD58 Antigens/metabolism , CRISPR-Cas Systems , Coculture Techniques , Computational Biology/methods , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epitopes/genetics , Gene Knockout Techniques , Humans , Immune Checkpoint Inhibitors/pharmacology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/drug therapy , Melanoma/immunology , Sequence Analysis, RNA , Tumor Escape/genetics
8.
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
10.
Nat Med ; 26(5): 792-802, 2020 05.
Article in English | MEDLINE | ID: mdl-32405060

ABSTRACT

Single-cell genomics is essential to chart tumor ecosystems. Although single-cell RNA-Seq (scRNA-Seq) profiles RNA from cells dissociated from fresh tumors, single-nucleus RNA-Seq (snRNA-Seq) is needed to profile frozen or hard-to-dissociate tumors. Each requires customization to different tissue and tumor types, posing a barrier to adoption. Here, we have developed a systematic toolbox for profiling fresh and frozen clinical tumor samples using scRNA-Seq and snRNA-Seq, respectively. We analyzed 216,490 cells and nuclei from 40 samples across 23 specimens spanning eight tumor types of varying tissue and sample characteristics. We evaluated protocols by cell and nucleus quality, recovery rate and cellular composition. scRNA-Seq and snRNA-Seq from matched samples recovered the same cell types, but at different proportions. Our work provides guidance for studies in a broad range of tumors, including criteria for testing and selecting methods from the toolbox for other tumors, thus paving the way for charting tumor atlases.


Subject(s)
Algorithms , Cell Nucleus/genetics , Genomics/methods , Neoplasms/genetics , RNA-Seq/methods , Single-Cell Analysis/methods , Adult , Animals , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Child , Computational Biology/methods , Female , Freezing , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Knockout , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Sequence Analysis, RNA/methods , Tumor Cells, Cultured , Exome Sequencing/methods
11.
Cancer Immunol Res ; 7(8): 1230-1236, 2019 08.
Article in English | MEDLINE | ID: mdl-31217164

ABSTRACT

Immunotherapy with checkpoint inhibitors, such as the programmed death-1 (PD-1) antibodies pembrolizumab and nivolumab, are effective in a variety of tumors, yet not all patients respond. Tumor microsatellite instability-high (MSI-H) has emerged as a biomarker of response to checkpoint blockade, leading to the tissue agnostic approval of pembrolizumab in MSI-H cancers. Here we describe a patient with MSI-H colorectal cancer that was treated with this immune checkpoint inhibitor and exhibited progression of disease. We examined this intrinsic resistance through genomic, transcriptional, and pathologic characterization of the patient's tumor and the associated immune microenvironment. The tumor had typical MSI-H molecular features, including a high neoantigen load. We also identified biallelic loss of the gene for ß2-microglobulin (B2M), whose product is critical for antigen presentation. Immune infiltration deconvolution analysis of bulk transcriptome data from this anti-PD-1-resistant tumor and hundreds of other colorectal cancer specimens revealed a high natural killer cell and M2 macrophage infiltration in the patient's cancer. This was confirmed by single-cell transcriptome analysis and multiplex immunofluorescence. Our study provides insight into resistance in MSI-H tumors and suggests immunotherapeutic strategies in additional genomic contexts of colorectal cancer.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Colorectal Neoplasms/genetics , DNA Mismatch Repair , Drug Resistance, Neoplasm/genetics , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , DNA Copy Number Variations , DNA Methylation , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Mutation , Single-Cell Analysis , Tomography, X-Ray Computed
12.
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
13.
Nat Commun ; 9(1): 2068, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802342

ABSTRACT

Transient activation of Src oncoprotein in non-transformed, breast epithelial cells can initiate an epigenetic switch to the stably transformed state via a positive feedback loop that involves the inflammatory transcription factors STAT3 and NF-κB. Here, we develop an experimental and computational pipeline that includes 1) a Bayesian network model (AccessTF) that accurately predicts protein-bound DNA sequence motifs based on chromatin accessibility, and 2) a scoring system (TFScore) that rank-orders transcription factors as candidates for being important for a biological process. Genetic experiments validate TFScore and suggest that more than 40 transcription factors contribute to the oncogenic state in this model. Interestingly, individual depletion of several of these factors results in similar transcriptional profiles, indicating that a complex and interconnected transcriptional network promotes a stable oncogenic state. The combined experimental and computational pipeline represents a general approach to comprehensively identify transcriptional regulators important for a biological process.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Transcription Factors/metabolism , Breast/cytology , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Regulatory Networks/genetics , Genome, Human/genetics , Humans , Models, Genetic , Transcription Factors/genetics
14.
Cancer Discov ; 8(2): 196-215, 2018 02.
Article in English | MEDLINE | ID: mdl-29101162

