1
|
Krushkal J, Zhao Y, Roney K, Zhu W, Brooks A, Wilsker D, Parchment RE, McShane LM, Doroshow JH. Association of changes in expression of HDAC and SIRT genes after drug treatment with cancer cell line sensitivity to kinase inhibitors. Epigenetics 2024; 19:2309824. [PMID: 38369747 PMCID: PMC10878021 DOI: 10.1080/15592294.2024.2309824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/14/2024] [Indexed: 02/20/2024] Open
Abstract
Histone deacetylases (HDACs) and sirtuins (SIRTs) are important epigenetic regulators of cancer pathways. There is a limited understanding of how transcriptional regulation of their genes is affected by chemotherapeutic agents, and how such transcriptional changes affect tumour sensitivity to drug treatment. We investigated the concerted transcriptional response of HDAC and SIRT genes to 15 approved antitumor agents in the NCI-60 cancer cell line panel. Antitumor agents with diverse mechanisms of action induced upregulation or downregulation of multiple HDAC and SIRT genes. HDAC5 was upregulated by dasatinib and erlotinib in the majority of the cell lines. Tumour cell line sensitivity to kinase inhibitors was associated with upregulation of HDAC5, HDAC1, and several SIRT genes. We confirmed changes in HDAC and SIRT expression in independent datasets. We also experimentally validated the upregulation of HDAC5 mRNA and protein expression by dasatinib in the highly sensitive IGROV1 cell line. HDAC5 was not upregulated in the UACC-257 cell line resistant to dasatinib. The effects of cancer drug treatment on expression of HDAC and SIRT genes may influence chemosensitivity and may need to be considered during chemotherapy.
Collapse
Affiliation(s)
- Julia Krushkal
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Yingdong Zhao
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Kyle Roney
- Department of Biostatistics and Bioinformatics, George Washington University, Washington, DC, USA
| | - Weimin Zhu
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Alan Brooks
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Deborah Wilsker
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ralph E. Parchment
- Clinical Pharmacodynamic Biomarkers Program, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Lisa M. McShane
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis and Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
2
|
Imyanitov EN, Mitiushkina NV, Kuligina ES, Tiurin VI, Venina AR. Pathways and targeting avenues of BRAF in non-small cell lung cancer. Expert Opin Ther Targets 2024; 28:613-622. [PMID: 38941191 DOI: 10.1080/14728222.2024.2374742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/27/2024] [Indexed: 06/30/2024]
Abstract
INTRODUCTION BRAF is a serine-threonine kinase implicated in the regulation of MAPK signaling cascade. BRAF mutation-driven activation occurs in approximately 2-4% of treatment-naive non-small cell carcinomas (NSCLCs). BRAF upregulation is also often observed in tumors with acquired resistance to receptor tyrosine kinase inhibitors (TKIs). AREAS COVERED This review describes the spectrum of BRAF mutations and their functional roles, discusses treatment options available for BRAF p.V600 and non-V600 mutated NSCLCs, and identifies some gaps in the current knowledge. EXPERT OPINION Administration of combined BRAF/MEK inhibitors usually produces significant, although often a short-term, benefit to NSCLC patients with BRAF V600 (class 1) mutations. There are no established treatments for BRAF class 2 (L597, K601, G464, G469A/V/R/S, fusions, etc.) and class 3 (D594, G596, G466, etc.) mutants, which account for up to two-thirds of BRAF-driven NSCLCs. Many important issues related to the use of immune therapy for the management of BRAF-mutated NSCLC deserve further investigation. The rare occurrence of BRAF mutations in NSCLC is compensated by high overall incidence of lung cancer disease; therefore, clinical studies on BRAF-associated NSCLC are feasible.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, Russia
| | - Natalia V Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Ekatherina Sh Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Vladislav I Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Aigul R Venina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| |
Collapse
|
3
|
Lazar R, Fischbach C, Schott R, Somme L. Outcomes of non-small cell lung cancer patients with non-V600E BRAF mutations: a series of case reports and literature review. Front Oncol 2024; 14:1307882. [PMID: 38601760 PMCID: PMC11004365 DOI: 10.3389/fonc.2024.1307882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most prevalent form of lung cancer, accounting for approximately 85% of cases of lung cancer. The standard first-line therapy for patients without oncogenic driver metastatic NSCLC is anti PD-L1 immune checkpoint inhibition (ICI) with platinum-based chemotherapy. Approximately 4% of NSCLC patients harbor BRAF mutations; the V600E mutation is the most common. Non-V600 mutations is an heterogeneous population and account for approximately 50% of BRAF-mutated NSCLC. BRAF mutations are classified into 3 functional classes based on their kinase activity and their signaling mechanism. The European Medicines Agency and the United States Food and Drug Administration have approved dabrafenib, an anti-BRAF tyrosine kinase inhibitor (TKI), in combination with trametinib, an anti-MEK TKI, for the treatment of patients with BRAF V600E-mutated metastatic NSCLC. The use of targeted therapies in NSCLC with BRAF non-V600E mutations remains controversial. There is a lack of guidelines regarding therapeutic options in non-V600E BRAF-mutated NSCLC. Herein, we presented 3 cases of NSCLC with BRAF non-V600E mutations and reviewed the current state of therapies for this particular population of lung cancer.
Collapse
Affiliation(s)
- Raluca Lazar
- Oncology Department, Institut De Cancérologie Strasbourg-Europe, Strasbourg, France
| | | | | | | |
Collapse
|
4
|
Guaitoli G, Zullo L, Tiseo M, Dankner M, Rose AAN, Facchinetti F. Non-small-cell lung cancer: how to manage BRAF-mutated disease. Drugs Context 2023; 12:dic-2022-11-3. [PMID: 37168877 PMCID: PMC10166262 DOI: 10.7573/dic.2022-11-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/17/2023] [Indexed: 05/13/2023] Open
Abstract
BRAF mutations are reported in about 3-5% of non-small-cell lung cancer (NSCLC), almost exclusively in adenocarcinoma histology, and are classified into three different classes. The segmentation of BRAF mutations into V600 (class 1) and non-V600 (classes 2 and 3) relies on their biological characteristics and is of interest for predicting the therapeutic benefit of targeted therapies and immunotherapy. Given the relative rarity of this molecular subset of disease, evidence supporting treatment choices is limited. This review aims to offer a comprehensive update about available therapeutic options for patients with NSCLC harbouring BRAF mutations to guide the physician in the choice of treatment strategies. We collected the most relevant available data, from single-arm phase II studies and retrospective analyses conducted in advanced NSCLC, regarding the efficacy of BRAF and MEK inhibitors in both V600 and non-V600 BRAF mutations. We included case reports and smaller experiences that could provide information on specific alterations. With respect to immunotherapy, we reviewed retrospective evidence on immune-checkpoint inhibitors in this molecular subset, whereas data about chemo-immunotherapy in this molecular subgroup are lacking. Moreover, we included the available, though limited, retrospective evidence of immunotherapy as consolidation after chemo-radiation for unresectable stage III BRAF-mutant NSCLC, and an overview of ongoing clinical trials in the peri-operative setting that could open new perspectives in the future.
Collapse
Affiliation(s)
- Giorgia Guaitoli
- Université Paris-Saclay, Gustave Roussy, INSERM U981, Villejuif, France
- PhD Program Clinical & Experimental Medicine, University of Modena & Reggio Emilia, Modena, Italy
| | - Lodovica Zullo
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marcello Tiseo
- Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Matthew Dankner
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - April AN Rose
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Francesco Facchinetti
- Université Paris-Saclay, Gustave Roussy, INSERM U981, Villejuif, France
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
5
|
Guerin N, Feichtner A, Stefan E, Kaserer T, Donald BR. Resistor: An algorithm for predicting resistance mutations via Pareto optimization over multistate protein design and mutational signatures. Cell Syst 2022; 13:830-843.e3. [PMID: 36265469 PMCID: PMC9589925 DOI: 10.1016/j.cels.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/29/2022] [Accepted: 09/13/2022] [Indexed: 01/26/2023]
Abstract
Resistance to pharmacological treatments is a major public health challenge. Here, we introduce Resistor-a structure- and sequence-based algorithm that prospectively predicts resistance mutations for drug design. Resistor computes the Pareto frontier of four resistance-causing criteria: the change in binding affinity (ΔKa) of the (1) drug and (2) endogenous ligand upon a protein's mutation; (3) the probability a mutation will occur based on empirically derived mutational signatures; and (4) the cardinality of mutations comprising a hotspot. For validation, we applied Resistor to EGFR and BRAF kinase inhibitors treating lung adenocarcinoma and melanoma. Resistor correctly identified eight clinically significant EGFR resistance mutations, including the erlotinib and gefitinib "gatekeeper" T790M mutation and five known osimertinib resistance mutations. Furthermore, Resistor predictions are consistent with BRAF inhibitor sensitivity data from both retrospective and prospective experiments using KinCon biosensors. Resistor is available in the open-source protein design software OSPREY.
Collapse
Affiliation(s)
- Nathan Guerin
- Department of Computer Science, Duke University, Durham, NC 27708, USA
| | - Andreas Feichtner
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, 6020 Tyrol, Austria
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, 6020 Tyrol, Austria; Tyrolean Cancer Research Institute, Innsbruck, 6020 Tyrol, Austria
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry, University of Innsbruck, Innsbruck, 6020 Tyrol, Austria.
| | - Bruce R Donald
- Department of Computer Science, Duke University, Durham, NC 27708, USA; Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA; Department of Chemistry, Duke University, Durham, NC 27708, USA; Department of Mathematics, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
6
|
Riudavets M, Cascetta P, Planchard D. Targeting BRAF-mutant non-small cell lung cancer: current status and future directions. Lung Cancer 2022; 169:102-114. [DOI: 10.1016/j.lungcan.2022.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
|
7
|
Tabbò F, Pisano C, Mazieres J, Mezquita L, Nadal E, Planchard D, Pradines A, Santamaria D, Swalduz A, Ambrogio C, Novello S, Ortiz-Cuaran S. How far we have come targeting BRAF-mutant non-small cell lung cancer (NSCLC). Cancer Treat Rev 2022; 103:102335. [DOI: 10.1016/j.ctrv.2021.102335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 12/27/2022]
|
8
|
2-Hydroxyestradiol Overcomes Mesenchymal Stem Cells-Mediated Platinum Chemoresistance in Ovarian Cancer Cells in an ERK-Independent Fashion. Molecules 2022; 27:molecules27030804. [PMID: 35164068 PMCID: PMC8839885 DOI: 10.3390/molecules27030804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer (OC) is the second most common type of gynecological malignancy. Platinum (Pt)-based chemotherapy is the standard of care for OC, but toxicity and acquired chemoresistance has proven challenging. Recently, we reported that sensitivity to platinum was significantly reduced in a co-culture of OC cells with MSC. To discover compounds capable of restoring platinum sensitivity, we screened a number of candidates and monitored ability to induce PARP cleavage. Moreover, we monitored platinum uptake and expression of ABC transporters in OC cells. Our results showed that 2-hydroxyestradiol (2HE2), a metabolite of estradiol, and dasatinib, an Abl/Src kinase inhibitor, were significantly effective in overcoming MSC-mediated platinum drug resistance. Dasatinib activity was dependent on ERK1/2 activation, whereas 2HE2 was independent of the activation of ERK1/2. MSC-mediated platinum drug resistance was accompanied by reduced intracellular platinum concentrations in OC cells. Moreover, MSC co-cultured with OC cells resulted in downregulation of the expression of cellular transporters required for platinum uptake and efflux. Exposure to 2HE2 and other modulators resulted in an increase in intracellular platinum concentrations. Thus, 2HE2 and dasatinib might act as sensitizers to restore platinum drug sensitivity to OC cells and thus to limit TME-mediated chemoresistance in OC.
