Targeting FGFR Signaling in Cancer Mehdi Touat1,2, Ecaterina Ileana1, Sophie Postel-Vinay1,2, Fabrice Andre2,3, and Jean-Charles Soria1,2

Total Page:16

File Type:pdf, Size:1020Kb

Targeting FGFR Signaling in Cancer Mehdi Touat1,2, Ecaterina Ileana1, Sophie Postel-Vinay1,2, Fabrice Andre�2,3, and Jean-Charles Soria1,2 Review Clinical Cancer Research Targeting FGFR Signaling in Cancer Mehdi Touat1,2, Ecaterina Ileana1, Sophie Postel-Vinay1,2, Fabrice Andre2,3, and Jean-Charles Soria1,2 Abstract The fibroblast growth factor signaling pathway (FGFR signal- profiles in early-phase trials. Promising treatment efficacy has ing) is an evolutionary conserved signaling cascade that regulates been observed in different types of malignancies, particularly in several basic biologic processes, including tissue development, tumors harboring aberrant FGFR signaling, thus offering novel angiogenesis, and tissue regeneration. Substantial evidence indi- therapeutic opportunities in the era of precision medicine. The cates that aberrant FGFR signaling is involved in the pathogenesis most exciting challenges now focus on selecting patients who are of cancer. Recent developments of deep sequencing technologies most likely to benefit from these agents, increasing the efficacy of have allowed the discovery of frequent molecular alterations in therapies with the development of novel potent compounds and components of FGFR signaling among several solid tumor types. combination strategies, and overcoming toxicities associated with Moreover, compelling preclinical models have demonstrated the FGFR inhibitors. After examination of the basic and translational oncogenic potential of these aberrations in driving tumor growth, research studies that validated the oncogenic potential of aberrant promoting angiogenesis, and conferring resistance mechanisms FGFR signaling, this review focuses on recent data from clinical to anticancer therapies. Recently, the field of FGFR targeting has trials evaluating FGFR targeting therapies and discusses the chal- exponentially progressed thanks to the development of novel lenges and perspectives for the development of these agents. Clin agents inhibiting FGFs or FGFRs, which had manageable safety Cancer Res; 21(12); 2684–94. Ó2015 AACR. Disclosure of Potential Conflicts of Interest F. Andre is a consultant/advisory board member for Novartis. J.-C. Soria is a consultant/advisory board member for AstraZeneca, Clovis Oncology, EOS, Johnson & Johnson, and Servier. No potential conflicts of interest were disclosed by the other authors. Editor's Disclosures The following editor(s) reported relevant financial relationships: S.E. Bates reports receiving a commercial research grant from Celgene via CRADA with NCI. CME Staff Planners' Disclosures The members of the planning committee have no real or apparent conflicts of interest to disclose. Learning Objectives Upon completion of this activity, the participant should have a better understanding of the biologic rationale for targeting the FGFR signaling pathway in cancer, and of the different approaches currently under clinical development. Acknowledgment of Financial or Other Support This activity does not receive commercial support. Introduction embryonic development, differentiation, proliferation, survival, migration, and angiogenesis. FGFR signaling components are Fibroblast growth factors (FGF) and their receptors (FGFR) frequently altered in human cancer, and several preclinical mod- regulate a wide range of physiologic cellular processes, such as els have provided compelling evidence for the oncogenic poten- tial of aberrant FGFR signaling in carcinogenesis, thereby validat- ing FGFR signaling as an attractive target for cancer treatment. 1 Drug Development Department (DITEP), Gustave Roussy, Villejuif, Depending on the type of genomic aberration and the cellular France. 