ABSTRACT

Ex vivo systems that incorporate features of the tumor microenvironment and model the dynamic response to immune checkpoint blockade (ICB) may facilitate efforts in precision immuno-oncology and the development of effective combination therapies. Here, we demonstrate the ability to interrogate ex vivo response to ICB using murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). MDOTS/PDOTS isolated from mouse and human tumors retain autologous lymphoid and myeloid cell populations and respond to ICB in short-term three-dimensional microfluidic culture. Response and resistance to ICB was recapitulated using MDOTS derived from established immunocompetent mouse tumor models. MDOTS profiling demonstrated that TBK1/IKKε inhibition enhanced response to PD-1 blockade, which effectively predicted tumor response in vivo Systematic profiling of secreted cytokines in PDOTS captured key features associated with response and resistance to PD-1 blockade. Thus, MDOTS/PDOTS profiling represents a novel platform to evaluate ICB using established murine models as well as clinically relevant patient specimens.Significance: Resistance to PD-1 blockade remains a challenge for many patients, and biomarkers to guide treatment are lacking. Here, we demonstrate feasibility of ex vivo profiling of PD-1 blockade to interrogate the tumor immune microenvironment, develop therapeutic combinations, and facilitate precision immuno-oncology efforts. Cancer Discov; 8(2); 196-215. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Deng et al., p. 216This article is highlighted in the In This Issue feature, p. 127.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Cell Culture Techniques , Cell Line, Tumor , Cytokines/metabolism , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Immunohistochemistry , Immunophenotyping , Mice , Microfluidic Analytical Techniques , Programmed Cell Death 1 Receptor/metabolism , Spheroids, Cellular , Time-Lapse Imaging , Tumor Cells, Cultured
16.
Genes Dev ; 31(7): 674-687, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28446596

ABSTRACT

MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression critical for organismal viability. Changes in miRNA activity are common in cancer, but how these changes relate to subsequent alterations in transcription and the process of tumorigenesis is not well understood. Here, we report a deep transcriptional, oncogenic network regulated by miRNAs. We present analysis of the gene expression and phenotypic changes associated with global miRNA restoration in miRNA-deficient fibroblasts. This analysis uncovers a miRNA-repressed network containing oncofetal genes Imp1, Imp2, and Imp3 (Imp1-3) that is up-regulated primarily transcriptionally >100-fold upon Dicer loss and is resistant to resilencing by complete restoration of miRNA activity. This Dicer-resistant epigenetic switch confers tumorigenicity to these cells. Let-7 targets Imp1-3 are required for this tumorigenicity and feed back to reinforce and sustain expression of the oncogenic network. Together, these Dicer-resistant genes constitute an mRNA expression signature that is present in numerous human cancers and is associated with poor survival.


Subject(s)
Antigens, Neoplasm/genetics , Cell Transformation, Neoplastic/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/physiology , MicroRNAs/genetics , Ribonuclease III/genetics , Ribonuclease III/physiology , Animals , Antigens, Neoplasm/metabolism , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Knockout , Oncogenes , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcriptional Activation
17.
Mol Syst Biol ; 13(1): 905, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28069687

ABSTRACT

Treatment of BRAF-mutant melanomas with MAP kinase pathway inhibitors is paradigmatic of the promise of precision cancer therapy but also highlights problems with drug resistance that limit patient benefit. We use live-cell imaging, single-cell analysis, and molecular profiling to show that exposure of tumor cells to RAF/MEK inhibitors elicits a heterogeneous response in which some cells die, some arrest, and the remainder adapt to drug. Drug-adapted cells up-regulate markers of the neural crest (e.g., NGFR), a melanocyte precursor, and grow slowly. This phenotype is transiently stable, reverting to the drug-naïve state within 9 days of drug withdrawal. Transcriptional profiling of cell lines and human tumors implicates a c-Jun/ECM/FAK/Src cascade in de-differentiation in about one-third of cell lines studied; drug-induced changes in c-Jun and NGFR levels are also observed in xenograft and human tumors. Drugs targeting the c-Jun/ECM/FAK/Src cascade as well as BET bromodomain inhibitors increase the maximum effect (Emax) of RAF/MEK kinase inhibitors by promoting cell killing. Thus, analysis of reversible drug resistance at a single-cell level identifies signaling pathways and inhibitory drugs missed by assays that focus on cell populations.


