1
|
Das S, Berlin J. Systemic Therapy Improvements Will Render Locoregional Treatments Obsolete for Patients with Cancer with Liver Metastases. Hematol Oncol Clin North Am 2025; 39:191-206. [PMID: 39510673 DOI: 10.1016/j.hoc.2024.08.008] [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] [Indexed: 11/15/2024]
Abstract
Hepatic metastases are a major cause of morbidity and mortality for patients with cancer. Apart from curative resection, which offers patients the potential for long-term survival, an array of locoregional therapies, with limited evidence of improving survival, are used to treat them. The authors use examples from the realm of gastrointestinal cancer, largely focusing on the experience of patients with neuroendocrine cancer, hepatobiliary cancer, and colorectal cancer, to suggest that current systemic therapies offer, at minimum, similar survival outcomes for patients compared with these locoregional approaches.
Collapse
Affiliation(s)
- Satya Das
- Department of Medicine, Division of Hematology Oncology, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232, USA.
| | - Jordan Berlin
- Department of Medicine, Division of Hematology Oncology, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232, USA. https://twitter.com/jordanberlin5
| |
Collapse
|
2
|
Ellis H, Braconi C, Valle JW, Bardeesy N. Cholangiocarcinoma Targeted Therapies: Mechanisms of Action and Resistance. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00446-2. [PMID: 39730074 DOI: 10.1016/j.ajpath.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024]
Abstract
Cholangiocarcinoma is an aggressive bile duct malignancy with heterogeneous genomic features. Although most patients receive standard-of-care chemotherapy/immunotherapy, genomic changes that can be targeted with established or emerging therapeutics are common. Accordingly, precision medicine strategies are transforming the next-line treatment for patient subsets. Hotspot IDH1 mutations and activating fibroblast growth factor receptor 2 fusions occur frequently, and small-molecule inhibitors against these alterations are US Food and Drug Administration approved. Translational and basic science studies have elucidated the mechanisms of response and resistance in cholangiocarcinoma, providing insights into these targets that extend to other cancers. Additional US Food and Drug Administration-approved and National Comprehensive Cancer Network guideline-recommended treatments for recurrent genomic changes include BRAF inhibition (BRAF-V600E) and trastumazab deruxtecan (human epidermal growth factor receptor 2 amplification). Furthermore, ongoing clinical trials show promising results with KRAS inhibition (KRAS-codon 12 mutations), PRTM5 inhibition, alone or with methylthioadenosine inhibition (5-methylthioadenosine phosphorylase deletion), and murine double minute 2 inhibition (murine double minute 2 amplification). Despite these advances, the rate, depth, and duration of response to each treatment need improvement. Moreover, many patients do not have currently targetable genotypes. In this review, we examine the clinical efficacy and mechanisms of resistance associated with these treatments, as well as insights into the molecular and biological effects of pathway activation and inhibition, based on study of patient samples and preclinical models. We also explore strategies to overcome resistance and possible precision medicine approaches for additional patient subsets.
Collapse
Affiliation(s)
- Haley Ellis
- Massachusetts General Hospital Cancer Center, Krantz Family Center for Cancer Research, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Chiara Braconi
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom; Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Juan W Valle
- Cholangiocarcinoma Foundation, Herriman, Utah; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Krantz Family Center for Cancer Research, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
3
|
Goyal L, DiToro D, Facchinetti F, Martin EE, Peng P, Baiev I, Iyer R, Maurer J, Reyes S, Zhang K, Majeed U, Berchuck JE, Chen CT, Walmsley C, Pinto C, Vasseur D, Gordan JD, Mody K, Borad M, Karasic T, Damjanov N, Danysh BP, Wehrenberg-Klee E, Kambadakone AR, Saha SK, Hoffman ID, Nelson KJ, Iyer S, Qiang X, Sun C, Wang H, Li L, Javle M, Lin B, Harris W, Zhu AX, Cleary JM, Flaherty KT, Harris T, Shroff RT, Leshchiner I, Parida L, Kelley RK, Fan J, Stone JR, Uboha NV, Hirai H, Sootome H, Wu F, Bensen DC, Hollebecque A, Friboulet L, Lennerz JK, Getz G, Juric D. A Model for Decoding Resistance in Precision Oncology: Acquired Resistance to FGFR inhibitors in Cholangiocarcinoma. Ann Oncol 2024:S0923-7534(24)04990-1. [PMID: 39706336 DOI: 10.1016/j.annonc.