Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Cancer Discov ; 14(1): 36-48, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38047596

ABSTRACT

Cancer cells adapt and survive through the acquisition and selection of molecular modifications. This process defines cancer evolution. Building on a theoretical framework based on heritable genetic changes has provided insights into the mechanisms supporting cancer evolution. However, cancer hallmarks also emerge via heritable nongenetic mechanisms, including epigenetic and chromatin topological changes, and interactions between tumor cells and the tumor microenvironment. Recent findings on tumor evolutionary mechanisms draw a multifaceted picture where heterogeneous forces interact and influence each other while shaping tumor progression. A comprehensive characterization of the cancer evolutionary toolkit is required to improve personalized medicine and biomarker discovery. SIGNIFICANCE: Tumor evolution is fueled by multiple enabling mechanisms. Importantly, genetic instability, epigenetic reprogramming, and interactions with the tumor microenvironment are neither alternative nor independent evolutionary mechanisms. As demonstrated by findings highlighted in this perspective, experimental and theoretical approaches must account for multiple evolutionary mechanisms and their interactions to ultimately understand, predict, and steer tumor evolution.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Neoplasms/pathology , Epigenomics , Precision Medicine , Tumor Microenvironment/genetics
2.
Immunity ; 56(6): 1341-1358.e11, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37315536

ABSTRACT

Type 1 conventional dendritic cells (cDC1s) are critical for anti-cancer immunity. Protective anti-cancer immunity is thought to require cDC1s to sustain T cell responses within tumors, but it is poorly understood how this function is regulated and whether its subversion contributes to immune evasion. Here, we show that tumor-derived prostaglandin E2 (PGE2) programmed a dysfunctional state in intratumoral cDC1s, disabling their ability to locally orchestrate anti-cancer CD8+ T cell responses. Mechanistically, cAMP signaling downstream of the PGE2-receptors EP2 and EP4 was responsible for the programming of cDC1 dysfunction, which depended on the loss of the transcription factor IRF8. Blockade of the PGE2-EP2/EP4-cDC1 axis prevented cDC1 dysfunction in tumors, locally reinvigorated anti-cancer CD8+ T cell responses, and achieved cancer immune control. In human cDC1s, PGE2-induced dysfunction is conserved and associated with poor cancer patient prognosis. Our findings reveal a cDC1-dependent intratumoral checkpoint for anti-cancer immunity that is targeted by PGE2 for immune evasion.


Subject(s)
Dinoprostone , Neoplasms , Humans , Antibodies , CD8-Positive T-Lymphocytes , Dendritic Cells , Receptors, Prostaglandin E
3.
Cancer Discov ; 13(1): 70-84, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36213965

ABSTRACT

The skin is exposed to viral pathogens, but whether they contribute to the oncogenesis of skin cancers has not been systematically explored. Here we investigated 19 skin tumor types by analyzing off-target reads from commonly available next-generation sequencing data for viral pathogens. We identified human papillomavirus 42 (HPV42) in 96% (n = 45/47) of digital papillary adenocarcinoma (DPA), an aggressive cancer occurring on the fingers and toes. We show that HPV42, so far considered a nononcogenic, "low-risk" HPV, recapitulates the molecular hallmarks of oncogenic, "high-risk" HPVs. Using machine learning, we find that HPV-driven transformation elicits a germ cell-like transcriptional program conserved throughout all HPV-driven cancers (DPA, cervical carcinoma, and head and neck cancer). We further show that this germ cell-like transcriptional program, even when reduced to the top two genes (CDKN2A and SYCP2), serves as a fingerprint of oncogenic HPVs with implications for early detection, diagnosis, and therapy of all HPV-driven cancers. SIGNIFICANCE: We identify HPV42 as a uniform driver of DPA and add a new member to the short list of tumorigenic viruses in humans. We discover that all oncogenic HPVs evoke a germ cell-like transcriptional program with important implications for detecting, diagnosing, and treating all HPV-driven cancers. See related commentary by Starrett et al., p. 17. This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Adenocarcinoma, Clear Cell , Adenocarcinoma, Papillary , Bone Neoplasms , Breast Neoplasms , Papillomavirus Infections , Skin Neoplasms , Uterine Cervical Neoplasms , Female , Humans , Human Papillomavirus Viruses , Papillomavirus Infections/complications , Papillomaviridae/genetics , Germ Cells/pathology
4.
Sci Transl Med ; 14(655): eadd0887, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35895833

ABSTRACT

Functional interrogation of cancer and immune cells during tumor evolution reveals cancer drivers and informs mechanism-based combination therapies.