2University Paris Sud, INSERM U981, Predictive Biomarkers and New Therapeutic Strategies in Oncology,Gustave Roussy,Villejuif, context, the oncogenic potential of dysregulated FGFR signaling France. 3Department of Medical Oncology, Gustave Roussy, Villejuif, ranges from a driver event—responsible for oncogenesis and France. oncogene addiction—to an escape mechanism of secondary Corresponding Author: Fabrice Andre, Gustave Roussy Institute, 114 rue E acquired resistance to other anticancer agents. Vaillant, Villejuif 94800, France. Phone: þ33-1-42-11-43-71; Fax: þ33-1-42-11- 52-74; E-mail: [email protected] FGFR Signaling Pathway doi: 10.1158/1078-0432.CCR-14-2329 FGFRs are transmembrane, receptor tyrosine kinases (RTK) Ó2015 American Association for Cancer Research. consisting of three extracellular immunoglobulin-like domains 2684 Clin Cancer Res; 21(12) June 15, 2015 Downloaded from clincancerres.aacrjournals.org on July 20, 2015. © 2015 American Association for Cancer Research. Targeting FGFR Signaling in Cancer FGF FGF Extracellular FGF FGF FGFR HSPG FGF FGF Plasma membrane SEF FGFRL1 P P SPRY RAS RAF P P PIP2 P P FRS2 P P GRB2 SOS Cytoplasm PLC-γ GAB1 MEK IP3 P P DAG P P STAT PI3K ERK MKP3 PKC AKT Nucleus MKP1 Transcription of target genes Proliferation Resistance to Neoangiogenesis and survival anticancer agents © 2015 American Association for Cancer Research Figure 1. FGFR structure, network, and dysregulation in cancer. FGFRs are transmembrane RTKs consisting of three extracellular immunoglobulin-like domains and one intracellular split tyrosine kinase domain. A complex is formed among FGF, HSPG, and FGFR leading to receptor dimerization, and transphosphorylation of tyrosine kinase domains. Activation of downstream signaling occurs via FRS2, which functions as a key adaptor protein associated with GRB2, resulting in subsequent activation of MAPK and PI3K/AKT signaling pathways. Operating independently from FRS2, phospholipase C-g (PLC-g) binds to a phosphotyrosine at the COOH tail and hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-tri-phosphate (IP3) and diacylglycerol (DAG), thus activating protein kinase C (PKC), which converges with the MAPK pathway. Depending on the cellular context, several other pathways may be activated by FGFRs, including the p38 MAPK and Jun N-terminal kinase pathways, STAT signaling, and ribosomal protein S6 kinase 2 (RSK2). Multiple negative regulators may attenuate signaling at different levels, including FGFRL1, SEF, SPRY, and MAPK phosphatase 1 and 3 (MKP1 and MKP3). Aberrant FGFR signaling may result from (i) increased availability of FGFs (secreted by tumor or stromal cells) leading to ligand-dependent FGFR signaling (autocrine/paracrine loops), or (ii) ligand-independent FGFR signaling when a molecular alteration of an FGFR (mutation, translocation, or amplification) induces a constitutive activation of the kinase domain. and one intracellular split tyrosine kinase domain (1, 2). In FGFs are secreted glycoproteins that are readily sequestered by contrast with the multiple fibroblast growth factor (FGF) genes the extracellular matrix and the cell surface by heparan sulfate encoding 22 functionally distinct ligands, only four different proteoglycans (HPSG), which stabilize the FGF–FGFR interac- FGFRs (FGFR1–4) are known. However, alternative splicing tion by protecting FGFs from protease-mediated degradation. events of FGFR1–3 allow the generation of multiple isoforms, The binding of an FGF to an FGFR leads to receptor dimerization presenting a dramatically variable FGF-binding specificity (3). and transphosphorylation of tyrosine kinase domains (Fig. 1; www.aacrjournals.org Clin Cancer Res; 21(12) June 15, 2015 2685 Downloaded from clincancerres.aacrjournals.org on July 20, 2015. © 2015 American Association for Cancer Research. Touat et al. refs. 4, 5). Activation of downstream signaling occurs via the potential of these alterations, and high sensitivity to FGFR intracellular receptor