Subject(s)
Indoles/administration & dosage , MAP Kinase Signaling System/drug effects , Melanoma/genetics , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Receptors, Nerve Growth Factor/genetics , Sulfonamides/administration & dosage , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Melanoma/drug therapy , Mice , Mutation , Single-Cell Analysis , Sulfonamides/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays
18.
Curr Protoc Mol Biol ; 116: 28.8.1-28.8.12, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27723082

ABSTRACT

For the last five decades, measuring the ability of cells to grow in soft agar has served as the gold standard assay for in vitro cellular transformation. Nevertheless, the soft agar colony formation assay is time consuming and ill-suited for high-throughput screens. This unit describes an equally qualitative and quantitative assay known as growth in low attachment or GILA. The GILA assay is suitable for high-throughput pharmacological or genetic screens and allows the simultaneous examination of multiple cell lines and experimental perturbations. GILA conditions are specific and relevant to the transformed state because they depend on a property of cancer cells that is not shared by non-transformed cells. The GILA assay enables ex vivo drug sensitivity testing of patient-derived tumor cells to define precise treatments for individual patients. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Drug Screening Assays, Antitumor/methods , Genetic Testing/methods , High-Throughput Screening Assays/methods , Agar/chemistry , Cell Adhesion , Cell Culture Techniques/methods , Cell Separation/methods , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Cells, Cultured
19.
Science ; 352(6282): 189-96, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-27124452

ABSTRACT

To explore the distinct genotypic and phenotypic states of melanoma tumors, we applied single-cell RNA sequencing (RNA-seq) to 4645 single cells isolated from 19 patients, profiling malignant, immune, stromal, and endothelial cells. Malignant cells within the same tumor displayed transcriptional heterogeneity associated with the cell cycle, spatial context, and a drug-resistance program. In particular, all tumors harbored malignant cells from two distinct transcriptional cell states, such that tumors characterized by high levels of the MITF transcription factor also contained cells with low MITF and elevated levels of the AXL kinase. Single-cell analyses suggested distinct tumor microenvironmental patterns, including cell-to-cell interactions. Analysis of tumor-infiltrating T cells revealed exhaustion programs, their connection to T cell activation and clonal expansion, and their variability across patients. Overall, we begin to unravel the cellular ecosystem of tumors and how single-cell genomics offers insights with implications for both targeted and immune therapies.


Subject(s)
Melanoma/genetics , Melanoma/secondary , Skin Neoplasms/pathology , Tumor Microenvironment , Base Sequence , Cell Communication , Cell Cycle , Drug Resistance, Neoplasm/genetics , Endothelial Cells/pathology , Genomics , Humans , Immunotherapy , Lymphocyte Activation , Melanoma/therapy , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Metastasis , RNA/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Stromal Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcriptome
20.
Proc Natl Acad Sci U S A ; 112(18): 5708-13, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25902495

ABSTRACT

Colony formation in soft agar is the gold-standard assay for cellular transformation in vitro, but it is unsuited for high-throughput screening. Here, we describe an assay for cellular transformation that involves growth in low attachment (GILA) conditions and is strongly correlated with the soft-agar assay. Using GILA, we describe high-throughput screens for drugs and genes that selectively inhibit or increase transformation, but not proliferation. Such molecules are unlikely to be found through conventional drug screening, and they include kinase inhibitors and drugs for noncancer diseases. In addition to known oncogenes, the genetic screen identifies genes that contribute to cellular transformation. Lastly, we demonstrate the ability of Food and Drug Administration-approved noncancer drugs to selectively kill ovarian cancer cells derived from patients with chemotherapy-resistant disease, suggesting this approach may provide useful information for personalized cancer treatment.


Subject(s)
Agar/chemistry , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Ovarian Neoplasms/drug therapy , Adenosine Triphosphate/chemistry , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Chemistry, Pharmaceutical , Drug Design , Female , Fibroblasts/pathology , Flow Cytometry , High-Throughput Screening Assays , Humans , Open Reading Frames , Ovarian Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...