Collapse
|
9
|
Liu X, Song C, Huang F, Fu H, Xiao W, Zhang W. GraphCDR: a graph neural network method with contrastive learning for cancer drug response prediction. Brief Bioinform 2021; 23:6415314. [PMID: 34727569 DOI: 10.1093/bib/bbab457] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/25/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022] Open
Abstract
Predicting the response of a cancer cell line to a therapeutic drug is an important topic in modern oncology that can help personalized treatment for cancers. Although numerous machine learning methods have been developed for cancer drug response (CDR) prediction, integrating diverse information about cancer cell lines, drugs and their known responses still remains a great challenge. In this paper, we propose a graph neural network method with contrastive learning for CDR prediction. GraphCDR constructs a graph neural network based on multi-omics profiles of cancer cell lines, the chemical structure of drugs and known cancer cell line-drug responses for CDR prediction, while a contrastive learning task is presented as a regularizer within a multi-task learning paradigm to enhance the generalization ability. In the computational experiments, GraphCDR outperforms state-of-the-art methods under different experimental configurations, and the ablation study reveals the key components of GraphCDR: biological features, known cancer cell line-drug responses and contrastive learning are important for the high-accuracy CDR prediction. The experimental analyses imply the predictive power of GraphCDR and its potential value in guiding anti-cancer drug selection.
Collapse
Affiliation(s)
- Xuan Liu
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Congzhi Song
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Feng Huang
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Haitao Fu
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjie Xiao
- Information School, University of Washington, Washington, 98105, USA
| | - Wen Zhang
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
10
|
Alabi S, Jaime-Figueroa S, Yao Z, Gao Y, Hines J, Samarasinghe KTG, Vogt L, Rosen N, Crews CM. Mutant-selective degradation by BRAF-targeting PROTACs. Nat Commun 2021; 12:920. [PMID: 33568647 PMCID: PMC7876048 DOI: 10.1038/s41467-021-21159-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/14/2021] [Indexed: 12/26/2022] Open
Abstract
Over 300 BRAF missense mutations have been identified in patients, yet currently approved drugs target V600 mutants alone. Moreover, acquired resistance inevitably emerges, primarily due to RAF lesions that prevent inhibition of BRAF V600 with current treatments. Therefore, there is a need for new therapies that target other mechanisms of activated BRAF. In this study, we use the Proteolysis Targeting Chimera (PROTAC) technology, which promotes ubiquitination and degradation of neo-substrates, to address the limitations of BRAF inhibitor-based therapies. Using vemurafenib-based PROTACs, we achieve low nanomolar degradation of all classes of BRAF mutants, but spare degradation of WT RAF family members. Our lead PROTAC outperforms vemurafenib in inhibiting cancer cell growth and shows in vivo efficacy in a Class 2 BRAF xenograft model. Mechanistic studies reveal that BRAFWT is spared due to weak ternary complex formation in cells owing to its quiescent inactivated conformation, and activation of BRAFWT sensitizes it to degradation. This study highlights the degree of selectivity achievable with degradation-based approaches by targeting mutant BRAF-driven cancers while sparing BRAFWT, providing an anti-tumor drug modality that expands the therapeutic window. Hundreds of BRAF mutations have been identified in patients with cancer but currently approved drugs only target BRAF V600 mutants. Here, the authors develop a vemurafenib-based PROTAC that induces degradation of all classes of BRAF mutants without affecting wild-type RAF proteins.
Collapse
Affiliation(s)
| | - Saul Jaime-Figueroa
- Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Zhan Yao
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yijun Gao
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Hines
- Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | | | - Lea Vogt
- Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Neal Rosen
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Craig M Crews
- Department of Pharmacology, New Haven, CT, USA. .,Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA. .,Department of Chemistry, Yale University, New Haven, CT, USA.
| |
Collapse
|
11
|
Zhao Y, Yu H, Ida CM, Halling KC, Kipp BR, Geiersbach K, Rumilla KM, Gupta S, Lin MT, Zheng G. Assessment of RAS Dependency for BRAF Alterations Using Cancer Genomic Databases. JAMA Netw Open 2021; 4:e2035479. [PMID: 33507258 PMCID: PMC7844594 DOI: 10.1001/jamanetworkopen.2020.35479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/06/2020] [Indexed: 11/28/2022] Open
Abstract
IMPORTANCE Understanding RAS dependency and mechanisms of RAS activation in non-V600 BRAF variant cancers has important clinical implications. This is the first study to date to systematically assess RAS dependency of BRAF alterations with real-world cancer genomic databases. OBJECTIVE To evaluate RAS dependency of individual BRAF alterations through alteration coexistence analysis using cancer genomic databases. DESIGN AND SETTING A cross-sectional data analysis of 119 538 nonredundant cancer samples using cancer genomics databases including GENIE (Genomics Evidence Neoplasia Information Exchange) and databases in cBioPortal including TCGA (The Cancer Genome Atlas) (accessed March 24, 2020), in addition to 2745 cancer samples from Mayo Clinic Genomics Laboratory (January 1, 2015, to July 1, 2020). Frequencies and odds ratios of coexisting alterations of RAS (KRAS, NRAS and HRAS) and RAS regulatory genes (NF1, PTPN11 and CBL) were calculated for individual BRAF alterations, and compared according to the current BRAF alteration classification; cancer type specificity of coexisting alterations of RAS or RAS regulatory genes was also evaluated. MAIN OUTCOMES AND MEASURES Primary outcome measurement is enrichment of RAS (KRAS, NRAS and HRAS) alterations in BRAF variant cancers. Secondary outcome measurement is enrichment of RAS regulatory gene (NF1, PTPN11, and CBL) in BRAF variant cancers. RESULTS A total of 2745 cancer samples from 2708 patients (female/male ratio: 1.0) tested by Mayo Clinic Genomics Laboratory and 119 538 patients (female/male ratio: 1.1) from GENIE and cBioPortal database were included in the study. In 119 538 nonredundant cancer samples, class 1 BRAF alterations and BRAF fusions were found to be mutually exclusive to alterations of RAS or RAS regulatory genes (odds ratio range 0.03-0.13 and 0.03-0.73 respectively), confirming their RAS independency. Both class 2 and class 3 BRAF alterations show variable and overlapping levels of enriched RAS alterations (odds ratio range: 0.03-5.9 and 0.63-2.52 respectively), suggesting heterogeneity in RAS dependency and a need to revisit BRAF alteration classification. For RAS-dependent BRAF alterations, the coexisting alterations also involve RAS regulatory genes by enrichment analysis (for example, S467L shows an odds ratio of 8.26 for NF1, 9.87 for PTPN11, and 15.23 for CBL) and occur in a variety of cancer types with some coalterations showing cancer type specificity (for example, HRAS variations account for 46.7% of all coexisting RAS alterations in BRAF variant bladder cancers, but 0% in non-small cell lung cancers). Variant-level assessment shows that BRAF alterations involving the same codon may differ in RAS dependency. In addition, RAS dependency of previously unclassified BRAF alterations could be assessed. CONCLUSIONS AND RELEVANCE Current BRAF alteration classification based on in vitro assays does not accurately predict RAS dependency in vivo for non-V600 BRAF alterations. RAS-dependent BRAF variant cancers with different mechanisms of RAS activation suggest the need for different treatment strategies.
Collapse
Affiliation(s)
- Yiqing Zhao
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Hanzhong Yu
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Cris M. Ida
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kevin C. Halling
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | - Benjamin R. Kipp
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | - Katherine Geiersbach
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Sounak Gupta
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gang Zheng
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
12
|
Zhang J, Chen Y, He Q. Distinct characteristics of dasatinib-induced pyroptosis in gasdermin E-expressing human lung cancer A549 cells and neuroblastoma SH-SY5Y cells. Oncol Lett 2020; 20:145-154. [PMID: 32565942 PMCID: PMC7285962 DOI: 10.3892/ol.2020.11556] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/21/2020] [Indexed: 02/04/2023] Open
Abstract
Dasatinib, a multikinase inhibitor, is used in the treatment of chronic myeloid leukemia and was developed to overcome imatinib resistance. Its mechanism of action involves the induction of apoptosis, autophagy and necroptosis. However, it remains unclear whether dasatinib can induce pyroptosis. In the present study, gasdermin E (GSDME)-expressing SH-SY5Y and A549 cells were chosen for investigation. Typical pyroptotic features, such as cleavage of GSDME protein, leakage of lactate dehydrogenase and large bubbled morphology, were observed in both cell lines after exposure to dasatinib. The generation of GSDME fragments was inhibited by specific caspase-3 inhibitor zDEVD in SH-SY5Y cells and pan-caspase inhibitor zVAD in A549 cells. Moreover, distinct characteristics of pyroptosis were observed in A549 cells, which occurred only with a high percentage of Annexin V/propidium iodide double-stained cells and low level of GSDME protein cleavage. The sensitivity of A549 cells to dasatinib is significantly reduced by increasing cell numbers. The elevation of GSDMD and GSDME protein levels was induced by low concentrations of dasatinib, which was not influenced by the reduction of p53 protein with RNA interference. In conclusion, to the best of our knowledge, this is the first study to report that dasatinib can induce pyroptosis in tumor cells and increase the protein levels of GSDMD and GSDME in a p53-independent manner.
Collapse
Affiliation(s)
- Juan Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Yang Chen
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Qiyang He
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| |
Collapse
|
13
|
Saleh T, Bloukh S, Carpenter VJ, Alwohoush E, Bakeer J, Darwish S, Azab B, Gewirtz DA. Therapy-Induced Senescence: An "Old" Friend Becomes the Enemy. Cancers (Basel) 2020; 12:cancers12040822. [PMID: 32235364 PMCID: PMC7226427 DOI: 10.3390/cancers12040822] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 01/10/2023] Open
Abstract
For the past two decades, cellular senescence has been recognized as a central component of the tumor cell response to chemotherapy and radiation. Traditionally, this form of senescence, termed Therapy-Induced Senescence (TIS), was linked to extensive nuclear damage precipitated by classical genotoxic chemotherapy. However, a number of other forms of therapy have also been shown to induce senescence in tumor cells independently of direct genomic damage. This review attempts to provide a comprehensive summary of both conventional and targeted anticancer therapeutics that have been shown to induce senescence in vitro and in vivo. Still, the utility of promoting senescence as a therapeutic endpoint remains under debate. Since senescence represents a durable form of growth arrest, it might be argued that senescence is a desirable outcome of cancer therapy. However, accumulating evidence suggesting that cells have the capacity to escape from TIS would support an alternative conclusion, that senescence provides an avenue whereby tumor cells can evade the potentially lethal action of anticancer drugs, allowing the cells to enter a temporary state of dormancy that eventually facilitates disease recurrence, often in a more aggressive state. Furthermore, TIS is now strongly connected to tumor cell remodeling, potentially to tumor dormancy, acquiring more ominous malignant phenotypes and accounts for several untoward adverse effects of cancer therapy. Here, we argue that senescence represents a barrier to effective anticancer treatment, and discuss the emerging efforts to identify and exploit agents with senolytic properties as a strategy for elimination of the persistent residual surviving tumor cell population, with the goal of mitigating the tumor-promoting influence of the senescent cells and to thereby reduce the likelihood of cancer relapse.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Sarah Bloukh
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Valerie J. Carpenter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Enas Alwohoush
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Jomana Bakeer
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
| | - Sarah Darwish
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan; (T.S.); (S.D.)
| | - Belal Azab
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan; (S.B.); (E.A.); (J.B.); (B.A.)