substrates FGFR substrate 2 (FRS2) and inhibitors (17). phospholipase Cg (PLC-g), leading to subsequent upregulation These preclinical data provide a strong rationale for enrolling of RAS/mitogen-activated protein kinase (MAPK) and phosphoi- patients with tumors harboring FGFR fusions in clinical trials nositide 3-kinase (PI3K)/AKT signaling pathways. Other path- evaluating FGFR inhibitors, and preliminary data from early- ways can be activated, including STAT-dependent signaling phase trials are very encouraging (18). (Fig. 1; refs. 1–5). FGFR-activating mutations. The screening of more than 1,000 Dysregulation of FGFR Signaling in Human exon mutations of protein kinase genes from 210 different malig- fi Malignancies nancies (19) identi ed the FGFR signaling pathway as the most commonly mutated tyrosine kinase signaling pathway. Activating Aberrant FGFR signaling contributes to carcinogenesis in three mutations in FGFRs may result in aberrant FGFR signaling main situations: (i) "driver mutations," where the acquisition of through multiple mechanisms, including the following: (i) somatic molecular alterations directly stimulates cancer cell pro- enhanced activation of the kinase domain; (ii) ligand-indepen- liferation and survival; (ii) neoangiogenesis; and (iii) resistance to dent dimerization of the receptors; and (iii) altered affinity for anticancer agents (1–5). FGF ligands. Urothelial bladder carcinoma has the most established associ- Molecular alterations of the FGFR signaling pathway as driver ation with altered FGFR signaling, with up to 80% of low-grade events: the oncogene addiction phenomenon tumors harboring FGFR mutations and compelling in vivo and in FGFR family fusion genes. Fusion genes are hybrid genes formed by vitro data (Table 1). Comprehensive molecular characterization of the rearrangement of two previously independent genes. They can this cancer (16) revealed a cluster of tumors with papillary mor- occur as a result of translocation, chromosomal inversion,
Recommended publications
  • Chronic Myeloid Leukemia: Mechanisms of Blastic Transformation
    Chronic myeloid leukemia: mechanisms of blastic transformation Danilo Perrotti, … , John Goldman, Tomasz Skorski J Clin Invest. 2010;120(7):2254-2264. https://doi.org/10.1172/JCI41246. Science in Medicine The BCR-ABL1 oncoprotein transforms pluripotent HSCs and initiates chronic myeloid leukemia (CML). Patients with early phase (also known as chronic phase [CP]) disease usually respond to treatment with ABL tyrosine kinase inhibitors (TKIs), although some patients who respond initially later become resistant. In most patients, TKIs reduce the leukemia cell load substantially, but the cells from which the leukemia cells are derived during CP (so-called leukemia stem cells [LSCs]) are intrinsically insensitive to TKIs and survive long term. LSCs or their progeny can acquire additional genetic and/or epigenetic changes that cause the leukemia to transform from CP to a more advanced phase, which has been subclassified as either accelerated phase or blastic phase disease. The latter responds poorly to treatment and is usually fatal. Here, we discuss what is known about the molecular mechanisms leading to blastic transformation of CML and propose some novel therapeutic approaches. Find the latest version: https://jci.me/41246/pdf Science in medicine Chronic myeloid leukemia: mechanisms of blastic transformation Danilo Perrotti,1 Catriona Jamieson,2 John Goldman,3 and Tomasz Skorski4 1Department of Molecular Virology, Immunology and Medical Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA. 2Division of Hematology-Oncology, Department of Internal Medicine, University of California at San Diego, La Jolla, California, USA. 3Department of Haematology, Imperial College at Hammersmith Hospital, London, United Kingdom. 4Department of Microbiology and Immunology, Temple University, Philadelphia, Pennsylvania, USA.