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Fibroblast growth factor receptor (FGFR) inhibitors have significantly improved outcomes for patients with FGFR-altered cholangiocarcinoma, leading to their regulatory approval in multiple countries. However, as with many targeted therapies, acquired resistance limits their efficacy. A comprehensive, multimodal approach is crucial to characterizing resistance patterns to FGFR inhibitors. PATIENTS AND METHODS This study integrated data from six investigative strategies: cell-free DNA, tissue biopsy, rapid autopsy, statistical genomics, in vitro and in vivo studies, and pharmacology. We characterized the diversity, clonality, frequency, and mechanisms of acquired resistance to FGFR inhibitors in patients with FGFR-altered cholangiocarcinoma. Clinical samples were analyzed longitudinally as part of routine care across 10 institutions. RESULTS Among 138 patients evaluated, 77 met eligibility, yielding a total of 486 clinical samples. Patients with clinical benefit exhibited a significantly higher rate of FGFR2 kinase domain mutations compared to those without clinical benefit (65% vs 10%, p<0.0001). We identified 26 distinct FGFR2 kinase domain mutations, with 63% of patients harboring multiple. While IC50 assessments indicated strong potency of pan-FGFR inhibitors against common resistance mutations, pharmacokinetic studies revealed that low clinically achievable drug concentrations may underly polyclonal resistance. Molecular brake and gatekeeper mutations predominated, with 94% of patients with FGFR2 mutations exhibiting one or both, whereas mutations at the cysteine residue targeted by covalent inhibitors were rare. Statistical genomics and functional studies demonstrated that mutation frequencies were driven by their combined effects on drug binding and kinase activity rather than intrinsic mutational processes. CONCLUSION Our multimodal analysis led to a model characterizing the biology of acquired resistance, informing the rational design of next-generation FGFR inhibitors. FGFR inhibitors should be small, high-affinity, and selective for specific FGFR family members. Tinengotinib, a novel small molecule inhibitor with these characteristics, exhibited preclinical and clinical activity against key resistance mutations. This integrated approach offers a blueprint for advancing drug resistance research across cancer types.
Collapse
Affiliation(s)
- L Goyal
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA; Department of Medicine, Stanford Cancer Center, Stanford University School of Medicine, Palo Alto, CA.
| | - D DiToro
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - F Facchinetti
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - E E Martin
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - P Peng
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - I Baiev
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - R Iyer
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - J Maurer
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - S Reyes
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - K Zhang
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - U Majeed
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| | - J E Berchuck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - C T Chen
- Department of Medicine, Stanford Cancer Center, Stanford University School of Medicine, Palo Alto, CA
| | - C Walmsley
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - C Pinto
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - D Vasseur
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - J D Gordan
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - K Mody
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| | - M Borad
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ
| | - T Karasic
- University of Pennsylvania Abramson Cancer Center, Philadelphia, PA
| | - N Damjanov
- University of Pennsylvania Abramson Cancer Center, Philadelphia, PA
| | - B P Danysh
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - E Wehrenberg-Klee
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - A R Kambadakone
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - S K Saha
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | | | - S Iyer
- Tyra Biosciences, San Diego, CA
| | - X Qiang
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - C Sun
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - H Wang
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - L Li
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | - M Javle
- MD Anderson Cancer Center, Houston, TX
| | - B Lin
- Virginia Mason Medical Center, Seattle, WA
| | - W Harris
- University of Washington/Fred Hutchinson Cancer Center, Seattle, WA
| | - A X Zhu
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - J M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - K T Flaherty
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | | | - R T Shroff
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - I Leshchiner
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - L Parida
- IBM Research, Yorktown Heights, NY, USA
| | - R K Kelley
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - J Fan
- TransThera Sciences, (US), Inc., Gaithersburg, MD
| | - J R Stone
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - N V Uboha
- University of Wisconsin Carbone Cancer Center, Madison, WI
| | - H Hirai
- Tsukuba Research Institute, Taiho Pharmaceutical Co., Ltd., Japan
| | - H Sootome
- Tsukuba Research Institute, Taiho Pharmaceutical Co., Ltd., Japan