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Correspondence:
| |
Collapse
|
14
|
Ahmed E, Masud MK, Hossain MSA, Na J, Sina AAI, Yamauchi Y, Trau M. Nanostructured mesoporous gold electrodes detect protein phosphorylation in cancer with electrochemical signal amplification. Analyst 2020; 145:6639-6648. [DOI: 10.1039/d0an01096k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A nanostructured mesoporous gold electrode is demonstrated to detect the phosphorylated protein over non-phosphorylated in cancer using electrochemical signal amplification through differential pulse voltammetry in the presence of the [Fe(CN)6]3−/4−.
Collapse
Affiliation(s)
- Emtiaz Ahmed
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- The University of Queensland
- Brisbane
- Australia
| | - Mostafa Kamal Masud
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- The University of Queensland
- Brisbane
- Australia
| | - Md. Shahriar A. Hossain
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- The University of Queensland
- Brisbane
- Australia
- School of Mechanical and Mining Engineering
| | - Jongbeom Na
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- The University of Queensland
- Brisbane
- Australia
| | - Abu Ali Ibn Sina
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- The University of Queensland
- Brisbane
- Australia
| | - Yusuke Yamauchi
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- The University of Queensland
- Brisbane
- Australia
- School of Chemical Engineering
| | - Matt Trau
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- The University of Queensland
- Brisbane
- Australia
- School of Chemistry and Molecular Biosciences
| |
Collapse
|
15
|
Giopanou I, Pintzas A. RAS and BRAF in the foreground for non-small cell lung cancer and colorectal cancer: Similarities and main differences for prognosis and therapies. Crit Rev Oncol Hematol 2019; 146:102859. [PMID: 31927392 DOI: 10.1016/j.critrevonc.2019.102859] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Lung and colorectal cancer are included in the most tremendously threatening diseases in terms of incidence and death. Although they are located in completely different organs and differ in various characteristics they do share some common features, especially regarding their molecular mutational profile. Among several commonly mutated genes KRAS and BRAF are spotted to be highly associated with patient's poor disease outcome and resistance to targeted therapies mostly in liaison with other mutant activated genes. Many studies have shed light in these mechanisms for disease progression and numerous preclinical models, clinical trials and meta-analysis reports investigate the impact of specific treatments or combination of therapies. The present review is an effort to compare the mutational imprint of these genes between the two diseases and their impact in prognosis, current therapy, mechanisms of therapy resistance and future therapeutic plans and provide a spherical perspective regarding the systemic molecular profile of cancer.
Collapse
Affiliation(s)
- Ioanna Giopanou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece.
| | - Alexandros Pintzas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece.
| |
Collapse
|
16
|
Multi-Institutional Evaluation of Interrater Agreement of Variant Classification Based on the 2017 Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists Standards and Guidelines for the Interpretation and Reporting of Sequence Variants in Cancer. J Mol Diagn 2019; 22:284-293. [PMID: 31837433 DOI: 10.1016/j.jmoldx.2019.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 10/08/2019] [Accepted: 10/24/2019] [Indexed: 11/22/2022] Open
Abstract
This multi-institutional study was undertaken to evaluate interrater reliability of the 2017 Association for Molecular Pathology/American Society of Clinical Oncology/College of American Pathologists guidelines for interpretation and reporting of oncology sequence variants and to assess current practices and perceptions surrounding these guidelines. Fifty-one variants were distributed to 20 participants from 10 institutions for classification using the new guidelines. Agreement was assessed using chance-corrected agreement (Cohen κ). κ was 0.35. To evaluate if data sharing could help resolve disagreements, a summary of variant classifications and additional information about each variant were distributed to all participants. κ improved to 0.7 after the original classifications were revised. Participants were invited to take a web-based survey regarding their perceptions of the guidelines. Only 20% (n = 3) of the survey respondents had prior experience with the guidelines in clinical practice. The main perceived barriers to guideline implementation included the complexity of the guidelines, discordance between clinical actionability and pathobiologic relevance, lack of familiarity with the new classifications, and uncertainty when applying criteria to potential germline variants. This study demonstrates noteworthy discordances between pathologists for variant classification in solid tumors when using the 2017 Association for Molecular Pathology/American Society of Clinical Oncology/College of American Pathologists guidelines. These findings highlight potential areas for clarification/refinement before mainstream clinical adoption.
Collapse
|
17
|
Liu Q, Muglia LJ, Huang LF. Network as a Biomarker: A Novel Network-Based Sparse Bayesian Machine for Pathway-Driven Drug Response Prediction. Genes (Basel) 2019; 10:genes10080602. [PMID: 31405013 PMCID: PMC6723660 DOI: 10.3390/genes10080602] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 01/17/2023] Open
Abstract
With the advances in different biological networks including gene regulation, gene co-expression, protein–protein interaction networks, and advanced approaches for network reconstruction, analysis, and interpretation, it is possible to discover reliable and accurate molecular network-based biomarkers for monitoring cancer treatment. Such efforts will also pave the way toward the realization of biomarker-driven personalized medicine against cancer. Previously, we have reconstructed disease-specific driver signaling networks using multi-omics profiles and cancer signaling pathway data. In this study, we developed a network-based sparse Bayesian machine (NBSBM) approach, using previously derived disease-specific driver signaling networks to predict cancer cell responses to drugs. NBSBM made use of the information encoded in a disease-specific (differentially expressed) network to improve its prediction performance in problems with a reduced amount of training data and a very high-dimensional feature space. Sparsity in NBSBM is favored by a spike and slab prior distribution, which is combined with a Markov random field prior that encodes the network of feature dependencies. Gene features that are connected in the network are assumed to be both relevant and irrelevant to drug responses. We compared the proposed method with network-based support vector machine (NBSVM) approaches and found that the NBSBM approach could achieve much better accuracy than the other two NBSVM methods. The gene modules selected from the disease-specific driver networks for predicting drug sensitivity might be directly involved in drug sensitivity or resistance. This work provides a disease-specific network-based drug sensitivity prediction approach and can uncover the potential mechanisms of the action of drugs by selecting the most predictive sub-networks from the disease-specific network.
Collapse
Affiliation(s)
- Qi Liu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Louis J Muglia
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lei Frank Huang
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA.
- Department of Information Science, School of Mathematical Sciences and LAMA, Peking University, Beijing 100871, China.
| |
Collapse
|
18
|
Singh R, Peng S, Viswanath P, Sambandam V, Shen L, Rao X, Fang B, Wang J, Johnson FM. Non-canonical cMet regulation by vimentin mediates Plk1 inhibitor-induced apoptosis. EMBO Mol Med 2019; 11:e9960. [PMID: 31040125 PMCID: PMC6505578 DOI: 10.15252/emmm.201809960] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/28/2019] [Accepted: 03/12/2019] [Indexed: 12/26/2022] Open
Abstract
To address the need for improved systemic therapy for non-small-cell lung cancer (NSCLC), we previously demonstrated that mesenchymal NSCLC was sensitive to polo-like kinase (Plk1) inhibitors, but the mechanisms of resistance in epithelial NSCLC remain unknown. Here, we show that cMet was differentially regulated in isogenic pairs of epithelial and mesenchymal cell lines. Plk1 inhibition inhibits cMet phosphorylation only in mesenchymal cells. Constitutively active cMet abrogates Plk1 inhibitor-induced apoptosis. Likewise, cMet silencing or inhibition enhances Plk1 inhibitor-induced apoptosis. Cells with acquired resistance to Plk1 inhibitors are more epithelial than their parental cells and maintain cMet activation after Plk1 inhibition. In four animal NSCLC models, mesenchymal tumors were more sensitive to Plk1 inhibition alone than were epithelial tumors. The combination of cMet and Plk1 inhibition led to regression of tumors that did not regrow when drug treatment was stopped. Plk1 inhibition did not affect HGF levels but did decrease vimentin phosphorylation, which regulates cMet phosphorylation via β1-integrin. This research defines a heretofore unknown mechanism of ligand-independent activation of cMet downstream of Plk1 and an effective combination therapy.
Collapse
Affiliation(s)
- Ratnakar Singh
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shaohua Peng
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pavitra Viswanath
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Vaishnavi Sambandam
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Shen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiayu Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faye M Johnson
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
19
|
Zheng G, Tseng LH, Haley L, Ibrahim J, Bynum J, Xian R, Gocke CD, Eshleman JR, Lin MT. Clinical validation of coexisting driver mutations in colorectal cancers. Hum Pathol 2019; 86:12-20. [PMID: 30481508 PMCID: PMC6467705 DOI: 10.1016/j.humpath.2018.11.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 12/17/2022]
Abstract
Mutational profiling is recommended for selecting targeted therapy and predicting prognosis of metastatic colorectal cancer (CRC). Detection of coexisting mutations within the same pathway, which are usually mutually exclusive, raises the concern for potential laboratory errors. In this retrospective study for quality assessment of a next-generation sequencing assay, we examined BRAF, KRAS, and NRAS genes within the mitogen-activated protein kinase (MAPK) pathway and the PIK3CA gene within the phosphatidylinositol 3-kinase (mTOR) pathway in 744 CRC specimens submitted to our clinical diagnostics laboratory. Although coexistence of mutations between the MAPK and mTOR pathways was observed, it rarely occurred within the MAPK pathway. Retrospective quality assessments identified false detection of coexisting activating KRAS and NRAS mutations in 1 specimen and confirmed 2 activating KRAS mutations in 2 specimens and coexisting activating KRAS and NRAS mutations in 2 specimens, but no coexisting activating RAS and BRAF mutations. There were 15 CRCs with a kinase-impaired BRAF mutation, including 3 with a coexisting activating KRAS mutation, which may have therapeutic implications. Multiregional analysis based on different histologic features demonstrated that coexisting KRAS and NRAS mutations may be present in the same or different tumor populations and showed that invasion of adenomas by synchronous adenocarcinomas of different clonal origin may result in detection of coexisting mutations within the MAPK pathway. In this study, we proposed an operating procedure for clinical validation of unexpected coexisting mutations. Further studies are warranted to elucidate the biological significance and clinical implications of coexisting mutations within the MAPK pathway.
Collapse
Affiliation(s)
- Gang Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Li-Hui Tseng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Medical Genetics, National Taiwan University Hospital, Taipei 100,Taiwan
| | - Lisa Haley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Junaid Ibrahim
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jennifer Bynum
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rena Xian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christopher D Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Schumacher D, Andrieux G, Boehnke K, Keil M, Silvestri A, Silvestrov M, Keilholz U, Haybaeck J, Erdmann G, Sachse C, Templin M, Hoffmann J, Boerries M, Schäfer R, Regenbrecht CRA. Heterogeneous pathway activation and drug response modelled in colorectal-tumor-derived 3D cultures. PLoS Genet 2019; 15:e1008076. [PMID: 30925167 PMCID: PMC6457557 DOI: 10.1371/journal.pgen.1008076] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 04/10/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022] Open
Abstract
Organoid cultures derived from colorectal cancer (CRC) samples are increasingly used as preclinical models for studying tumor biology and the effects of targeted therapies under conditions capturing in vitro the genetic make-up of heterogeneous and even individual neoplasms. While 3D cultures are initiated from surgical specimens comprising multiple cell populations, the impact of tumor heterogeneity on drug effects in organoid cultures has not been addressed systematically. Here we have used a cohort of well-characterized CRC organoids to study the influence of tumor heterogeneity on the activity of the KRAS/MAPK-signaling pathway and the consequences of treatment by inhibitors targeting EGFR and downstream effectors. MAPK signaling, analyzed by targeted proteomics, shows unexpected heterogeneity irrespective of RAS mutations and is associated with variable responses to EGFR inhibition. In addition, we obtained evidence for intratumoral heterogeneity in drug response among parallel “sibling” 3D cultures established from a single KRAS-mutant CRC. Our results imply that separate testing of drug effects in multiple subpopulations may help to elucidate molecular correlates of tumor heterogeneity and to improve therapy response prediction in patients. Commonly occurring genetic alterations and patient-specific genetic features are increasingly used to predict the possible action of targeted cancer therapies. Although several lines of evidence have suggested that preclinical and clinical responses concur, the heterogeneity of tumors remains a severe obstacle in routinely translating preclinical data to patient treatments. Here we present a rapid work flow that integrates drug testing of three-dimensional patient tumor-derived (organoid) cultures and assessment of their genetic make-up as well as that of their donor tumors by amplicon sequencing and targeted proteomics. While the organoid cultures largely recapitulated the genomic profiles of donor tumors, the overall treatment responses and inhibitor effects on the intracellular signaling system were quite variable. Notably, organoid cultures obtained by synchronous multi-regional sampling of the same colorectal tumor showed an up to 30-fold difference in drug response. A combinatorial drug treatment improved the response. These data were confirmed in matched mouse xenograft models from the same tumor. Our findings may help to refine preclinical testing of individual tumors by modelling heterogeneity in cultures, to better understand therapeutic failure in clinical settings and to find ways to overcome treatment resistance.