    [Show full text]
  • Targeting Non-Oncogene Addiction for Cancer Therapy
    biomolecules Review Targeting Non-Oncogene Addiction for Cancer Therapy Hae Ryung Chang 1,*,†, Eunyoung Jung 1,†, Soobin Cho 1, Young-Jun Jeon 2 and Yonghwan Kim 1,* 1 Department of Biological Sciences and Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea; [email protected] (E.J.); [email protected] (S.C.) 2 Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; [email protected] * Correspondence: [email protected] (H.R.C.); [email protected] (Y.K.); Tel.: +82-2-710-9552 (H.R.C.); +82-2-710-9552 (Y.K.) † These authors contributed equally. Abstract: While Next-Generation Sequencing (NGS) and technological advances have been useful in identifying genetic profiles of tumorigenesis, novel target proteins and various clinical biomarkers, cancer continues to be a major global health threat. DNA replication, DNA damage response (DDR) and repair, and cell cycle regulation continue to be essential systems in targeted cancer therapies. Although many genes involved in DDR are known to be tumor suppressor genes, cancer cells are often dependent and addicted to these genes, making them excellent therapeutic targets. In this review, genes implicated in DNA replication, DDR, DNA repair, cell cycle regulation are discussed with reference to peptide or small molecule inhibitors which may prove therapeutic in cancer patients. Additionally, the potential of utilizing novel synthetic lethal genes in these pathways is examined, providing possible new targets for future therapeutics. Specifically, we evaluate the potential of TONSL as a novel gene for targeted therapy. Although it is a scaffold protein with no known enzymatic activity, the strategy used for developing PCNA inhibitors can also be utilized to target TONSL.
    [Show full text]
  • 1078-0432.CCR-20-1706.Full.Pdf
    Author Manuscript Published OnlineFirst on June 29, 2020; DOI: 10.1158/1078-0432.CCR-20-1706 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Gastrointestinal stromal tumor: challenges and opportunities for a new decade César Serrano1,2, Suzanne George3 1Sarcoma Translational Research Laboratory, Vall d’Hebron Institute of Oncology, Barcelona, Spain. 2Department of Medical Oncology, Vall d’Hebron University Hospital, Barcelona, Spain. 3Department of Medical Oncology, Sarcoma Center, Dana-Farber Cancer Institute, Boston, US. Running title: Review on gastrointestinal stromal tumor Key words: Avapritinib; circulating tumor DNA; gastrointestinal stromal tumor; imatinib; regorafenib; ripretinib; sunitinib. Financial support: This work was supported by grants (to C.S.) from SARC Career Development Award, FIS ISCIII (PI19/01271), PERIS 2018 (SLT006/17/221) and FERO Foundation. Conflicts of interest: C.S. has received research grants from Deciphera Pharmaceuticals, Bayer AG and Pfizer, Inc; consulting fees (advisory role) from Deciphera Pharmaceuticals and Blueprint 1 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 29, 2020; DOI: 10.1158/1078-0432.CCR-20-1706 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Medicines; payment for lectures from Bayer AG and Blueprint Medicines; and travel grants from Pharmamar, Pfizer, Bayer AG, Novartis and Lilly. S.G. has received research funding to her institution from Blueprint Medicines, Deciphera Pharmaceutical, Bayer AG, Pfizer, Novartis; consulting fees (advisory role) from Blueprint Medicines, Deciphera Pharmaceuticals, Eli Lilly, Bayer AG. Corresponding author: Dr. César Serrano, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital.
    [Show full text]
  • Anti-Aging: Senolytics Or Gerostatics (Unconventional View)
    www.oncotarget.com Oncotarget, 2021, Vol. 12, (No. 18), pp: 1821-1835 Review Anti-aging: senolytics or gerostatics (unconventional view) Mikhail V. Blagosklonny1 1Roswell Park Cancer Institute, Buffalo, NY 14263, USA Correspondence to: Mikhail V. Blagosklonny, email: [email protected], [email protected] Keywords: geroscience; senolytics; hyperfunction theory; aging; sirolimus Received: June 07, 2021 Accepted: July 05, 2021 Published: August 31, 2021 Copyright: © 2021 Blagosklonny. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Senolytics are basically anti-cancer drugs, repurposed to kill senescent cells selectively. It is even more difficult to selectively kill senescent cells than to kill cancer cells. Based on lessons of cancer therapy, here I suggest how to exploit oncogene-addiction and to combine drugs to achieve selectivity. However, even if selective senolytic combinations will be developed, there is little evidence that a few senescent cells are responsible for organismal aging. I also discuss gerostatics, such as rapamycin and other rapalogs, pan-mTOR inhibitors, dual PI3K/mTOR inhibitors, which inhibit growth- and aging-promoting pathways. Unlike senolytics, gerostatics do not kill cells but slow down cellular geroconversion to senescence. Numerous studies demonstrated that inhibition of the mTOR pathways by any means (genetic, pharmacological and dietary) extends lifespan. Currently, only two studies demonstrated that senolytics (fisetin and a combination Dasatinib plus Quercetin) extend lifespan in mice. These senolytics slightly inhibit the mTOR pathway. Thus, life extension by these senolytics can be explained by their slight rapamycin-like (gerostatic) effects.