Subject(s)
Neoplasms , Combined Modality Therapy , Humans , Neoplasms/therapy
6.
Sci Transl Med ; 13(601)2021 07 07.
Article in English | MEDLINE | ID: mdl-34233950

ABSTRACT

Inflammation is a well-known driver of lung tumorigenesis. One strategy by which tumor cells escape tight homeostatic control is by decreasing the expression of the potent anti-inflammatory protein tumor necrosis factor alpha-induced protein 3 (TNFAIP3), also known as A20. We observed that tumor cell intrinsic loss of A20 markedly enhanced lung tumorigenesis and was associated with reduced CD8+ T cell-mediated immune surveillance in patients with lung cancer and in mouse models. In mice, we observed that this effect was completely dependent on increased cellular sensitivity to interferon-γ (IFN-γ) signaling by aberrant activation of TANK-binding kinase 1 (TBK1) and increased downstream expression and activation of signal transducer and activator of transcription 1 (STAT1). Interrupting this autocrine feed forward loop by knocking out IFN-α/ß receptor completely restored infiltration of cytotoxic T cells and rescued loss of A20 depending tumorigenesis. Downstream of STAT1, programmed death ligand 1 (PD-L1) was highly expressed in A20 knockout lung tumors. Accordingly, immune checkpoint blockade (ICB) treatment was highly efficient in mice harboring A20-deficient lung tumors. Furthermore, an A20 loss-of-function gene expression signature positively correlated with survival of melanoma patients treated with anti-programmed cell death protein 1. Together, we have identified A20 as a master immune checkpoint regulating the TBK1-STAT1-PD-L1 axis that may be exploited to improve ICB therapy in patients with lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Adenocarcinoma of Lung/genetics , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Down-Regulation , Humans , Interferon-gamma/metabolism , Lung Neoplasms/genetics , Mice , Signal Transduction
7.
Mol Imaging Biol ; 23(6): 874-893, 2021 12.
Article in English | MEDLINE | ID: mdl-34101107

ABSTRACT

Tumor vasculature and angiogenesis play a crucial role in tumor progression. Their visualization is therefore of utmost importance to the community. In this proof-of-principle study, we have established a novel cross-modality imaging (CMI) pipeline to characterize exactly the same murine tumors across scales and penetration depths, using orthotopic models of melanoma cancer. This allowed the acquisition of a comprehensive set of vascular parameters for a single tumor. The workflow visualizes capillaries at different length scales, puts them into the context of the overall tumor vessel network and allows quantification and comparison of vessel densities and morphologies by different modalities. The workflow adds information about hypoxia and blood flow rates. The CMI approach includes well-established technologies such as magnetic resonance imaging (MRI), positron emission tomography (PET), computed tomography (CT), and ultrasound (US), and modalities that are recent entrants into preclinical discovery such as optical coherence tomography (OCT) and high-resolution episcopic microscopy (HREM). This novel CMI platform establishes the feasibility of combining these technologies using an extensive image processing pipeline. Despite the challenges pertaining to the integration of microscopic and macroscopic data across spatial resolutions, we also established an open-source pipeline for the semi-automated co-registration of the diverse multiscale datasets, which enables truly correlative vascular imaging. Although focused on tumor vasculature, our CMI platform can be used to tackle a multitude of research questions in cancer biology.