Collapse
Affiliation(s)
- Dirk Schumacher
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Geoffroy Andrieux
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Karsten Boehnke
- Eli Lilly and Company, Lilly Research Laboratories, Oncology Translational Research, New York, NY, United States of America
| | - Marlen Keil
- EPO Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin, Germany
| | | | | | | | - Johannes Haybaeck
- Department of Pathology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Department of Pathology, Neuropathology, and Molecular Pathology, Medical University of Innsbruck, Austria.,Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, Austria
| | - Gerrit Erdmann
- NMI TT Pharmaservices, Berlin, Germany.,ASC Oncology GmbH, Berlin, Germany
| | - Christoph Sachse
- NMI TT Pharmaservices, Berlin, Germany.,ASC Oncology GmbH, Berlin, Germany
| | - Markus Templin
- ASC Oncology GmbH, Berlin, Germany.,NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Jens Hoffmann
- EPO Experimental Pharmacology and Oncology Berlin-Buch GmbH, Berlin, Germany
| | - Melanie Boerries
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Reinhold Schäfer
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Charité Comprehensive Cancer Center, Berlin, Germany
| | - Christian R A Regenbrecht
- cpo-Cellular Phenomics & Oncology Berlin-Buch GmbH, Berlin, Germany.,Department of Pathology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,ASC Oncology GmbH, Berlin, Germany
| |
Collapse
|
21
|
Alvarez JGB, Otterson GA. Agents to treat BRAF-mutant lung cancer. Drugs Context 2019; 8:212566. [PMID: 30899313 PMCID: PMC6419923 DOI: 10.7573/dic.212566] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 01/06/2023] Open
Abstract
BRAF mutations are seen in up to 3.5–4% of the non-small cell lung cancer (NSCLC) patients. BRAF V600E mutations account for 50% of these cases, and the remaining BRAF mutations are non-V600E. The biologic behavior of BRAF-mutated lung tumors tends to be more aggressive and resistant to chemotherapy, but responses to tyrosine kinase inhibitors such as BRAF inhibitors with or without MEK inhibitors have provided another effective tool to attain better response rates when compared to cytotoxic chemotherapy. New strategies such as immunotherapy are becoming as well another option to treat in the second-line setting patients with BRAF-mutated NSCLC.
Collapse
Affiliation(s)
- Jean G Bustamante Alvarez
- Division of Medical Oncology, Department of Internal Medicine, The James Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Gregory A Otterson
- Division of Medical Oncology, Department of Internal Medicine, The James Cancer Center and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
22
|
Boussemart L, Nelson A, Wong M, Ross JS, Sosman J, Mehnert J, Daniels G, Kendra K, Ali SM, Miller VA, Schrock AB. Hybrid Capture-Based Genomic Profiling Identifies BRAF V600 and Non-V600 Alterations in Melanoma Samples Negative by Prior Testing. Oncologist 2019; 24:657-663. [PMID: 30683711 DOI: 10.1634/theoncologist.2018-0271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND BRAF and MEK inhibitors are approved for BRAF V600-mutated advanced melanoma, with response rates of up to 70%. Responses to targeted therapies have also been observed for diverse non-V600 BRAF alterations. Thus, sensitive, accurate, and broad detection of BRAF alterations is critical to match patients with available targeted therapies. MATERIALS AND METHODS Pathology reports were reviewed for 385 consecutive melanoma cases with BRAF mutations or rearrangements identified using a hybrid capture-based next-generation sequencing comprehensive genomic profiling (CGP) assay during the course of clinical care. RESULTS Records of prior BRAF molecular testing were available for 79 (21%) cases. Of cases with BRAF V600 mutations, 11/57 (19%) with available data were negative by prior BRAF testing. Prior negative BRAF results were also identified in 16/20 (80%) cases with non-V600 mutations, 2 of which harbored multiple BRAF alterations, and in 2/2 (100%) cases with activating BRAF fusions. Clinical outcomes for a subset of patients are presented. CONCLUSION CGP identifies diverse activating BRAF alterations in a significant fraction of cases with prior negative testing. Given the proven clinical benefit of BRAF/MEK inhibitors in BRAF-mutated melanoma, CGP should be considered for patients with metastatic melanoma, particularly if other testing is negative. IMPLICATIONS FOR PRACTICE Published guidelines for melanoma treatment recommend BRAF mutational analysis, but little guidance is provided as to selection criteria for testing methodologies, or as to clinical implications for non-V600 alterations. This study found that hybrid capture-based next-generation sequencing can detect BRAF alterations in samples from a significant fraction of patients with advanced melanoma with prior negative BRAF results. This study highlights the need for oncologists and pathologists to be critically aware of coverage and sensitivity limitations of various assays, particularly regarding non-V600E alterations, of which many are potentially targetable.
Collapse
Affiliation(s)
- Lise Boussemart
- Department of Dermatology, Pontchaillou Hospital, CHU de Rennes, Rennes, France
- University of Rennes, CNRS, IGDR, UMR 6290, Rennes, France
| | - Annie Nelson
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA
| | | | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA
- Department of Pathology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jeffrey Sosman
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Janice Mehnert
- Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Gregory Daniels
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Kari Kendra
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
23
|
BRAF Mutant Lung Cancer: Programmed Death Ligand 1 Expression, Tumor Mutational Burden, Microsatellite Instability Status, and Response to Immune Check-Point Inhibitors. J Thorac Oncol 2018; 13:1128-1137. [DOI: 10.1016/j.jtho.2018.04.024] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 03/23/2018] [Accepted: 04/23/2018] [Indexed: 11/20/2022]
|
24
|
Leonetti A, Facchinetti F, Rossi G, Minari R, Conti A, Friboulet L, Tiseo M, Planchard D. BRAF in non-small cell lung cancer (NSCLC): Pickaxing another brick in the wall. Cancer Treat Rev 2018; 66:82-94. [PMID: 29729495 DOI: 10.1016/j.ctrv.2018.04.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/28/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
Molecular characterization of non-small cell lung cancer (NSCLC) marked an historical turning point for the treatment of lung tumors harboring kinase alterations suitable for specific targeted drugs inhibition, translating into major clinical improvements. Besides EGFR, ALK and ROS1, BRAF represents a novel therapeutic target for the treatment of advanced NSCLC. BRAF mutations, found in 1.5-3.5% of NSCLC, are responsible of the constitutive activation of mitogen activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway. Clinical trials evaluating the efficacy of the BRAF inhibitor dabrafenib in combination with the downstream MEK inhibitor trametinib in metastatic BRAFV600E-mutated NSCLC guaranteed FDA and EMA rapid approval of the combination regimen in this clinical setting. In line with the striking results observed in metastatic melanoma harboring the same molecular alteration, BRAF and MEK inhibition should be considered a new standard of care in this molecular subtype of NSCLC. In the present review, we propose an overview of the available evidence about BRAF in NSCLC mutations (V600E and non-V600E), from biological significance to emerging clinical implications of BRAF mutations detection. Focusing on the current strategies to act against the mutated kinase, we moreover approach additional strategies to overcome treatment resistance.
Collapse
Affiliation(s)
| | | | - Giulio Rossi
- Pathology Unit, Santa Maria delle Croci Hospital, Ravenna, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - Luc Friboulet
- INSERM, U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.
| | - David Planchard
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
25
|
Toledo RA, Garralda E, Mitsi M, Pons T, Monsech J, Vega E, Otero Á, Albarran MI, Baños N, Durán Y, Bonilla V, Sarno F, Camacho-Artacho M, Sanchez-Perez T, Perea S, Álvarez R, De Martino A, Lietha D, Blanco-Aparicio C, Cubillo A, Domínguez O, Martínez-Torrecuadrada JL, Hidalgo M. Exome Sequencing of Plasma DNA Portrays the Mutation Landscape of Colorectal Cancer and Discovers Mutated VEGFR2 Receptors as Modulators of Antiangiogenic Therapies. Clin Cancer Res 2018; 24:3550-3559. [PMID: 29588308 DOI: 10.1158/1078-0432.ccr-18-0103] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/15/2018] [Accepted: 03/21/2018] [Indexed: 12/18/2022]
Abstract
Purpose: Despite the wide use of antiangiogenic drugs in the clinical setting, predictive biomarkers of response to these drugs are still unknown.Experimental Design: We applied whole-exome sequencing of matched germline and basal plasma cell-free DNA samples (WES-cfDNA) on a RAS/BRAF/PIK3CA wild-type metastatic colorectal cancer patient with primary resistance to standard treatment regimens, including inhibitors to the VEGF:VEGFR2 pathway. We performed extensive functional experiments, including ectopic expression of VEGFR2 mutants in different cell lines, kinase and drug sensitivity assays, and cell- and patient-derived xenografts.Results: WES-cfDNA yielded a 77% concordance rate with tumor exome sequencing and enabled the identification of the KDR/VEGFR2 L840F clonal, somatic mutation as the cause of therapy refractoriness in our patient. In addition, we found that 1% to 3% of samples from cancer sequencing projects harbor KDR somatic mutations located in protein residues frequently mutated in other cancer-relevant kinases, such as EGFR, ABL1, and ALK. Our in vitro and in vivo functional assays confirmed that L840F causes strong resistance to antiangiogenic drugs, whereas the KDR hot-spot mutant R1032Q confers sensitivity to strong VEGFR2 inhibitors. Moreover, we showed that the D717V, G800D, G800R, L840F, G843D, S925F, R1022Q, R1032Q, and S1100F VEGFR2 mutants promote tumor growth in mice.Conclusions: Our study supports WES-cfDNA as a powerful platform for portraying the somatic mutation landscape of cancer and discovery of new resistance mechanisms to cancer therapies. Importantly, we discovered that VEGFR2 is somatically mutated across tumor types and that VEGFR2 mutants can be oncogenic and control sensitivity/resistance to antiangiogenic drugs. Clin Cancer Res; 24(15); 3550-9. ©2018 AACR.