| | - F Wu
- TransThera Sciences (Nanjing), Inc., Nanjing, China
| | | | - A Hollebecque
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - L Friboulet
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - J K Lennerz
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - G Getz
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - D Juric
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| |
Collapse
|
4
|
Goyal L, DiToro D, Hollebecque A, Bridgewater JA, Shimura M, Kano A, Okamura S, Silhavy JL, Wacheck V, Halim A, Meric-Bernstam F. Genomic correlates of response and resistance to the irreversible FGFR1-4 inhibitor futibatinib based on biopsy and circulating tumor DNA profiling. Ann Oncol 2024:S0923-7534(24)04976-7. [PMID: 39672383 DOI: 10.1016/j.annonc.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Futibatinib is the only covalent inhibitor of FGFR1-4 to gain regulatory approval in oncology. In this article, we present genomic analyses of tissue biopsies and circulating tumor DNA (ctDNA) from patients with 1 of nearly 20 tumor types treated with futibatinib in the phase I/II FOENIX study. PATIENTS AND METHODS Eligible patients included those with ctDNA samples collected per protocol at baseline and/or progression on futibatinib in the phase Ib portion of the study for FGF/FGFR-altered advanced solid tumors or the phase II portion of the study for FGFR2 fusion/rearrangement-positive cholangiocarcinoma. Assessments included analytical concordance between tumor and ctDNA analyses for detection of FGFR alterations, association of ctDNA-detected co-occurring genomic alterations with response to futibatinib, and determination of patterns of acquired resistance following progression on futibatinib. RESULTS Among 300 patients treated with futibatinib, 226 were eligible for this analysis, including 139 (62%) with cholangiocarcinoma. Among patients with known FGFR2 fusions/rearrangements, FGFR1 fusions, FGFR3 fusions, or FGFR2 amplifications per tissue analysis, detection rates in ctDNA for these aberrations were 84%, 0%, 11%, and 59%, respectively. Objective response rates on futibatinib were not significantly different between patients with TP53-altered versus -unaltered solid tumors; progression-free survival was reduced in patients with CDKN2B-altered versus -unaltered cholangiocarcinoma (median 4.8 versus 11.0 months; P = 0.03). Acquired resistance to futibatinib was frequently polyclonal and driven by an array of mutations within the relevant FGFR kinase domain, predominantly V565L, V565F, and N550K variants. CONCLUSIONS In this largest and most systematic analysis of acquired resistance to an FGFR inhibitor from prospective clinical trials, emergence of secondary FGFR2 kinase domain mutations was observed in most patients receiving clinical benefit to futibatinib. ctDNA analysis shows clinically relevant potential as a noninvasive method for assessing genomic profiles, identifying patients who may benefit from FGFR inhibitor treatment, and exploring acquired resistance mechanisms.
Collapse
Affiliation(s)
- L Goyal
- Department of Medicine, Stanford Cancer Center, Stanford University School of Medicine, Palo Alto, USA; Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, USA.
| | - D DiToro
- Brigham and Women's Hospital, Boston, USA
| | | | - J A Bridgewater
- University College London Cancer Institute, University College London, London, UK
| | - M Shimura
- Taiho Oncology, Inc., Princeton, USA
| | - A Kano
- Taiho Oncology, Inc., Princeton, USA
| | - S Okamura
- Taiho Oncology, Inc., Princeton, USA
| | | | - V Wacheck
- Taiho Oncology, Inc., Princeton, USA
| | - A Halim
- Taiho Oncology, Inc., Princeton, USA
| | | |
Collapse
|
5
|
Facchinetti F, Loriot Y, Brayé F, Vasseur D, Bahleda R, Bigot L, Barbé R, Nobre C, Combarel D, Michiels S, Italiano A, Smolenschi C, Tselikas L, Scoazec JY, Ponce-Aix S, Besse B, André F, Olaussen KA, Hollebecque A, Friboulet L. Understanding and Overcoming Resistance to Selective FGFR Inhibitors across FGFR2-Driven Malignancies. Clin Cancer Res 2024; 30:4943-4956. [PMID: 39226398 PMCID: PMC7616615 DOI: 10.1158/1078-0432.ccr-24-1834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/26/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
PURPOSE Understanding resistance to selective FGFR inhibitors is crucial to improve the clinical outcomes of patients with FGFR2-driven malignancies. EXPERIMENTAL DESIGN We analyzed sequential ctDNA, ± whole-exome sequencing, or targeted next-generation sequencing on tissue biopsies from patients with tumors harboring activating FGFR2 alterations progressing on pan-FGFR-selective inhibitors, collected in the prospective UNLOCK program. FGFR2::BICC1 Ba/F3 and patient-derived xenograft models were used for functional studies. RESULTS Thirty-six patients were included. In cholangiocarcinoma, at resistance to both reversible inhibitors (e.g., pemigatinib and erdafitinib) and the irreversible inhibitor futibatinib, polyclonal FGFR2 kinase domain mutations were frequent (14/27 patients). Tumors other than cholangiocarcinoma shared the same mutated FGFR2 residues, but