Subject(s)
Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Animals , Feasibility Studies , Mice , Positron-Emission Tomography , Tomography, X-Ray Computed
8.
Nat Cancer ; 2(7): 693-708, 2021 07.
Article in English | MEDLINE | ID: mdl-35121945

ABSTRACT

How targeted therapies and immunotherapies shape tumors, and thereby influence subsequent therapeutic responses, is poorly understood. In the present study, we show, in melanoma patients and mouse models, that when tumors relapse after targeted therapy with MAPK pathway inhibitors, they are cross-resistant to immunotherapies, despite the different modes of action of these therapies. We find that cross-resistance is mediated by a cancer cell-instructed, immunosuppressive tumor microenvironment that lacks functional CD103+ dendritic cells, precluding an effective T cell response. Restoring the numbers and functionality of CD103+ dendritic cells can re-sensitize cross-resistant tumors to immunotherapy. Cross-resistance does not arise from selective pressure of an immune response during evolution of resistance, but from the MAPK pathway, which not only is reactivated, but also exhibits an increased transcriptional output that drives immune evasion. Our work provides mechanistic evidence for cross-resistance between two unrelated therapies, and a scientific rationale for treating patients with immunotherapy before they acquire resistance to targeted therapy.


Subject(s)
Melanoma , Tumor Microenvironment , Animals , Humans , Immune Evasion , Immunologic Factors/therapeutic use , Immunotherapy , Melanoma/drug therapy , Mice , Neoplasm Recurrence, Local , Protein Kinase Inhibitors/pharmacology
9.
Nat Biotechnol ; 39(2): 174-178, 2021 02.
Article in English | MEDLINE | ID: mdl-32719478

ABSTRACT

We developed a functional lineage tracing tool termed CaTCH (CRISPRa tracing of clones in heterogeneous cell populations). CaTCH combines precise clonal tracing of millions of cells with the ability to retrospectively isolate founding clones alive before and during selection, allowing functional experiments. Using CaTCH, we captured rare clones representing as little as 0.001% of a population and investigated the emergence of resistance to targeted melanoma therapy in vivo.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Separation , Clone Cells/metabolism , Genes, Reporter , Animals , Cell Line , Female , Humans , Melanoma/pathology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , raf Kinases/antagonists & inhibitors
10.
EMBO Mol Med ; 12(11): e12525, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33026191

ABSTRACT

Merkel cell carcinoma (MCC) is a highly aggressive, neuroendocrine skin cancer that lacks actionable mutations, which could be utilized for targeted therapies. Epigenetic regulators governing cell identity may represent unexplored therapeutic entry points. Here, we targeted epigenetic regulators in a pharmacological screen and discovered that the lysine-specific histone demethylase 1A (LSD1/KDM1A) is required for MCC growth in vitro and in vivo. We show that LSD1 inhibition in MCC disrupts the LSD1-CoREST complex leading to displacement and degradation of HMG20B (BRAF35), a poorly characterized complex member that is essential for MCC proliferation. Inhibition of LSD1 causes derepression of transcriptional master regulators of the neuronal lineage, activates a gene expression signature resembling normal Merkel cells, and induces cell cycle arrest and cell death. Our study unveils the importance of LSD1 for maintaining cellular plasticity and proliferation in MCC. There is also growing evidence that cancer cells exploit cellular plasticity and dedifferentiation programs to evade destruction by the immune system. The combination of LSD1 inhibitors with checkpoint inhibitors may thus represent a promising treatment strategy for MCC patients.


Subject(s)
Carcinoma, Merkel Cell , Skin Neoplasms , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/genetics , Cell Death , Cell Differentiation , Histone Demethylases/genetics , Humans , Skin Neoplasms/drug therapy
11.
Front Immunol ; 10: 2746, 2019.
Article in English | MEDLINE | ID: mdl-31849950

ABSTRACT

For decades, cancer was considered a disease driven by genetic mutations in tumor cells, therefore afflicting a single cell type. This simplified view was slowly replaced by the understanding that interactions between malignant cells and neighboring stromal and immune cells-the tumor microenvironment (TME)-profoundly shape cancer progression. This understanding paved the way for an entirely new form of therapy that targets the immune cell compartment, which has revolutionized the treatment of cancer. In particular, agents activating T lymphocytes have become a key focus of these therapies, as they can induce durable responses in several cancer types. However, T cell targeting agents only benefit a fraction of patients. Thus, it is crucial to identify the roles of other immune cell types in the TME and understand how they influence T cell function and/or whether they present valuable therapeutic targets themselves. In this review, we focus on the myeloid compartment of the TME, a heterogeneous mix of cell types with diverse effector functions. We describe how distinct myeloid cell types can act as enemies of cancer cells by inducing or enhancing an existing immune response, while others act as strong allies, supporting tumor cells in their malignant growth and establishing an immune evasive TME. Specifically, we focus on the role of myeloid cells in the response and resistance to immunotherapy, and how modulating their numbers and/or state could provide alternative therapeutic entry-points.