    [Show full text]
  • Oncogene Addiction As a Rationale for Targeted Anti-Cancer Therapy in Hepatocellular Carcinoma
    호암상 수상 기념특강 Oncogene addiction as a rationale for targeted anti-cancer therapy in hepatocellular carcinoma Dae-Ghon Kim Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Jeonbuk, Korea Abstract The concept of oncogene addiction was first introduced by Bernard Weinstein in 2000, with particular reference to the observation that some cyclin D-overexpressing cancers reverse their malignant phenotype upon cyclin-D deple- tion by means of RNA interference. It postulates that some tumours rely on one single dominant oncogene for growth and survival, so that inhibition of this specific oncogene is sufficient to halt the neoplastic phenotype. A large amount of evidence has proven the pervasive power of this notion, both in basic research and in therapeutic applications. Application of this concept to the clinical setting has achieved variable success in some various cancer types, including chronic myelogeneous leukaemia harbouring the BCR-ABL translocation, Erb2 overexpressing breast cancer, and non-small cell lung cancer harbouring a subset of EGFR mutations (Table1). However, in the face of such a considerable body of knowledge, the intimate molecular mechanisms mediating this phenomenon remain elusive. At the clinical level, successful translation of the oncogene addiction model into the rational and effective design of targeted therapeutics against individual oncoproteins still faces major obstacles, mainly due to the emergence of escape mechanisms and drug resistance. Sorafenib, a tyrosine kinase inhibitor (TKI), has demon- strated clinical efficacy in patients with HCC. Studies in patients with lung, breast, or colorectal cancers indicated that the genetic heterogeneity of cancer cells within a tumor affect its response to therapeutics designed to target specific molecules.
    [Show full text]
  • Emerging Insights of Tumor Heterogeneity and Drug Resistance Mechanisms in Lung Cancer Targeted Therapy Zuan-Fu Lim1,2,3 and Patrick C
    Lim and Ma Journal of Hematology & Oncology (2019) 12:134 https://doi.org/10.1186/s13045-019-0818-2 REVIEW Open Access Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy Zuan-Fu Lim1,2,3 and Patrick C. Ma3* Abstract The biggest hurdle to targeted cancer therapy is the inevitable emergence of drug resistance. Tumor cells employ different mechanisms to resist the targeting agent. Most commonly in EGFR-mutant non-small cell lung cancer, secondary resistance mutations on the target kinase domain emerge to diminish the binding affinity of first- and second-generation inhibitors. Other alternative resistance mechanisms include activating complementary bypass pathways and phenotypic transformation. Sequential monotherapies promise to temporarily address the problem of acquired drug resistance, but evidently are limited by the tumor cells’ ability to adapt and evolve new resistance mechanisms to persist in the drug environment. Recent studies have nominated a model of drug resistance and tumor progression under targeted therapy as a result of a small subpopulation of cells being able to endure the drug (minimal residual disease cells) and eventually develop further mutations that allow them to regrow and become the dominant population in the therapy-resistant tumor. This subpopulation of cells appears to have developed through a subclonal event, resulting in driver mutations different from the driver mutation that is tumor- initiating in the most common ancestor. As such, an understanding of intratumoral heterogeneity—the driving force behind minimal residual disease—is vital for the identification of resistance drivers that results from branching evolution. Currently available methods allow for a more comprehensive and holistic analysis of tumor heterogeneity in that issues associated with spatial and temporal heterogeneity can now be properly addressed.