polyclonality was rare (1/9 patients). At resistance to reversible inhibitors, 14 residues in the FGFR2 kinase domain were mutated-after futibatinib, only the molecular brake N550 and the gatekeeper V565. Off-target alterations in PI3K/mTOR and MAPK pathways were found in 11 patients, often together with on-target mutations. At progression to a first FGFR inhibitor, 12 patients received futibatinib or lirafugratinib (irreversible inhibitors), with variable clinical outcomes depending on previous resistance mechanisms. Two patients with TSC1 or PIK3CA mutations benefited from everolimus. In cell viability assays on Ba/F3 and in pharmacologic studies on patient-derived xenografts, irreversible inhibitors retained better activity against FGFR2 kinase domain mutations, with lirafugratinib active against the recalcitrant V565L/F/Y. CONCLUSIONS At progression to FGFR inhibitors, FGFR2-driven malignancies are characterized by high intra- and interpatient molecular heterogeneity, particularly in cholangiocarcinoma. Resistance to FGFR inhibitors can be overcome by sequential, molecularly oriented treatment strategies across FGFR2-driven tumors.
Collapse
Affiliation(s)
| | - Yohann Loriot
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département d’Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Floriane Brayé
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Damien Vasseur
- Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
- AMMICa UAR3655/US23, Gustave Roussy, Villejuif, France
| | - Rastislav Bahleda
- Département d’Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
| | - Ludovic Bigot
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Rémy Barbé
- Département de Radiologie, Gustave Roussy, Villejuif, France
| | - Catline Nobre
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - David Combarel
- Service de Pharmacocinétique, Faculté de Pharmacie, Université Paris-Saclay, Orsay, France
- Département de Biologie et Pathologies Médicales, Service de Pharmacologie, Gustave Roussy, Villejuif, France
| | - Stefan Michiels
- Université Paris-Saclay, Inserm, CESP, Villejuif, France
- Gustave Roussy, Office of Biostatistics and Epidemiology, Villejuif, France
| | - Antoine Italiano
- Département d’Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Cristina Smolenschi
- Département d’Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- BIOTHERIS, Department of Interventional Radiology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Jean-Yves Scoazec
- Département de Biologie et Pathologie Médicales, Service de Pathologie Moléculaire, Gustave Roussy, AMMICa, CNRS UAR3655 INSERM US23, Université Paris Saclay, Villejuif, France
| | - Santiago Ponce-Aix
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Fabrice André
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Ken A. Olaussen
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| | - Antoine Hollebecque
- Département d’Innovation Thérapeutique (DITEP), Gustave Roussy, Villejuif, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Luc Friboulet
- Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif, France
| |
Collapse
|
6
|
González-Medina A, Vila-Casadesús M, Gomez-Rey M, Fabregat-Franco C, Sierra A, Tian TV, Castet F, Castillo G, Matito J, Martinez P, Miquel JM, Nuciforo P, Pérez-López R, Macarulla T, Vivancos A. Clinical Value of Liquid Biopsy in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma During Targeted Therapy. Clin Cancer Res 2024; 30:4491-4504. [PMID: 39078735 PMCID: PMC11443220 DOI: 10.1158/1078-0432.ccr-23-3780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/06/2024] [Accepted: 07/25/2024] [Indexed: 10/02/2024]
Abstract
PURPOSE FGFR2 fusions occur in 10% to 15% of patients with intrahepatic cholangiocarcinoma (iCCA), potentially benefiting from FGFR inhibitors (FGFRi). We aimed to assess the feasibility of detecting FGFR2 fusions in plasma and explore plasma biomarkers for managing FGFRi treatment. EXPERIMENTAL DESIGN We conducted a retrospective study in 18 patients with iCCA and known FGFR2 fusions previously identified in tissue samples from prior FGFRi treatment. Both tissue and synchronous plasma samples were analyzed using a custom hybrid capture gene panel with next-generation sequencing (VHIO-iCCA panel) and validated against commercial vendor results. Longitudinal plasma analysis during FGFRi was performed. Subsequently, we explored the correlation between plasma biomarkers, liver enzymes, tumor volume, and clinical outcomes. RESULTS Sixteen patients (88.9%) were positive for FGFR2 fusion events in plasma. Remarkably, the analysis of plasma suggests that lower levels of ctDNA are linked to clinical benefits from targeted therapy and result in improved progression-free survival and overall survival. Higher concentrations of cell-free DNA before FGFRi treatment were linked to worse overall survival, correlating with impaired liver function and indicating compromised cell-free DNA removal by the liver. Additionally, increased ctDNA or the emergence of resistance mutations allowed earlier detection of disease progression compared with standard radiologic imaging methods. CONCLUSIONS VHIO-iCCA demonstrated accurate detection of FGFR2 fusions in plasma. The integration of information from various plasma biomarkers holds the potential to predict clinical outcomes and identify treatment failure prior to radiologic progression, offering valuable guidance for the clinical management of patients with iCCA.