Subject(s)
Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Tumor Microenvironment , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunity , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/immunology , Macrophages/metabolism , Myeloid Cells/pathology , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Escape , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
12.
Science ; 360(6390): 800-805, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29622725

ABSTRACT

Defining direct targets of transcription factors and regulatory pathways is key to understanding their roles in physiology and disease. We combined SLAM-seq [thiol(SH)-linked alkylation for the metabolic sequencing of RNA], a method for direct quantification of newly synthesized messenger RNAs (mRNAs), with pharmacological and chemical-genetic perturbation in order to define regulatory functions of two transcriptional hubs in cancer, BRD4 and MYC, and to interrogate direct responses to BET bromodomain inhibitors (BETis). We found that BRD4 acts as general coactivator of RNA polymerase II-dependent transcription, which is broadly repressed upon high-dose BETi treatment. At doses triggering selective effects in leukemia, BETis deregulate a small set of hypersensitive targets including MYC. In contrast to BRD4, MYC primarily acts as a selective transcriptional activator controlling metabolic processes such as ribosome biogenesis and de novo purine synthesis. Our study establishes a simple and scalable strategy to identify direct transcriptional targets of any gene or pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Genes, Regulator , Leukemia, Myeloid/drug therapy , Nuclear Proteins/metabolism , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins , Dose-Response Relationship, Drug , Humans , Leukemia, Myeloid/genetics , Molecular Targeted Therapy , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Purines/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Ribosomes/metabolism , Sequence Analysis, RNA , Transcription Factors/genetics , Transcription, Genetic
13.
Oncotarget ; 9(17): 13565-13581, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29568378

ABSTRACT

Tripartite motif (TRIM) proteins have been shown to play important roles in cancer development and progression by modulating cell proliferation or resistance from cell death during non-homeostatic stress conditions found in tumor micro-environments. In this study, we set out to investigate the importance for cellular fitness of the virtually uncharacterized family member TRIM52. The human TRIM52 gene has arisen recently in evolution, making it unlikely that TRIM52 is required for basic cellular functions in normal cells. However, a recent genome-wide ablation screening study has suggested that TRIM52 may be essential for optimal proliferation or survival in certain genetic cancer backgrounds. Identifying genes which fit this concept of genetic context-dependent fitness in cancer cells is of interest as they are promising targets for tumor-specific therapy. We report here that TRIM52 ablation significantly diminished the proliferation of specific glioblastoma cell lines in cell culture and mouse xenografts by compromising their cell cycle progression in a p53-dependent manner. Together, our findings point to a non-redundant TRIM52 function that is required for optimal proliferation.

14.
Immunity ; 44(3): 698-711, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26982367

ABSTRACT

Microsatellite instability in colorectal cancer predicts favorable outcomes. However, the mechanistic relationship between microsatellite instability, tumor-infiltrating immune cells, Immunoscore, and their impact on patient survival remains to be elucidated. We found significant differences in mutational patterns, chromosomal instability, and gene expression that correlated with patient microsatellite instability status. A prominent immune gene expression was observed in microsatellite-instable (MSI) tumors, as well as in a subgroup of microsatellite-stable (MSS) tumors. MSI tumors had increased frameshift mutations, showed genetic evidence of immunoediting, had higher densities of Th1, effector-memory T cells, in situ proliferating T cells, and inhibitory PD1-PDL1 cells, had high Immunoscores, and were infiltrated with mutation-specific cytotoxic T cells. Multivariate analysis revealed that Immunoscore was superior to microsatellite instability in predicting patients' disease-specific recurrence and survival. These findings indicate that assessment of the immune status via Immunoscore provides a potent indicator of tumor recurrence beyond microsatellite-instability staging that could be an important guide for immunotherapy strategies.