    [Show full text]
  • Discovery of Dihydroartemisinin and Dasatinib Drug Combination To
    University of Tennessee Health Science Center UTHSC Digital Commons Theses and Dissertations (ETD) College of Graduate Health Sciences 8-2012 Discovery of Dihydroartemisinin and Dasatinib Drug Combination to Cure Pooroutcome BCR- ABL+ Acute Lymphoblastic Leukemia Harpreet Singh University of Tennessee Health Science Center Follow this and additional works at: https://dc.uthsc.edu/dissertations Part of the Medical Sciences Commons, Neoplasms Commons, Pharmaceutical Preparations Commons, and the Pharmaceutics and Drug Design Commons Recommended Citation Singh, Harpreet , "Discovery of Dihydroartemisinin and Dasatinib Drug Combination to Cure Pooroutcome BCR-ABL+ Acute Lymphoblastic Leukemia" (2012). Theses and Dissertations (ETD). Paper 249. http://dx.doi.org/10.21007/etd.cghs.2012.0292. This Dissertation is brought to you for free and open access by the College of Graduate Health Sciences at UTHSC Digital Commons. It has been accepted for inclusion in Theses and Dissertations (ETD) by an authorized administrator of UTHSC Digital Commons. For more information, please contact [email protected]. Discovery of Dihydroartemisinin and Dasatinib Drug Combination to Cure Pooroutcome BCR-ABL+ Acute Lymphoblastic Leukemia Document Type Dissertation Degree Name Doctor of Philosophy (PhD) Program Biomedical Sciences Track Cancer and Developmental Biology Research Advisor Gerard P. Zambetti, Ph.D. (for Richard T. Williams, M.D., Ph.D.) Committee Suzanne J. Baker, Ph.D. R. Kiplin Guy, Ph.D. Sima Jeha, Ph.D. Rajendra S. Raghow, Ph.D. DOI 10.21007/etd.cghs.2012.0292 This dissertation is available at UTHSC Digital Commons: https://dc.uthsc.edu/dissertations/249 Discovery of Dihydroartemisinin and Dasatinib Drug Combination to Cure Poor-outcome BCR-ABL+ Acute Lymphoblastic Leukemia A Dissertation Presented for The Graduate Studies Council The University of Tennessee Health Science Center In Partial Fulfillment Of the Requirements for the Degree Doctor of Philosophy From The University of Tennessee By Harpreet Singh August 2012 Copyright © 2012 by Harpreet Singh.
    [Show full text]
  • Oncogene Addiction As a Foundational Rationale for Targeted Anti-Cancer Therapy: Promises and Perils
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Institutional Research Information System University of Turin Review Oncogene addiction and targeted anti-cancer therapy Oncogene addiction as a foundational rationale for targeted anti-cancer therapy: promises and perils Davide Torti1, Livio Trusolino1* Keywords: DNA damage; drug development; oncogene addiction; targeted therapies; tyrosine kinases DOI 10.1002/emmm.201100176 Received April 15, 2011 / Revised July 07, 2011 / Accepted August 04, 2011 A decade has elapsed since the concept of oncogene addiction was first proposed. It postulates that – despite the diverse array of genetic lesions typical of cancer – some tumours rely on one single dominant oncogene for growth and survival, so that Introduction inhibition of this specific oncogene is sufficient to halt the neoplastic phenotype. A large amount of evidence has proven the pervasive power of this notion, both in Coordinated efforts by the Interna- tional Cancer Genome Consortium basic research and in therapeutic applications. However, in the face of such a and other Institutions have started to considerable body of knowledge, the intimate molecular mechanisms mediating this reveal the complex and heterogeneous phenomenon remain elusive. At the clinical level, successful translation of the ‘genomic landscapes’ of many human oncogene addiction model into the rational and effective design of targeted cancer types with unprecedented accu- therapeutics against individual oncoproteins still faces major obstacles, mainly racy (Hudson et al, 2010; Stratton et al, due to the emergence of escape mechanisms and drug resistance. Here, we offer 2009). A variegate repertoire of genetic an overview of the relevant literature, encompassing both biological aspects and aberrations – including point muta- recent clinical insights.