Collapse
Affiliation(s)
| | - Maria Vila-Casadesús
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Marina Gomez-Rey
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Carles Fabregat-Franco
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Upper Gastrointestinal and Endocrine Tumor Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Alexandre Sierra
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Tian V Tian
- Upper Gastrointestinal and Endocrine Tumor Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Florian Castet
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Upper Gastrointestinal and Endocrine Tumor Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Gloria Castillo
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Judit Matito
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paola Martinez
- Molecular Oncology Lab, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Josep M Miquel
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Lab, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Raquel Pérez-López
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Teresa Macarulla
- Gastrointestinal and Endocrine Tumor Unit, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Upper Gastrointestinal and Endocrine Tumor Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
7
|
Wang J, Liu S, Cao Y, Chen Y. Overcoming treatment resistance in cholangiocarcinoma: current strategies, challenges, and prospects. Front Cell Dev Biol 2024; 12:1408852. [PMID: 39156971 PMCID: PMC11327014 DOI: 10.3389/fcell.2024.1408852] [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: 03/28/2024] [Accepted: 06/26/2024] [Indexed: 08/20/2024] Open
Abstract
Significant advancements in our understanding and clinical treatment of cholangiocarcinoma (CCA) have been achieved over the past 5 years. Groundbreaking studies have illuminated the immune landscape and pathological characteristics of the tumor microenvironment in CCA. The development of immune- and metabolism-based classification systems has enabled a nuanced exploration of the tumor microenvironment and the origins of CCA, facilitating a detailed understanding of tumor progression modulation. Despite these insights, targeted therapies have not yet yielded satisfactory clinical results, highlighting the urgent need for innovative therapeutic strategies. This review delineates the complexity and heterogeneity of CCA, examines the current landscape of therapeutic strategies and clinical trials, and delves into the resistance mechanisms underlying targeted therapies. Finally, from a single-cell and spatial transcriptomic perspective, we address the challenge of therapy resistance, discussing emerging mechanisms and potential strategies to overcome this barrier and enhance treatment efficacy.
Collapse
Affiliation(s)
- Jiayi Wang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Siyan Liu
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yi Cao
- Second Clinical College, Chongqing Medical University, Chongqing, China
| | - Yong Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Mahapatra S, Kar P. Computational biophysical characterization of the effect of gatekeeper mutations on the binding of ponatinib to the FGFR kinase. Arch Biochem Biophys 2024; 758:110070. [PMID: 38909834 DOI: 10.1016/j.abb.2024.110070] [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: 02/29/2024] [Revised: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Fibroblast Growth Factor Receptor (FGFR) is connected to numerous downstream signalling cascades regulating cellular behavior. Any dysregulation leads to a plethora of illnesses, including cancer. Therapeutics are available, but drug resistance driven by gatekeeper mutation impedes the treatment. Ponatinib is an FDA-approved drug against BCR-ABL kinase and has shown effective results against FGFR-mediated carcinogenesis. Herein, we undertake molecular dynamics simulation-based analysis on ponatinib against all the FGFR isoforms having Val to Met gatekeeper mutations. The results suggest that ponatinib is a potent and selective inhibitor for FGFR1, FGFR2, and FGFR4 gatekeeper mutations. The extensive electrostatic and van der Waals interaction network accounts for its high potency. The FGFR3_VM mutation has shown resistance towards ponatinib, which is supported by their lesser binding affinity than wild-type complexes. The disengaged molecular brake and engaged hydrophobic spine were believed to be the driving factors for weak protein-ligand interaction. Taken together, the inhibitory and structural characteristics exhibited by ponatinib may aid in thwarting resistance based on Val-to-Met gatekeeper mutations at an earlier stage of treatment and advance the design and development of other inhibitors targeted at FGFRs harboring gatekeeper mutations.