Collapse
Affiliation(s)
- Rodrigo A Toledo
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain. .,Vall d'Hebron Institute of Oncology (VHIO), CIBERONC, Barcelona, Spain
| | - Elena Garralda
- Vall d'Hebron Institute of Oncology (VHIO), CIBERONC, Barcelona, Spain.,Centro Integral Oncológico Clara Campal (CIOCC), Hospital Universitario HM Sanchinarro, Madrid, Spain.,Universidad San Pablo CEU, Madrid, Spain
| | - Maria Mitsi
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Tirso Pons
- Structural Computational Biology Group, CNIO, Madrid, Spain
| | | | - Estela Vega
- Centro Integral Oncológico Clara Campal (CIOCC), Hospital Universitario HM Sanchinarro, Madrid, Spain.,Universidad San Pablo CEU, Madrid, Spain
| | - Álvaro Otero
- Crystallography and Protein Engineering Unit, CNIO, Madrid, Spain
| | | | - Natalia Baños
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Yolanda Durán
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Victoria Bonilla
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Francesca Sarno
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Tania Sanchez-Perez
- Molecular Genetics of Angiogenesis Laboratory, Spanish National Center for Cardiovascular Research (CNIC), Madrid, Spain
| | - Sofia Perea
- Gastrointestinal Cancer Clinical Research Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Rafael Álvarez
- Centro Integral Oncológico Clara Campal (CIOCC), Hospital Universitario HM Sanchinarro, Madrid, Spain.,Universidad San Pablo CEU, Madrid, Spain
| | | | - Daniel Lietha
- Cell Signalling and Adhesion Group, CNIO, Madrid, Spain
| | | | - Antonio Cubillo
- Centro Integral Oncológico Clara Campal (CIOCC), Hospital Universitario HM Sanchinarro, Madrid, Spain.,Universidad San Pablo CEU, Madrid, Spain
| | | | | | - Manuel Hidalgo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
26
|
|
27
|
Type II RAF inhibitor causes superior ERK pathway suppression compared to type I RAF inhibitor in cells expressing different BRAF mutant types recurrently found in lung cancer. Oncotarget 2018; 9:16110-16123. [PMID: 29662630 PMCID: PMC5882321 DOI: 10.18632/oncotarget.24576] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 02/20/2018] [Indexed: 12/19/2022] Open
Abstract
A large fraction of somatic driver BRAF mutations in lung cancer are non-V600 and impaired-kinase. Non-V600 BRAF mutations predict sensitivity to combination of a type I RAF inhibitor, Dabrafenib, and a MEK inhibitor, Trametinib. Singly, Dabrafenib only weakly suppresses mutant BRAF-induced ERK signaling and can induce ERK paradoxical activation in CRAF-overexpressing cells. The present study compared the effects of Dabrafenib and a type II RAF inhibitor, AZ628, on ERK activity in HEK293T cells expressing several tumor-derived BRAF mutants, and in a non-V600 and impaired-kinase BRAF-mutant lung cancer cell line (H1666). Unlike Dabrafenib, AZ628 did not induce paradoxical ERK activation in CRAF-overexpressing cells and BRAF-mutant cells overexpressing CRAF were more responsive to AZ628 compared to Dabrafenib in terms of ERK inhibition. AZ628 inhibited ERK more effectively than Dabrafenib in both H1666 cells and HEK293T cells co-expressing several different BRAF-mutants with CRAF. Similarly, AZ628 plus Trametinib had better MEK-inhibitory and pro-apoptotic effects in H1666 cells than Dabrafenib plus Trametinib. Moreover, prolonged treatment of H1666 cells with AZ628 plus Trametinib produced greater inhibition of cell growth than Dabrafenib plus Trametinib. These results indicate that AZ628 has greater potential than Dabrafenib, both as a single agent and combined with Trametinib, for the treatment of non-V600 BRAF mutant lung cancer.
Collapse
|
28
|
Li SD, Ma M, Li H, Waluszko A, Sidorenko T, Schadt EE, Zhang DY, Chen R, Ye F. Cancer gene profiling in non-small cell lung cancers reveals activating mutations in JAK2 and JAK3 with therapeutic implications. Genome Med 2017; 9:89. [PMID: 29082853 PMCID: PMC5662094 DOI: 10.1186/s13073-017-0478-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/04/2017] [Indexed: 01/05/2023] Open
Abstract
Background Next-generation sequencing (NGS) of cancer gene panels are widely applied to enable personalized cancer therapy and to identify novel oncogenic mutations. Methods We performed targeted NGS on 932 clinical cases of non-small-cell lung cancers (NSCLCs) using the Ion AmpliSeq™ Cancer Hotspot panel v2 assay. Results Actionable mutations were identified in 65% of the cases with available targeted therapeutic options, including 26% of the patients with mutations in National Comprehensive Cancer Network (NCCN) guideline genes. Most notably, we discovered JAK2 p.V617F somatic mutation, a hallmark of myeloproliferative neoplasms, in 1% (9/932) of the NSCLCs. Analysis of cancer cell line pharmacogenomic data showed that a high level of JAK2 expression in a panel of NSCLC cell lines is correlated with increased sensitivity to a selective JAK2 inhibitor. Further analysis of TCGA genomic data revealed JAK2 gain or loss due to genetic alterations in NSCLC clinical samples are associated with significantly elevated or reduced PD-L1 expression, suggesting that the activating JAK2 p.V617F mutation could confer sensitivity to both JAK inhibitors and anti-PD1 immunotherapy. We also detected JAK3 germline activating mutations in 6.7% (62/932) of the patients who may benefit from anti-PD1 treatment, in light of recent findings that JAK3 mutations upregulate PD-L1 expression. Conclusion Taken together, this study demonstrated the clinical utility of targeted NGS with a focused hotspot cancer gene panel in NSCLCs and identified activating mutations in JAK2 and JAK3 with clinical implications inferred through integrative analysis of cancer genetic, genomic, and pharmacogenomic data. The potential of JAK2 and JAK3 mutations as response markers for the targeted therapy against JAK kinases or anti-PD1 immunotherapy warrants further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0478-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shuyu D Li
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sema4, a Mount Sinai venture, Stamford, CT, 06902, USA
| | - Meng Ma
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sema4, a Mount Sinai venture, Stamford, CT, 06902, USA
| | - Hui Li
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sema4, a Mount Sinai venture, Stamford, CT, 06902, USA
| | - Aneta Waluszko
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tatyana Sidorenko
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sema4, a Mount Sinai venture, Stamford, CT, 06902, USA
| | - David Y Zhang
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rong Chen
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Sema4, a Mount Sinai venture, Stamford, CT, 06902, USA.
| | - Fei Ye
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Pathology, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
29
|
Phase I study of the combination of crizotinib (as a MET inhibitor) and dasatinib (as a c-SRC inhibitor) in patients with advanced cancer. Invest New Drugs 2017; 36:416-423. [PMID: 29047029 DOI: 10.1007/s10637-017-0513-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/20/2017] [Indexed: 01/19/2023]
Abstract
Background Both MET and c-SRC are important mediators of cancer progression and there is cross talk between the two molecules. Preclinical studies have demonstrated combination of MET and c-SRC inhibitors is effective in multiple cancer types. Methods We analyzed the safety and efficacy of administering a c-SRC inhibitor (dasatinib) in combination with a MET inhibitor (crizotinib) in a two-arm concurrent phase I study. Arm A consisted of crizotinib fixed at 250 mg twice per day with escalation of dasatinib. Arm B consisted of dasatinib fixed at 140 mg daily with escalation of crizotinib. Endpoints included dose-limiting toxicities (DLTs), recommended phase II dose (RP2D), and response (RECIST 1.1). Results We enrolled 61 patients (arm A: 31, arm B: 30). The most common cancers were sarcoma (21%) and prostate cancer (16%). In Arm A, at dose level 2 (DL2), 40% (2/5) experienced DLTs. In the expanded DL1, 21% (4/19) experienced DLTs (all grade 3). In Arm B, at DL2, 50% (2/4) experienced DLTs. In the expanded DL1, 22% (4/18) experienced DLTs (all grade 3). RP2D was determined to be arm A, DL1 (250 mg crizotinib orally twice per day plus 50 mg dasatinib orally daily). Partial response (N = 1) and stable disease for ≥6 months (N = 3) were seen. Conclusions The combination of crizotinib and dasatinib is safe to administer but tolerability is limited given the high rate of adverse events. Responses and durable stable disease were limited. Further precision therapy approach using this specific combination may be difficult given the toxicity.
Collapse
|
30
|
Planchard D, Smit EF, Groen HJM, Mazieres J, Besse B, Helland Å, Giannone V, D'Amelio AM, Zhang P, Mookerjee B, Johnson BE. Dabrafenib plus trametinib in patients with previously untreated BRAF V600E-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol 2017; 18:1307-1316. [PMID: 28919011 DOI: 10.1016/s1470-2045(17)30679-4] [Citation(s) in RCA: 676] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND BRAFV600E mutation occurs in 1-2% of lung adenocarcinomas and acts as an oncogenic driver. Dabrafenib, alone or combined with trametinib, has shown substantial antitumour activity in patients with previously treated BRAFV600E-mutant metastatic non-small-cell lung cancer (NSCLC). We aimed to assess the activity and safety of dabrafenib plus trametinib treatment in previously untreated patients with BRAFV600E-mutant metastatic NSCLC. METHODS In this phase 2, sequentially enrolled, multicohort, multicentre, non-randomised, open-label study, adults (≥18 years of age) with previously untreated metastatic BRAFV600E-mutant NSCLC were enrolled into cohort C from 19 centres in eight countries within North America, Europe, and Asia. Patients received oral dabrafenib 150 mg twice per day plus oral trametinib 2 mg once per day until disease progression, unacceptable adverse events, consent withdrawal, or death. The primary endpoint was investigator-assessed overall response, defined as the percentage of patients who achieved a confirmed complete response or partial response per Response Evaluation Criteria In Solid Tumors version 1.1. The primary and safety analyses were by intention to treat in the protocol-defined population (previously untreated patients). The study is ongoing, but no longer recruiting patients. This trial is registered with ClinicalTrials.gov, number NCT01336634. FINDINGS Between April 16, 2014, and Dec 28, 2015, 36 patients were enrolled and treated with first-line dabrafenib plus trametinib. Median follow-up was 15·9 months (IQR 7·8-22·0) at the data cutoff (April 28, 2017). The proportion of patients with investigator-assessed confirmed overall response was 23 (64%, 95% CI 46-79), with two (6%) patients achieving a complete response and 21 (58%) a partial response. All patients had one or more adverse event of any grade, and 25 (69%) had one or more grade 3 or 4 event. The most common (occurring in more than two patients) grade 3 or 4 adverse events were pyrexia (four [11%]), alanine aminotransferase increase (four [11%]), hypertension (four [11%]), and vomiting (three [8%]). Serious adverse events occurring in more than two patients included alanine aminotransferase increase (five [14%]), pyrexia (four [11%]), aspartate aminotransferase increase (three [8%]), and ejection fraction decrease (three [8%]). One fatal serious adverse event deemed unrelated to study treatment was reported (cardiorespiratory arrest). INTERPRETATION Dabrafenib plus trametinib represents a new therapy with clinically meaningful antitumour activity and a manageable safety profile in patients with previously untreated BRAFV600E-mutant NSCLC. FUNDING Novartis.
Collapse
Affiliation(s)
| | - Egbert F Smit
- Vrije Universiteit VU Medical Centre, Amsterdam, Netherlands
| | - Harry J M Groen
- University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Julien Mazieres
- Rangueil-Larrey Hospital and Paul Sabatier University, Toulouse, France
| | | | - Åslaug Helland
- Oslo University Hospital, Department of Oncology, Norwegian Radium Hospital, Oslo, Norway
| | | | | | - Pingkuan Zhang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | |
Collapse
|
31
|
Lee CS, Baek J, Han SY. The Role of Kinase Modulators in Cellular Senescence for Use in Cancer Treatment. Molecules 2017; 22:molecules22091411. [PMID: 28841181 PMCID: PMC6151769 DOI: 10.3390/molecules22091411] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022] Open
Abstract
Recently, more than 30 small molecules and eight monoclonal antibodies that modulate kinase signaling have been approved for the treatment of several pathological conditions, including cancer, idiopathic pulmonary fibrosis, and rheumatoid arthritis. Among them, kinase modulators have been a primary focus for use in cancer treatment. Cellular senescence is believed to protect cells from tumorigenesis by irreversibly halting cell cycle progression and avoiding the growth of damaged cells and tissues. Senescence can also contribute to tumor suppression and be utilized as a mechanism by anti-cancer agents. Although the role of kinase modulators in cancer treatment and their effects on senescence in tumor development have been extensively studied, the relationship between kinase modulators for cancer treatment and senescence has not been fully discussed. In this review, we discuss the pro- and anti-tumorigenesis functions of senescence and summarize the key roles of kinase modulators in the regulation of senescence against tumors.