    [Show full text]
  • Focus on ROS1-Positive Non-Small Cell Lung Cancer (NSCLC): Crizotinib, Resistance Mechanisms and the Newer Generation of Targeted Therapies
    cancers Review Focus on ROS1-Positive Non-Small Cell Lung Cancer (NSCLC): Crizotinib, Resistance Mechanisms and the Newer Generation of Targeted Therapies Alberto D’Angelo 1,* , Navid Sobhani 2 , Robert Chapman 3 , Stefan Bagby 1, Carlotta Bortoletti 4, Mirko Traversini 5, Katia Ferrari 6, Luca Voltolini 7, Jacob Darlow 1 and Giandomenico Roviello 8 1 Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; [email protected] (S.B.); [email protected] (J.D.) 2 Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; [email protected] 3 University College London Hospitals NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK; [email protected] 4 Department of Dermatology, University of Padova, via Vincenzo Gallucci 4, 35121 Padova, Italy; [email protected] 5 Unità Operativa Anatomia Patologica, Ospedale Maggiore Carlo Alberto Pizzardi, AUSL Bologna, Largo Bartolo Nigrisoli 2, 40100 Bologna, Italy; [email protected] 6 Respiratory Medicine, Careggi University Hospital, 50139 Florence, Italy; [email protected] 7 Thoracic Surgery Unit, Careggi University Hospital, Largo Brambilla, 1, 50134 Florence, Italy; luca.voltolini@unifi.it 8 Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy; [email protected] * Correspondence: [email protected] Received: 22 September 2020; Accepted: 5 November 2020; Published: 6 November 2020 Simple Summary: Genetic rearrangements of the ROS1 gene account for up to 2% of NSCLC patients who sometimes develop brain metastasis, resulting in poor prognosis. This review discusses the tyrosine kinase inhibitor crizotinib plus updates and preliminary results with the newer generation of tyrosine kinase inhibitors, which have been specifically conceived to overcome crizotinib resistance, including brigatinib, cabozantinib, ceritinib, entrectinib, lorlatinib and repotrectinib.
    [Show full text]
  • An Integrated Approach to Dissecting Oncogene Addiction Implicates a Myb-Coordinated Self-Renewal Program As Essential for Leukemia Maintenance
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press An integrated approach to dissecting oncogene addiction implicates a Myb-coordinated self-renewal program as essential for leukemia maintenance Johannes Zuber,1,2,8 Amy R. Rappaport,1,3,8 Weijun Luo,1 Eric Wang,1 Chong Chen,1 Angelina V. Vaseva,1,4 Junwei Shi,1,4 Susann Weissmueller,1,3 Christof Fellmann,1 Meredith J. Taylor,1 Martina Weissenboeck,2 Thomas G. Graeber,5 Scott C. Kogan,6 Christopher R. Vakoc,1 and Scott W. Lowe1,3,7,9 1Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA; 2Research Institute of Molecular Pathology (IMP), A-1030 Vienna, Austria; 3Watson School of Biological Sciences, Cold Spring Harbor, New York 11724, USA; 4Genetics Program, Stony Brook University, Stony Brook, New York 11794, USA; 5Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California at Los Angeles, California 90095, USA; 6Department of Laboratory Medicine, University of California at San Francisco, California 94143, USA; 7Howard Hughes Medical Institute, Cold Spring Harbor, New York 11724, USA Although human cancers have complex genotypes and are genomically unstable, they often remain dependent on the continued presence of single-driver mutations—a phenomenon dubbed ‘‘oncogene addiction.’’ Such de- pendencies have been demonstrated in mouse models, where conditional expression systems have revealed that oncogenes able to initiate cancer are often required for tumor maintenance and progression, thus validating the pathways they control as therapeutic targets. Here, we implement an integrative approach that combines genetically defined mouse models, transcriptional profiling, and a novel inducible RNAi platform to characterize cellular programs that underlie addiction to MLL-AF9—a fusion oncoprotein involved in aggressive forms of acute myeloid leukemia (AML).