Collapse
Affiliation(s)
- Subhasmita Mahapatra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, 453552, Madhya Pradesh, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore, 453552, Madhya Pradesh, India.
| |
Collapse
|
9
|
Hanssens C, Mouna O, Meyers M, Hendlisz A. State-of-the-art and trends in fibroblast growth factor receptor-directed therapies in gastro-intestinal malignancies. Curr Opin Oncol 2024; 36:320-325. [PMID: 38726837 DOI: 10.1097/cco.0000000000001047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW This review is timely and relevant due to the increasing recognition of the significance of the fibroblast growth factor receptor (FGFR) family in cancer biology. Understanding the role of FGFRs and their dysregulation in various cancers is crucial for developing targeted therapies and improving patient outcomes. RECENT FINDINGS The review highlights the importance of the FGFR family in cellular processes such as growth, proliferation, and survival. It discusses how abnormalities in FGFR2, including overexpression, gene amplification, and other genetic alterations, contribute to cancer progression, particularly in gastro-intestinal cancers. The paper also emphasizes the promising results of FGFR-targeted therapies, especially tyrosine kinase inhibitors, in certain cancers such as cholangiocarcinoma and oesophagogastric cancers. SUMMARY The findings underscore the potential of FGFR-targeted therapies in treating cancers with FGFR dysregulation. However, the review also addresses the challenges associated with these therapies, including toxicities and mechanisms of resistance. Understanding these complexities is essential for optimizing the efficacy of FGFR-targeted treatments and improving patient outcomes in clinical practice and research efforts.
Collapse
Affiliation(s)
- Charlotte Hanssens
- Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | |
Collapse
|
10
|
Demir T, Moloney C, Mahalingam D. Emerging targeted therapies and strategies to overcome resistance in biliary tract cancers. Crit Rev Oncol Hematol 2024; 199:104388. [PMID: 38754771 DOI: 10.1016/j.critrevonc.2024.104388] [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: 01/22/2024] [Revised: 04/14/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
In the last decade, targeted therapies have shown rapid advancement in biliary tract cancer (BTC). Today, many targeted agents are available and under investigation for patients with BTC. More recently, immune checkpoint inhibitors (ICI) such as durvalumab and pembrolizumab in combination with gemcitabine plus cisplatin (gem/cis) have resulted in improved overall survival and progression-free survival in the first-line setting. However, the efficacy benefit of these novel therapeutics is often short-lived, with literature outlining concerns about both primary and secondary resistance to these agents. Investigators also need to consider toxicity profiles that can emerge using this strategy. There have been efforts to reduce evolving resistance through combinatory approaches, both pre-clinically and in early clinical settings. This review summarizes the emerging targeted therapies in BTC, evolving biomarkers of resistance, strategies to overcome them, and an analysis of ongoing clinical trials of patients with advanced BTC.
Collapse
Affiliation(s)
- Tarik Demir
- Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine1, Chicago, IL 60611, USA.