Subject(s)
Colorectal Neoplasms/diagnosis , Immunoassay/methods , Pathology, Molecular/methods , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Aged , Aged, 80 and over , Cells, Cultured , Colorectal Neoplasms/mortality , Cytotoxicity Tests, Immunologic , DNA Mutational Analysis , Female , Frameshift Mutation/genetics , Humans , Immunologic Memory , Male , Microsatellite Instability , Microsatellite Repeats , Predictive Value of Tests , Prognosis , Survival Analysis , Transcriptome
15.
Sci Transl Med ; 8(327): 327ra26, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26912905

ABSTRACT

Although distant metastases account for most of the deaths in cancer patients, fundamental questions regarding mechanisms that promote or inhibit metastasis remain unanswered. We show the impact of mutations, genomic instability, lymphatic and blood vascularization, and the immune contexture of the tumor microenvironment on synchronous metastases in large cohorts of colorectal cancer patients. We observed large genetic heterogeneity among primary tumors, but no major differences in chromosomal instability or key cancer-associated mutations. Similar patterns of cancer-related gene expression levels were observed between patients. No cancer-associated genes or pathways were associated with M stage. Instead, mutations of FBXW7 were associated with the absence of metastasis and correlated with increased expression of T cell proliferation and antigen presentation functions. Analyzing the tumor microenvironment, we observed two hallmarks of the metastatic process: decreased presence of lymphatic vessels and reduced immune cytotoxicity. These events could be the initiating factors driving both synchronous and metachronous metastases. Our data demonstrate the protective impact of the Immunoscore, a cytotoxic immune signature, and increased marginal lymphatic vessels, against the generation of distant metastases, regardless of genomic instability.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Tumor Microenvironment/immunology , Blood Vessels/pathology , Cell Count , Cell Death , Colorectal Neoplasms/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Lymphatic System/pathology , Lymphocytes/metabolism , Mutation/genetics , Neoplasm Metastasis , Tumor Microenvironment/genetics
16.
Nature ; 529(7586): 298-306, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26791720

ABSTRACT

Metastasis is the main cause of death in people with cancer. To colonize distant organs, circulating tumour cells must overcome many obstacles through mechanisms that we are only now starting to understand. These include infiltrating distant tissue, evading immune defences, adapting to supportive niches, surviving as latent tumour-initiating seeds and eventually breaking out to replace the host tissue. They make metastasis a highly inefficient process. However, once metastases have been established, current treatments frequently fail to provide durable responses. An improved understanding of the mechanistic determinants of such colonization is needed to better prevent and treat metastatic cancer.


Subject(s)
Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Animals , Cell Proliferation , Cell Survival , Humans , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/immunology , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/immunology , Organ Specificity , Stem Cell Niche , Tumor Microenvironment/immunology
17.
Nature ; 526(7573): 453-7, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26444240

ABSTRACT

Activation of oncogenes by mechanisms other than genetic aberrations such as mutations, translocations, or amplifications is largely undefined. Here we report a novel isoform of the anaplastic lymphoma kinase (ALK) that is expressed in ∼11% of melanomas and sporadically in other human cancer types, but not in normal tissues. The novel ALK transcript initiates from a de novo alternative transcription initiation (ATI) site in ALK intron 19, and was termed ALK(ATI). In ALK(ATI)-expressing tumours, the ATI site is enriched for H3K4me3 and RNA polymerase II, chromatin marks characteristic of active transcription initiation sites. ALK(ATI) is expressed from both ALK alleles, and no recurrent genetic aberrations are found at the ALK locus, indicating that the transcriptional activation is independent of genetic aberrations at the ALK locus. The ALK(ATI) transcript encodes three proteins with molecular weights of 61.1, 60.8 and 58.7 kilodaltons, consisting primarily of the intracellular tyrosine kinase domain. ALK(ATI) stimulates multiple oncogenic signalling pathways, drives growth-factor-independent cell proliferation in vitro, and promotes tumorigenesis in vivo in mouse models. ALK inhibitors can suppress the kinase activity of ALK(ATI), suggesting that patients with ALK(ATI)-expressing tumours may benefit from ALK inhibitors. Our findings suggest a novel mechanism of oncogene activation in cancer through de novo alternative transcription initiation.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Neoplasms/enzymology , Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Transcription Initiation, Genetic , Alleles , Anaplastic Lymphoma Kinase , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Female , HEK293 Cells , Histones/chemistry , Histones/metabolism , Humans , Introns/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/chemistry , Isoenzymes/genetics , Lysine/metabolism , Methylation , Mice , Molecular Sequence Data , Molecular Weight , NIH 3T3 Cells , Neoplasms/drug therapy , Oncogenes/genetics , Protein Structure, Tertiary/genetics , RNA Polymerase II/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/chemistry , Signal Transduction
18.
Cancer Res ; 75(18): 3713-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26208905