Collapse
Affiliation(s)
- Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju, Gyeongnam 52828, Korea.
| | - Juhwa Baek
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju, Gyeongnam 52828, Korea.
| | - Sun-Young Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju, Gyeongnam 52828, Korea.
| |
Collapse
|
32
|
Schallenberg S, Merkelbach-Bruse S, Buettner R. Lung cancer as a paradigm for precision oncology in solid tumours. Virchows Arch 2017; 471:221-233. [PMID: 28730537 DOI: 10.1007/s00428-017-2183-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/19/2017] [Accepted: 06/25/2017] [Indexed: 02/06/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death in the western world. However, the combination of molecular genotyping and subsequent systematic treatment of decoded target structures is a prime example of precision oncology in solid tumours. In this review, current targets of approved therapeutics and potential targets in clinical and preclinical trials are outlined. Furthermore, immune checkpoint inhibitors, as promising new therapeutic options, which have already been applied successfully in cases of lung cancer, are introduced. A major issue of targeted treatment of lung tumours is the persistent development of resistance. The underlying mechanisms and established and potentially applicable alternative therapeutic approaches are described. In this process of precision oncology, immunohistochemistry, fluorescence in situ hybridization, and parallel sequencing are crucial diagnostic tools.
Collapse
Affiliation(s)
- Simon Schallenberg
- Institute of Pathology, University Hospital and Center for Integrated Oncology Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, University Hospital and Center for Integrated Oncology Cologne, Kerpener Straße 62, 50937, Cologne, Germany.
| | - Reinhard Buettner
- Institute of Pathology, University Hospital and Center for Integrated Oncology Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| |
Collapse
|
33
|
Yang M, Topaloglu U, Petty WJ, Pagni M, Foley KL, Grant SC, Robinson M, Bitting RL, Thomas A, Alistar AT, Desnoyers RJ, Goodman M, Albright C, Porosnicu M, Vatca M, Qasem SA, DeYoung B, Kytola V, Nykter M, Chen K, Levine EA, Staren ED, D’Agostino RB, Petro RM, Blackstock W, Powell BL, Abraham E, Pasche B, Zhang W. Circulating mutational portrait of cancer: manifestation of aggressive clonal events in both early and late stages. J Hematol Oncol 2017; 10:100. [PMID: 28472989 PMCID: PMC5418716 DOI: 10.1186/s13045-017-0468-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/20/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Solid tumors residing in tissues and organs leave footprints in circulation through circulating tumor cells (CTCs) and circulating tumor DNAs (ctDNA). Characterization of the ctDNA portraits and comparison with tumor DNA mutational portraits may reveal clinically actionable information on solid tumors that is traditionally achieved through more invasive approaches. METHODS We isolated ctDNAs from plasma of patients of 103 lung cancer and 74 other solid tumors of different tissue origins. Deep sequencing using the Guardant360 test was performed to identify mutations in 73 clinically actionable genes, and the results were associated with clinical characteristics of the patient. The mutation profiles of 37 lung cancer cases with paired ctDNA and tumor genomic DNA sequencing were used to evaluate clonal representation of tumor in circulation. Five lung cancer cases with longitudinal ctDNA sampling were monitored for cancer progression or response to treatments. RESULTS Mutations in TP53, EGFR, and KRAS genes are most prevalent in our cohort. Mutation rates of ctDNA are similar in early (I and II) and late stage (III and IV) cancers. Mutation in DNA repair genes BRCA1, BRCA2, and ATM are found in 18.1% (32/177) of cases. Patients with higher mutation rates had significantly higher mortality rates. Lung cancer of never smokers exhibited significantly higher ctDNA mutation rates as well as higher EGFR and ERBB2 mutations than ever smokers. Comparative analysis of ctDNA and tumor DNA mutation data from the same patients showed that key driver mutations could be detected in plasma even when they were present at a minor clonal population in the tumor. Mutations of key genes found in the tumor tissue could remain in circulation even after frontline radiotherapy and chemotherapy suggesting these mutations represented resistance mechanisms. Longitudinal sampling of five lung cancer cases showed distinct changes in ctDNA mutation portraits that are consistent with cancer progression or response to EGFR drug treatment. CONCLUSIONS This study demonstrates that ctDNA mutation rates in the key tumor-associated genes are clinical parameters relevant to smoking status and mortality. Mutations in ctDNA may serve as an early detection tool for cancer. This study quantitatively confirms the hypothesis that ctDNAs in circulation is the result of dissemination of aggressive tumor clones and survival of resistant clones. This study supports the use of ctDNA profiling as a less-invasive approach to monitor cancer progression and selection of appropriate drugs during cancer evolution.
Collapse
Affiliation(s)
- Meng Yang
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, 300060 Tianjin, China
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Umit Topaloglu
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - W. Jeffrey Petty
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Matthew Pagni
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Kristie L. Foley
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Stefan C. Grant
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Mac Robinson
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Rhonda L. Bitting
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Alexandra Thomas
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Angela T. Alistar
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Rodwige J. Desnoyers
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Michael Goodman
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Carol Albright
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Mercedes Porosnicu
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Mihaela Vatca
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Shadi A. Qasem
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Laboratory Medicine and Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Barry DeYoung
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Laboratory Medicine and Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Ville Kytola
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Institute for Biosciences and Medical Technology, University of Tampere, 33520 Tampere, Finland
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Matti Nykter
- Institute for Biosciences and Medical Technology, University of Tampere, 33520 Tampere, Finland
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, 300060 Tianjin, China
| | - Edward A. Levine
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of General Surgery-Section of Surgical Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Edgar D. Staren
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of General Surgery-Section of Surgical Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Ralph B. D’Agostino
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Robin M. Petro
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - William Blackstock
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Bayard L. Powell
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Edward Abraham
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Boris Pasche
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Department of Internal Medicine-Section of Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Wei Zhang
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Medical Center Blvd., Winston-Salem, NC 27157 USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- Cancer Genomics and Precision Medicine, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157 USA
| |
Collapse
|
34
|
Abstract
Treatment options for patients with metastatic melanoma, and especially BRAF-mutant melanoma, have changed dramatically in the past 5 years, with the FDA approval of eight new therapeutic agents. During this period, the treatment paradigm for BRAF-mutant disease has evolved rapidly: the standard-of-care BRAF-targeted approach has shifted from single-agent BRAF inhibition to combination therapy with a BRAF and a MEK inhibitor. Concurrently, immunotherapy has transitioned from cytokine-based treatment to antibody-mediated blockade of the cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and, now, the programmed cell-death protein 1 (PD-1) immune checkpoints. These changes in the treatment landscape have dramatically improved patient outcomes, with the median overall survival of patients with advanced-stage melanoma increasing from approximately 9 months before 2011 to at least 2 years - and probably longer for those with BRAF-V600-mutant disease. Herein, we review the clinical trial data that established the standard-of-care treatment approaches for advanced-stage melanoma. Mechanisms of resistance and biomarkers of response to BRAF-targeted treatments and immunotherapies are discussed, and the contrasting clinical benefits and limitations of these therapies are explored. We summarize the state of the field and outline a rational approach to frontline-treatment selection for each individual patient with BRAF-mutant melanoma.
Collapse
|
35
|
Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors. Nat Commun 2017; 8:14262. [PMID: 28186126 PMCID: PMC5309787 DOI: 10.1038/ncomms14262] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 12/13/2016] [Indexed: 12/12/2022] Open
Abstract
Colorectal carcinoma represents a heterogeneous entity, with only a fraction of the tumours responding to available therapies, requiring a better molecular understanding of the disease in precision oncology. To address this challenge, the OncoTrack consortium recruited 106 CRC patients (stages I–IV) and developed a pre-clinical platform generating a compendium of drug sensitivity data totalling >4,000 assays testing 16 clinical drugs on patient-derived in vivo and in vitro models. This large biobank of 106 tumours, 35 organoids and 59 xenografts, with extensive omics data comparing donor tumours and derived models provides a resource for advancing our understanding of CRC. Models recapitulate many of the genetic and transcriptomic features of the donors, but defined less complex molecular sub-groups because of the loss of human stroma. Linking molecular profiles with drug sensitivity patterns identifies novel biomarkers, including a signature outperforming RAS/RAF mutations in predicting sensitivity to the EGFR inhibitor cetuximab. The heterogeneity of colorectal cancer has important clinical and therapeutic implications. Here the authors analysed the responses of a large biobank of organoids and xenografts derived from colorectal patients to a panel of clinically relevant therapeutic agents to identify genes signatures associated with drug response.
Collapse
|
36
|
Martinelli E, Morgillo F, Troiani T, Ciardiello F. Cancer resistance to therapies against the EGFR-RAS-RAF pathway: The role of MEK. Cancer Treat Rev 2016; 53:61-69. [PMID: 28073102 DOI: 10.1016/j.ctrv.2016.12.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022]
Abstract
The mitogen-activated protein kinases (MAPKs) mediate intracellular signals activated by a wide variety of extracellular stimuli. The activation of the RAS-RAF-MEK-MAPK cascade culminates in the regulation of gene transcription promoting cancer cell proliferation, survival, migration and angiogenesis. MEK (mitogen-activated protein kinase kinase-MAPKK) 1/2 is a transducer of the growth factor receptor-RAS-RAF-MAPK signalling cascade and plays a relevant role in development and progression of human cancers, such as colorectal cancer (CRC), non small cell lung cancer (NSCLC). Direct inhibition of MEK is a promising strategy and several inhibitors are currently under evaluation in clinical trials showing initial clinical activity in different tumours. MEK activation, by different genetic mechanisms, has been described for both intrinsic and acquired resistance to drugs targeting the EGFR (Epidermal Growth Factor Receptor)-RAS-RAF pathway in CRC, NSCLC. Combination therapies with chemotherapy and/or with molecular targeted agents are warranted and biomarkers studies are needed to identify those tumours dependent on MEK signalling.
Collapse
Affiliation(s)
- Erika Martinelli
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università degli Studi della Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Napoli, Italy.
| | - Floriana Morgillo
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università degli Studi della Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Napoli, Italy
| | - Teresa Troiani
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università degli Studi della Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Napoli, Italy
| | - Fortunato Ciardiello
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università degli Studi della Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Napoli, Italy
| |
Collapse
|
37
|
Dasatinib induces DNA damage and activates DNA repair pathways leading to senescence in non-small cell lung cancer cell lines with kinase-inactivating BRAF mutations. Oncotarget 2016; 7:565-79. [PMID: 26623721 PMCID: PMC4808018 DOI: 10.18632/oncotarget.6376] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/14/2015] [Indexed: 12/31/2022] Open
Abstract
Improved therapies are greatly needed for non-small cell lung cancer (NSCLC) that does not harbor targetable kinase mutations or translocations. We previously demonstrated that NSCLC cells that harbor kinase-inactivating BRAF mutations (KIBRAF) undergo senescence when treated with the multitargeted kinase inhibitor dasatinib. Similarly, treatment with dasatinib resulted in a profound and durable response in a patient with KIBRAF NSCLC. However, no canonical pathways explain dasatinib-induced senescence in KIBRAF NSCLC. To investigate the underlying mechanism, we used 2 approaches: gene expression and reverse phase protein arrays. Both approaches showed that DNA repair pathways were differentially modulated between KIBRAF NSCLC cells and those with wild-type (WT) BRAF. Consistent with these findings, dasatinib induced DNA damage and activated DNA repair pathways leading to senescence only in the KIBRAF cells. Moreover, dasatinib-induced senescence was dependent on Chk1 and p21, proteins known to mediate DNA damage-induced senescence. Dasatinib also led to a marked decrease in TAZ but not YAP protein levels. Overexpression of TAZ inhibited dasatinib-induced senescence. To investigate other vulnerabilities in KIBRAF NSCLC cells, we compared the sensitivity of these cells with that of WTBRAF NSCLC cells to 79 drugs and identified a pattern of sensitivity to EGFR and MEK inhibitors in the KIBRAF cells. Clinically approved EGFR and MEK inhibitors, which are better tolerated than dasatinib, could be used to treat KIBRAF NSCLC. Our novel finding that dasatinib induced DNA damage and subsequently activated DNA repair pathways leading to senescence in KIBRAF NSCLC cells represents a unique vulnerability with potential clinical applications.