| | - Carolyn Moloney
- Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine1, Chicago, IL 60611, USA
| | - Devalingam Mahalingam
- Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine1, Chicago, IL 60611, USA
| |
Collapse
|
11
|
Rodón J, Damian S, Furqan M, García-Donas J, Imai H, Italiano A, Spanggaard I, Ueno M, Yokota T, Veronese ML, Oliveira N, Li X, Gilmartin A, Schaffer M, Goyal L. Pemigatinib in previously treated solid tumors with activating FGFR1-FGFR3 alterations: phase 2 FIGHT-207 basket trial. Nat Med 2024; 30:1645-1654. [PMID: 38710951 PMCID: PMC11186762 DOI: 10.1038/s41591-024-02934-7] [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: 10/17/2023] [Accepted: 03/19/2024] [Indexed: 05/08/2024]
Abstract
Fibroblast growth factor receptor (FGFR) alterations drive oncogenesis in multiple tumor types. Here we studied pemigatinib, a selective, potent, oral FGFR1-FGFR3 inhibitor, in the phase 2 FIGHT-207 basket study of FGFR-altered advanced solid tumors. Primary end points were objective response rate (ORR) in cohorts A (fusions/rearrangements, n = 49) and B (activating non-kinase domain mutations, n = 32). Secondary end points were progression-free survival, duration of response and overall survival in cohorts A and B, and safety. Exploratory end points included ORR of cohort C (kinase domain mutations, potentially pathogenic variants of unknown significance, n = 26) and analysis of co-alterations associated with resistance and response. ORRs for cohorts A, B and C were 26.5% (13/49), 9.4% (3/32) and 3.8% (1/26), respectively. Tumors with no approved FGFR inhibitors or those with alterations not previously confirmed to be sensitive to FGFR inhibition had objective responses. In cohorts A and B, the median progression-free survival was 4.5 and 3.7 months, median duration of response was 7.8 and 6.9 months and median overall survival was 17.5 and 11.4 months, respectively. Safety was consistent with previous reports. The most common any-grade treatment-emergent adverse events were hyperphosphatemia (84%) and stomatitis (53%). TP53 co-mutations were associated with lack of response and BAP1 alterations with higher response rates. FGFR1-FGFR3 gatekeeper and molecular brake mutations led to acquired resistance. New therapeutic areas for FGFR inhibition and drug failure mechanisms were identified across tumor types. ClinicalTrials.gov identifier: NCT03822117 .
Collapse
MESH Headings
- Humans
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors
- Female
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Pyrimidines/adverse effects
- Pyrimidines/therapeutic use
- Male
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/pathology
- Middle Aged
- Adult
- Aged
- Mutation
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
- Progression-Free Survival
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Morpholines
- Pyrroles
Collapse
Affiliation(s)
- Jordi Rodón
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Silvia Damian
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | - Hiroo Imai
- Tohoku University Hospital, Sendai-Shi, Japan
| | - Antoine Italiano
- Institut Bergonié, Bordeaux, France
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Iben Spanggaard
- Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | | | | - Xin Li
- Incyte Corporation, Wilmington, DE, USA
| | | | | | - Lipika Goyal
- Mass General Cancer Center, Harvard Medical School, Boston, MA, USA.
- Stanford Cancer Center, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
12
|
Balasooriya ER, Wu Q, Ellis H, Zhen Y, Norden BL, Corcoran RB, Mohan A, Martin E, Franovic A, Tyhonas J, Lardy M, Grandinetti KB, Pelham R, Soroceanu L, Silveira VS, Bardeesy N. The Irreversible FGFR Inhibitor KIN-3248 Overcomes FGFR2 Kinase Domain Mutations. Clin Cancer Res 2024; 30:2181-2192. [PMID: 38437671 PMCID: PMC11229173 DOI: 10.1158/1078-0432.ccr-23-3588] [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: 11/17/2023] [Revised: 01/24/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
PURPOSE FGFR2 and FGFR3 show oncogenic activation in many cancer types, often through chromosomal fusion or extracellular domain mutation. FGFR2 and FGFR3 alterations are most prevalent in intrahepatic cholangiocarcinoma (ICC) and bladder cancers, respectively, and multiple selective reversible and covalent pan-FGFR tyrosine kinase inhibitors (TKI) have been approved in these contexts. However, resistance, often due to acquired secondary mutations in the FGFR2/3 kinase domain, limits efficacy. Resistance is typically polyclonal, involving a spectrum of different mutations that most frequently affect the molecular brake and gatekeeper residues (N550 and V565 in FGFR2). EXPERIMENTAL DESIGN Here, we characterize the activity of the next-generation covalent FGFR inhibitor, KIN-3248, in preclinical models of FGFR2 fusion+ ICC harboring a series of secondary kinase domain mutations, in vitro and in vivo. We also test select FGFR3 alleles in bladder cancer models. RESULTS KIN-3248 exhibits potent selectivity for FGFR1-3 and retains activity against various FGFR2 kinase domain mutations, in addition to being effective against FGFR3 V555M and N540K mutations. Notably, KIN-3248 activity extends to the FGFR2 V565F gatekeeper mutation, which causes profound resistance to currently approved FGFR inhibitors. Combination treatment with EGFR or MEK inhibitors potentiates KIN-3248 efficacy in vivo, including in models harboring FGFR2 kinase domain mutations. CONCLUSIONS Thus, KIN-3248 is a novel FGFR1-4 inhibitor whose distinct activity profile against FGFR kinase domain mutations highlights its potential for the treatment of ICC and other FGFR-driven cancers.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cholangiocarcinoma/drug therapy