    [Show full text]
  • The Role of MET in Chemotherapy Resistance
    Oncogene (2021) 40:1927–1941 https://doi.org/10.1038/s41388-020-01577-5 REVIEW ARTICLE The role of MET in chemotherapy resistance 1 1 1 1 Georgina E. Wood ● Helen Hockings ● Danielle M. Hilton ● Stéphanie Kermorgant Received: 16 July 2020 / Revised: 7 November 2020 / Accepted: 18 November 2020 / Published online: 1 February 2021 © The Author(s) 2021. This article is published with open access Abstract Chemotherapy remains the mainstay of treatment in the majority of solid and haematological malignancies. Resistance to cytotoxic chemotherapy is a major clinical problem and substantial research is ongoing into potential methods of overcoming this resistance. One major target, the receptor tyrosine kinase MET, has generated increasing interest with multiple clinical trials in progress. Overexpression of MET is frequently observed in a range of different cancers and is associated with poor prognosis. Studies have shown that MET promotes resistance to targeted therapies, including those targeting EGFR, BRAF and MEK. More recently, several reports suggest that MET also contributes to cytotoxic chemotherapy resistance. Here we review the preclinical evidence of MET’s role in chemotherapy resistance, the mechanisms by which this resistance is mediated and the translational relevance of MET inhibitor therapy for patients with chemotherapy resistant disease. 1234567890();,: 1234567890();,: Introduction activation of ERK1/2, STAT3 and Rac1 from within endosomes [3–5], as opposed to the classical view of RTK The protein MET (also termed Met, c-Met, c-MET), enco- signalling from the plasma membrane only (Fig. 1). Fur- ded by the proto-oncogene MET found on chromosome thermore, oncogenic forms of MET display modified 7q31, is a cell surface receptor tyrosine kinase (RTK) pre- endocytosis and/or ubiquitination leading to enhanced sta- dominantly expressed by epithelial cells [1, 2].
    [Show full text]
  • Update on Molecular Pathology and Role of Liquid Biopsy in Nonsmall Cell Lung Cancer
    EUROPEAN RESPIRATORY REVIEW SERIES ARTICLE P. ABDAYEM AND D. PLANCHARD Update on molecular pathology and role of liquid biopsy in nonsmall cell lung cancer Pamela Abdayem and David Planchard Number 6 in the Series “Thoracic oncology” Edited by Rudolf Huber and Peter Dorfmüller Dept of Cancer Medicine, Thoracic Group, Gustave Roussy Cancer Campus, Villejuif, France. Corresponding author: David Planchard ([email protected]) Shareable abstract (@ERSpublications) Screening for driver alterations in patients with advanced lung adenocarcinoma and those with squamous cell carcinoma and no/little smoking history is mandatory. Liquid biopsy is evolving constantly and will definitely improve outcomes in thoracic oncology. https://bit.ly/2XjuQrD Cite this article as: Abdayem P, Planchard D. Update on molecular pathology and role of liquid biopsy in nonsmall cell lung cancer. Eur Respir Rev 2021; 30: 200294 [DOI: 10.1183/16000617.0294-2020]. Abstract Copyright ©The authors 2021 Personalised medicine, an essential component of modern thoracic oncology, has been evolving continuously ever since the discovery of the epidermal growth factor receptor and its tyrosine kinase This version is distributed under the terms of the Creative inhibitors. Today, screening for driver alterations in patients with advanced lung adenocarcinoma as well as Commons Attribution those with squamous cell carcinoma and no/little history of smoking is mandatory. Multiplex molecular Non-Commercial Licence 4.0. platforms are preferred to sequential molecular testing since they are less time- and tissue-consuming. In For commercial reproduction this review, we present the latest updates on the nine most common actionable driver alterations in rights and permissions contact nonsmall cell lung cancer.
    [Show full text]