Collapse
|
38
|
McIntyre JB, Rambau PF, Chan A, Yap S, Morris D, Nelson GS, Köbel M. Molecular alterations in indolent, aggressive and recurrent ovarian low-grade serous carcinoma. Histopathology 2016; 70:347-358. [DOI: 10.1111/his.13071] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/29/2016] [Indexed: 02/04/2023]
Affiliation(s)
- John B McIntyre
- Translational Laboratory; Tom Baker Cancer Centre; Department of Oncology; University of Calgary; Calgary Alberta Canada
| | - Peter F Rambau
- Department of Pathology; Catholic University of Health and Allied Sciences-Bugando; Mwanza Tanzania
- Department of Pathology and Laboratory Medicine; Calgary Laboratory Services/Alberta Health Services and University of Calgary; Calgary Alberta Canada
| | - Angela Chan
- Translational Laboratory; Tom Baker Cancer Centre; Department of Oncology; University of Calgary; Calgary Alberta Canada
| | - Sidney Yap
- Department of Pathology and Laboratory Medicine; Calgary Laboratory Services/Alberta Health Services and University of Calgary; Calgary Alberta Canada
| | - Don Morris
- Translational Laboratory; Tom Baker Cancer Centre; Department of Oncology; University of Calgary; Calgary Alberta Canada
| | - Gregg S Nelson
- Department of Gynecological Oncology; Tom Baker Cancer Centre; University of Calgary; Calgary Alberta Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine; Calgary Laboratory Services/Alberta Health Services and University of Calgary; Calgary Alberta Canada
| |
Collapse
|
39
|
Chen YB, Xu J, Skanderup AJ, Dong Y, Brannon AR, Wang L, Won HH, Wang PI, Nanjangud GJ, Jungbluth AA, Li W, Ojeda V, Hakimi AA, Voss MH, Schultz N, Motzer RJ, Russo P, Cheng EH, Giancotti FG, Lee W, Berger MF, Tickoo SK, Reuter VE, Hsieh JJ. Molecular analysis of aggressive renal cell carcinoma with unclassified histology reveals distinct subsets. Nat Commun 2016; 7:13131. [PMID: 27713405 PMCID: PMC5059781 DOI: 10.1038/ncomms13131] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinomas with unclassified histology (uRCC) constitute a significant portion of aggressive non-clear cell renal cell carcinomas that have no standard therapy. The oncogenic drivers in these tumours are unknown. Here we perform a molecular analysis of 62 high-grade primary uRCC, incorporating targeted cancer gene sequencing, RNA sequencing, single-nucleotide polymorphism array, fluorescence in situ hybridization, immunohistochemistry and cell-based assays. We identify recurrent somatic mutations in 29 genes, including NF2 (18%), SETD2 (18%), BAP1 (13%), KMT2C (10%) and MTOR (8%). Integrated analysis reveals a subset of 26% uRCC characterized by NF2 loss, dysregulated Hippo–YAP pathway and worse survival, whereas 21% uRCC with mutations of MTOR, TSC1, TSC2 or PTEN and hyperactive mTORC1 signalling are associated with better clinical outcome. FH deficiency (6%), chromatin/DNA damage regulator mutations (21%) and ALK translocation (2%) distinguish additional cases. Altogether, this study reveals distinct molecular subsets for 76% of our uRCC cohort, which could have diagnostic and therapeutic implications. A subset of renal cell carcinomas have uncertain histology and are aggressive in nature. Here, the authors examine this group of unclassified renal cancers using genomics techniques and identify further subclasses of the tumours that have differing prognoses.
Collapse
Affiliation(s)
- Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Jianing Xu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Anders Jacobsen Skanderup
- Computational Biology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yiyu Dong
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - A Rose Brannon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Lu Wang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Helen H Won
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Patricia I Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Gouri J Nanjangud
- Molecular Cytogenetics Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Achim A Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Wei Li
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Virginia Ojeda
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Martin H Voss
- Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Nikolaus Schultz
- Computational Biology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Robert J Motzer
- Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Paul Russo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Emily H Cheng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Filippo G Giancotti
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - William Lee
- Computational Biology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Satish K Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - James J Hsieh
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Department of Medicine, Weill Cornell Medical College, 1300 York Ave, New York, New York 10065, USA
| |
Collapse
|
40
|
Abstract
BACKGROUND Several subsets of non-small-cell lung cancer (NSCLC) are defined by molecular alterations acting as tumor drivers, some of them being currently therapeutically actionable. The rat sarcoma (RAS)-rapidly accelerated fibrosarcoma (RAF)-mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway constitutes an attractive potential target, as v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations occur in 2-4% of NSCLC adenocarcinoma. METHODS Here, we review the latest clinical data on BRAF serine/threonine kinase inhibitors in NSCLC. RESULTS Treatment of V600E BRAF-mutated NSCLC with BRAF inhibitor monotherapy demonstrated encouraging antitumor activity. Combination of BRAF and MEK inhibitors using dabrafenib and trametinib is under evaluation. Preliminary data suggest superior efficacy compared with BRAF inhibitor monotherapy. CONCLUSION Targeting BRAF alterations represents a promising new therapeutic approach for a restricted subset of oncogene-addicted NSCLC. Prospect ive trials refining this strategy are ongoing. A next step will probably aim at combining BRAF inhibitors and immunotherapy or alternatively improve a multilevel mitogen-activated protein kinase (MAPK) pathway blockade by combining with ERK inhibitors.
Collapse
|
41
|
Tafe LJ, Pierce KJ, Peterson JD, de Abreu F, Memoli VA, Black CC, Pettus JR, Marotti JD, Gutmann EJ, Liu X, Shirai K, Dragnev KH, Amos CI, Tsongalis GJ. Clinical Genotyping of Non-Small Cell Lung Cancers Using Targeted Next-Generation Sequencing: Utility of Identifying Rare and Co-mutations in Oncogenic Driver Genes. Neoplasia 2016; 18:577-83. [PMID: 27659017 PMCID: PMC5031899 DOI: 10.1016/j.neo.2016.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 11/15/2022]
Abstract
Detection of somatic mutations in non-small cell lung cancers (NSCLCs), especially adenocarcinomas, is important for directing patient care when targeted therapy is available. Here, we present our experience with genotyping NSCLC using the Ion Torrent Personal Genome Machine (PGM) and the AmpliSeq Cancer Hotspot Panel v2. We tested 453 NSCLC samples from 407 individual patients using the 50 gene AmpliSeq Cancer Hotspot Panel v2 from May 2013 to July 2015. Using 10 ng of DNA, up to 11 samples were simultaneously sequenced on the Ion Torrent PGM (316 and 318 chips). We identified variants with the Ion Torrent Variant Caller Plugin, and Golden Helix's SVS software was used for annotation and prediction of the significance of the variants. Three hundred ninety-eight samples were successfully sequenced (12.1% failure rate). In all, 633 variants in 41 genes were detected with a median of 2 (range of 0 to 7) variants per sample. Mutations detected in BRAF, EGFR, ERBB2, KRAS, NRAS, and PIK3CA were considered potentially actionable and were identified in 237 samples, most commonly in KRAS (37.9%), EGFR (11.1%), BRAF (4.8%), and PIK3CA (4.3%). In our patient population, all mutations in EGFR, KRAS, and BRAF were mutually exclusive. The Ion Torrent Ampliseq technology can be utilized on small biopsy and cytology specimens, requires very little input DNA, and can be applied in clinical laboratories for genotyping of NSCLC. This targeted next-generation sequencing approach allows for detection of common and also rare mutations that are clinically actionable in multiple patients simultaneously.
Collapse
Affiliation(s)
- Laura J Tafe
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH.
| | - Kirsten J Pierce
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jason D Peterson
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Francine de Abreu
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Vincent A Memoli
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Candice C Black
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jason R Pettus
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jonathan D Marotti
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Edward J Gutmann
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Xiaoying Liu
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Keisuke Shirai
- Department of Oncology, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Konstantin H Dragnev
- Department of Oncology, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Christopher I Amos
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH; Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Gregory J Tsongalis
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| |
Collapse
|
42
|
Noeparast A, Teugels E, Giron P, Verschelden G, De Brakeleer S, Decoster L, De Grève J. Non-V600 BRAF mutations recurrently found in lung cancer predict sensitivity to the combination of Trametinib and Dabrafenib. Oncotarget 2016; 8:60094-60108. [PMID: 28947956 PMCID: PMC5601124 DOI: 10.18632/oncotarget.11635] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 06/09/2016] [Indexed: 12/12/2022] Open
Abstract
Approximately half of BRAF-mutated Non-small cell lung cancers (NSCLCs) harbor a non-V600 BRAF mutation, accounting for ∼40,000 annual deaths worldwide. Recent studies have revealed the benefits of combined targeted therapy with a RAF-inhibitor (Dabrafenib) and a MEK-inhibitor (Trametinib) in treating V600 BRAF mutant cancers, including NSCLC. In contrast, sensitivity of non-V600 BRAF mutations to these inhibitors is not documented. Non-V600 mutations can either increase or impair BRAF kinase activity. However, impaired BRAF kinases can still activate the ERK pathway in a CRAF-dependent manner. Herein, beyond describing a cohort of BRAF mutant NSCLC patients and functionally analyzing 13 tumor-derived BRAF mutations, we demonstrate that both types of non-V600 BRAF mutations can be sensitive to clinically relevant doses of Dabrafenib and Trametinib in HEK293T cells, in lung epithelial cellular model (BEAS-2B) and in human cancer cell lines harboring non-V600 BRAF mutations. ERK activity induced by both types of these mutations is further reduced by combinatorial drug treatment. Moreover, the combination leads to more prolonged ERK inhibition and has anti-proliferative and pro-apoptotic effects in cells harboring both types of non-V600 BRAF mutations. This study provides a basis for the clinical exploration of non-V600 BRAF mutant lung cancers upon treatment with Trametinib and Dabrafenib.