ABSTRACT

Several experimental models faithfully recapitulate many important facets of human metastatic disease. Here, we have performed whole-exome sequencing in five widely used experimental metastasis models that were independently derived through in vivo selection from heterogeneous human cancer cell lines. In addition to providing an important characterization of these model systems, our study examines the genetic evolution of metastatic phenotypes. We found that in vivo selected highly metastatic cell populations showed little genetic divergence from the corresponding parental population. However, selection of genetic variations that preexisted in parental populations, including the well-established oncogenic mutations KRAS(G13D) and BRAF(G464V), was associated with increased metastatic capability. Conversely, expression of the wild-type BRAF allele in metastatic cells inhibited metastatic outgrowth as well as tumor initiation in mice. Our findings establish that metastatic competence can arise from heterogeneous cancer cell populations without the need for acquisition of additional mutations and that such competence can benefit from further selection of tumor-initiating mutations that seed primary tumorigenesis.


Subject(s)
Exome/genetics , Neoplasm Metastasis/genetics , Oncogenes , Alleles , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/secondary , Cell Line, Tumor , Comparative Genomic Hybridization , DNA, Neoplasm/genetics , Female , Gene Dosage , Heterografts , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Metastasis/physiopathology , Neoplasm Transplantation , Organ Specificity , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Selection, Genetic , Sequence Analysis, DNA , ras Proteins/genetics
19.
Nature ; 520(7547): 368-72, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25807485

ABSTRACT

Drug resistance invariably limits the clinical efficacy of targeted therapy with kinase inhibitors against cancer. Here we show that targeted therapy with BRAF, ALK or EGFR kinase inhibitors induces a complex network of secreted signals in drug-stressed human and mouse melanoma and human lung adenocarcinoma cells. This therapy-induced secretome stimulates the outgrowth, dissemination and metastasis of drug-resistant cancer cell clones and supports the survival of drug-sensitive cancer cells, contributing to incomplete tumour regression. The tumour-promoting secretome of melanoma cells treated with the kinase inhibitor vemurafenib is driven by downregulation of the transcription factor FRA1. In situ transcriptome analysis of drug-resistant melanoma cells responding to the regressing tumour microenvironment revealed hyperactivation of several signalling pathways, most prominently the AKT pathway. Dual inhibition of RAF and the PI(3)K/AKT/mTOR intracellular signalling pathways blunted the outgrowth of the drug-resistant cell population in BRAF mutant human melanoma, suggesting this combination therapy as a strategy against tumour relapse. Thus, therapeutic inhibition of oncogenic drivers induces vast secretome changes in drug-sensitive cancer cells, paradoxically establishing a tumour microenvironment that supports the expansion of drug-resistant clones, but is susceptible to combination therapy.


Subject(s)
Disease Progression , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/metabolism , Melanoma/metabolism , Metabolome/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Anaplastic Lymphoma Kinase , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Clone Cells/pathology , Down-Regulation/drug effects , Enzyme Activation/drug effects , ErbB Receptors/antagonists & inhibitors , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma/drug therapy , Melanoma/pathology , Mice , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fos/deficiency , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
20.
Trends Cancer ; 1(1): 76-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-28741564

ABSTRACT

The clinical manifestation of metastasis in a vital organ is the final stage of cancer progression and the main culprit of cancer-related mortality. Once established, metastasis is devastating, but only a small proportion of the cancer cells that leave a tumor succeed at infiltrating, surviving, and ultimately overtaking a distant organ. The bottlenecks that challenge cancer cells in newly invaded microenvironments are organ-specific and consequently demand distinct mechanisms for metastatic colonization. We review the metastatic traits that allow cancer cells to colonize distinct organ sites.

SELECTION OF CITATIONS
SEARCH DETAIL