Collapse
Affiliation(s)
- Amir Noeparast
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Erik Teugels
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Philippe Giron
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gil Verschelden
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sylvia De Brakeleer
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lore Decoster
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jacques De Grève
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
43
|
Wang Y, Singh R, Wang L, Nilsson M, Goonatilake R, Tong P, Li L, Giri U, Villalobos P, Mino B, Rodriguez-Canales J, Wistuba I, Wang J, Heymach JV, Johnson FM. Polo-like kinase 1 inhibition diminishes acquired resistance to epidermal growth factor receptor inhibition in non-small cell lung cancer with T790M mutations. Oncotarget 2016; 7:47998-48010. [PMID: 27384992 PMCID: PMC5216995 DOI: 10.18632/oncotarget.10332] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/07/2016] [Indexed: 01/22/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are effective against non-small cell lung cancer (NSCLC) with activating EGFR mutations, but resistance is inevitable. Mechanisms of acquired resistance include T790M mutations and epithelial-mesenchymal transition (EMT). One potential strategy for overcoming this resistance is the inhibition of polo-like kinase 1 (PLK1) based on our previous studies showing that mesenchymal NSCLC cell lines are more sensitive to PLK1 inhibition than epithelial cell lines. To determine the extent to which PLK1 inhibition overcomes EGFR TKI resistance we measured the effects of the PLK1 inhibitor volasertib alone and in combination with the EGFR inhibitor erlotinib in vitro and in vivo in EGFR mutant NSCLC cell lines with acquired resistance to erlotinib. Two erlotinib-resistant cell lines that underwent EMT had higher sensitivity to volasertib, which caused G2/M arrest and apoptosis, than their parental cells. In all NSCLC cell lines with T790M mutations, volasertib markedly reduced erlotinib resistance. All erlotinib-resistant NSCLC cell lines with T790M mutations had higher sensitivity to erlotinib plus volasertib than to erlotinib alone, and the combination treatment caused G2/M arrest and apoptosis. Compared with either agent alone, the combination treatment also caused significantly more DNA damage and greater reductions in tumor size. Our results suggest that PLK1 inhibition is clinically effective against NSCLC that becomes resistant to EGFR inhibition through EMT or the acquisition of a T790M mutation. These results uncover new functions of PLK1 inhibition in the treatment of NSCLC with acquired resistance to EGFR TKIs.
Collapse
Affiliation(s)
- Yuehong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ratnakar Singh
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Liguang Wang
- Institute of Oncology, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, China
| | - Monique Nilsson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ruchitha Goonatilake
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pan Tong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lerong Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Uma Giri
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pamela Villalobos
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Barbara Mino
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ignacio Wistuba
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Faye M. Johnson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
44
|
Targeted sequencing of refractory myeloma reveals a high incidence of mutations in CRBN and Ras pathway genes. Blood 2016; 128:1226-33. [PMID: 27458004 DOI: 10.1182/blood-2016-02-698092] [Citation(s) in RCA: 199] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/28/2016] [Indexed: 12/20/2022] Open
Abstract
In this study, targeted sequencing to screen 50 multidrug refractory multiple myeloma (rMM) patients was performed by using the Multiple Myeloma Mutation Panel. Patients were pretreated with both immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs), and 88%, 78%, and 68% were refractory to an IMiD, a PI, or both, respectively. The majority of patients had progressive (82%) or refractory (78%) disease immediately before sampling, with 43% being IMiD refractory and 46% being PI refractory in the most recent line of therapy. Compared with newly diagnosed MM, an increased prevalence of mutations in the Ras pathway genes KRAS, NRAS, and/or BRAF (72%), as well as TP53 (26%), CRBN (12%), and CRBN pathway genes (10%) was observed. Longitudinal analyses performed in 3 patients with CRBN mutations at time of IMiD resistance confirmed that these mutations were undetectable at earlier, IMiD-sensitive time points. Furthermore, the functional introduction of these mutations in MM cells conferred lenalidomide resistance in vitro. These data indicate a differential genetic landscape in rMM associated with drug response.
Collapse
|
45
|
Somatic mutations in histiocytic sarcoma identified by next generation sequencing. Virchows Arch 2016; 469:233-41. [DOI: 10.1007/s00428-016-1965-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 04/24/2016] [Accepted: 05/17/2016] [Indexed: 01/04/2023]
|
46
|
Desai A, Menon SP, Dy GK. Alterations in genes other than EGFR/ALK/ROS1 in non-small cell lung cancer: trials and treatment options. Cancer Biol Med 2016; 13:77-86. [PMID: 27144064 PMCID: PMC4850130 DOI: 10.28092/j.issn.2095-3941.2016.0008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
During the last decade, we have seen tremendous progress in the therapy of lung cancer. Discovery of actionable mutations in EGFR and translocations in ALK and ROS1 have identified subsets of patients with excellent tumor response to oral targeted agents with manageable side effects. In this review, we highlight treatment options including corresponding clinical trials for oncogenic alterations affecting the receptor tyrosine kinases MET, FGFR, NTRK, RET, HER2, HER3, and HER4 as well as components of the RAS-RAF-MEK signaling pathway.
Collapse
Affiliation(s)
- Arpita Desai
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Smitha P Menon
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226-0509, USA
| | - Grace K Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
47
|
Ferrarotto R, Goonatilake R, Yoo SY, Tong P, Giri U, Peng S, Minna J, Girard L, Wang Y, Wang L, Li L, Diao L, Peng DH, Gibbons DL, Glisson BS, Heymach JV, Wang J, Byers LA, Johnson FM. Epithelial-Mesenchymal Transition Predicts Polo-Like Kinase 1 Inhibitor-Mediated Apoptosis in Non-Small Cell Lung Cancer. Clin Cancer Res 2016; 22:1674-1686. [PMID: 26597303 PMCID: PMC4818738 DOI: 10.1158/1078-0432.ccr-14-2890] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 11/01/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE To identify new therapeutic targets for non-small cell lung cancer (NSCLC), we systematically searched two cancer cell line databases for sensitivity data on a broad range of drugs. We identified polo-like kinase 1 (PLK1) as the most promising target for further investigation based on a subset of sensitive NSCLC cell lines and inhibitors that were in advanced clinical development. EXPERIMENTAL DESIGN To identify potential biomarkers of response of NSCLC to PLK1 inhibition and mechanisms of PLK1 inhibitor-induced apoptosis, integrated analysis of gene and protein expression, gene mutations, and drug sensitivity was performed using three PLK1 inhibitors (volasertib, BI2536, and GSK461364) with a large panel of NSCLC cell lines. RESULTS The NSCLC cell lines had different sensitivities to PLK1 inhibition, with a minority demonstrating sensitivity to all three inhibitors. PLK1 inhibition led to G2-M arrest, but only treatment-sensitive cell lines underwent substantial apoptosis following PLK1 inhibition. NSCLC lines with high epithelial-mesenchymal transition (EMT) gene signature scores (mesenchymal cell lines) were more sensitive to PLK1 inhibition than epithelial lines (P< 0.02). Likewise, proteomic profiling demonstrated that E-cadherin expression was higher in the resistant cell lines than in the sensitive ones (P< 0.01). Induction of an epithelial phenotype by expression of the miRNA miR-200 increased cellular resistance to PLK1 inhibition. Also, KRAS mutation and alterations in the tight-junction, ErbB, and Rho signaling pathways correlated with drug response of NSCLC. CONCLUSIONS In this first reported large-scale integrated analysis of PLK1 inhibitor sensitivity, we demonstrated that EMT leads to PLK1 inhibition sensitivity of NSCLC cells. Our findings have important clinical implications for mesenchymal NSCLC, a significant subtype of the disease that is associated with resistance to currently approved targeted therapies.
Collapse
Affiliation(s)
- Renata Ferrarotto
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ruchitha Goonatilake
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Suk Young Yoo
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pan Tong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Uma Giri
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shaohua Peng
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Minna
- Hamon Cancer Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Luc Girard
- Hamon Cancer Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yuehong Wang
- Department of Respiratory Medicine, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liguang Wang
- Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, China
| | - Lerong Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David H. Peng
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Don L. Gibbons
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bonnie S. Glisson
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V. Heymach
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Jing Wang
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren A. Byers
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Faye M. Johnson
- Departments of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
48
|
Caparica R, de Castro G, Gil-Bazo I, Caglevic C, Calogero R, Giallombardo M, Santos ES, Raez LE, Rolfo C. BRAF mutations in non-small cell lung cancer: has finally Janus opened the door? Crit Rev Oncol Hematol 2016; 101:32-9. [PMID: 26960735 DOI: 10.1016/j.critrevonc.2016.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/25/2016] [Accepted: 02/23/2016] [Indexed: 01/02/2023] Open
Abstract
B-Raf mutations occur in about 1-2% of non-small cell lung cancers (NSCLC). These mutations generate a permanent activation of the mitogen activated protein kinase (MAPK) pathway, which promotes tumor growth and proliferation. In the present review, we discuss B-Raf mutation epidemiology, diagnostic methods to detect B-Raf mutations, the role of B-Raf as a driver mutation and a potential therapeutic target in NSCLC. The results of clinical trials involving B-Raf or MAPK pathway inhibitors for the treatment of NSCLC are also discussed. Clinical trials evaluating B-Raf inhibitors in BRAF mutated NSCLC patients have shown promising results, and larger prospective studies are warranted to validate these findings. Enrollment of these patients in clinical trials is an interesting strategy to offer a potentially more effective and less toxic targeted therapy.
Collapse
Affiliation(s)
- Rafael Caparica
- Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da USP, São Paulo, Brazil
| | - Gilberto de Castro
- Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da USP, São Paulo, Brazil
| | - Ignacio Gil-Bazo
- Oncology Department, Clínica Universidad de Navarra, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | | | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Marco Giallombardo
- Biopathology and Medical Biotechnology Department, Biology section, University of Palermo, Italy; Phase I - Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital & Center for Oncological Research (CORE), Antwerp University, Belgium
| | - Edgardo S Santos
- Lynn Cancer Institute, Thoracic and Head/Neck Cancer Programs, Florida Atlantic University, Boca Raton, FL, USA
| | - Luis E Raez
- Memorial Cancer Institute, Memorial Health Care System, Florida International University, Miami, FL, USA
| | - Christian Rolfo
- Phase I - Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital & Center for Oncological Research (CORE), Antwerp University, Belgium.
| |
Collapse
|
49
|
Li F, Wang L, Kong R, Sheng J, Cao H, Mancuso J, Xia X, Stephan C, Wong STC. DrugMoaMiner: A computational tool for mechanism of action discovery and personalized drug sensitivity prediction. 2016 IEEE-EMBS INTERNATIONAL CONFERENCE ON BIOMEDICAL AND HEALTH INFORMATICS (BHI) 2016:368-371. [DOI: 10.1109/bhi.2016.7455911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
50
|
Terai H, Tan L, Beauchamp EM, Hatcher JM, Liu Q, Meyerson M, Gray NS, Hammerman PS. Characterization of DDR2 Inhibitors for the Treatment of DDR2 Mutated Nonsmall Cell Lung Cancer. ACS Chem Biol 2015; 10:2687-96. [PMID: 26390252 PMCID: PMC4685943 DOI: 10.1021/acschembio.5b00655] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Despite advances in precision medicine approaches over the past decade, the majority of nonsmall cell lung cancers (NSCLCs) are refractory to treatment with targeted small molecule inhibitors. Previous work has identified mutations in the Discoidin Domain Receptor 2 (DDR2) kinase as potential therapeutic targets in NSCLCs. While DDR2 is potently targeted by several multitargeted kinase inhibitors, most notably dasatinib, toxicity has limited the clinical application of anti-DDR2 therapy. Here, we have characterized compound 1 and other tool compounds demonstrating selectivity for DDR2 and show that while these compounds inhibit DDR2 in lung cancer model systems, they display limited antiproliferative activity in DDR2 mutated cell lines as compared to dual DDR2/SRC inhibitors. We show that DDR2 and SRC are binding partners, that SRC activity is tied to DDR2 activation, and that dual inhibition of both DDR2 and SRC leads to enhanced suppression of DDR2 mutated lung cancer cell lines. These results support the further evaluation of dual SRC/DDR2 targeting in NSCLC, and we report a tool compound, compound 5, which potently inhibits both SRC and DDR2 with a distinct selectivity profile as compared to dasatinib.
Collapse
Affiliation(s)
- Hideki Terai
- Department
of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
| | - Li Tan
- Department
of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Ellen M. Beauchamp
- Department
of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
| | - John M. Hatcher
- Department
of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Qingsong Liu
- Department
of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Matthew Meyerson
- Department
of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
- Department
of Pathology, Brigham and Women’s Hospital, Boston Massachusetts, United States
- Cancer
Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States
| | - Nathanael S. Gray
- Department
of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Peter S. Hammerman
- Department
of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States
- Cancer
Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States
| |
Collapse
|