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/pathology
- Drug Resistance, Neoplasm/genetics
- Mutation
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/pathology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Eranga R. Balasooriya
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Cancer Program, Broad Institute, Cambridge, MA, USA
| | - Qibiao Wu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Cancer Program, Broad Institute, Cambridge, MA, USA
| | - Haley Ellis
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Cancer Program, Broad Institute, Cambridge, MA, USA
| | - Yuanli Zhen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Cancer Program, Broad Institute, Cambridge, MA, USA
| | - Bryanna L. Norden
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Ryan B. Corcoran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | - Vanessa S. Silveira
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Cancer Program, Broad Institute, Cambridge, MA, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Cancer Program, Broad Institute, Cambridge, MA, USA
| |
Collapse
|
13
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [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: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
14
|
Wu Q, Ellis H, Siravegna G, Michel AG, Norden BL, Fece de la Cruz F, Balasooriya ER, Zhen Y, Silveira VS, Che J, Corcoran RB, Bardeesy N. Landscape of Clinical Resistance Mechanisms to FGFR Inhibitors in FGFR2-Altered Cholangiocarcinoma. Clin Cancer Res 2024; 30:198-208. [PMID: 37843855 PMCID: PMC10767308 DOI: 10.1158/1078-0432.ccr-23-1317] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/18/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
PURPOSE FGFR inhibitors are effective in FGFR2-altered cholangiocarcinoma, leading to approval of reversible FGFR inhibitors, pemigatinib and infigratinib, and an irreversible inhibitor, futibatinib. However, acquired resistance develops, limiting clinical benefit. Some mechanisms of resistance have been reported, including secondary FGFR2 kinase domain mutations. Here, we sought to establish the landscape of acquired resistance to FGFR inhibition and to validate findings in model systems. EXPERIMENTAL DESIGN We examined the spectrum of acquired resistance mechanisms detected in circulating tumor DNA or tumor tissue upon disease progression following FGFR inhibitor therapy in 82 FGFR2-altered cholangiocarcinoma patients from 12 published reports. Functional studies of candidate resistance alterations were performed. RESULTS Overall, 49 of 82 patients (60%) had one or more detectable secondary FGFR2 kinase domain mutations upon acquired resistance. N550 molecular brake and V565 gatekeeper mutations were most common, representing 63% and 47% of all FGFR2 kinase domain mutations, respectively. Functional studies showed different inhibitors displayed unique activity profiles against FGFR2 mutations. Interestingly, disruption of the cysteine residue covalently bound by futibatinib (FGFR2 C492) was rare, observed in 1 of 42 patients treated with this drug. FGFR2 C492 mutations were insensitive to inhibition by futibatinib but showed reduced signaling activity, potentially explaining their low frequency. CONCLUSIONS These data support secondary FGFR2 kinase domain mutations as the primary mode of acquired resistance to FGFR inhibitors, most commonly N550 and V565 mutations. Thus, development of combination strategies and next-generation FGFR inhibitors targeting the full spectrum of FGFR2 resistance mutations will be critical.
Collapse
Affiliation(s)
- Qibiao Wu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Haley Ellis
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Giulia Siravegna
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Alexa G. Michel
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Bryanna L. Norden
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Ferran Fece de la Cruz
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Eranga Roshan Balasooriya
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Yuanli Zhen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Vanessa S. Silveira
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| | - Jianwe Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ryan B. Corcoran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
- The Cancer Program, Broad Institute, Cambridge, Massachusetts
| |
Collapse
|
15
|
Lamarca A, Ostios L, McNamara MG, Garzon C, Gleeson JP, Edeline J, Herrero A, Hubner RA, Moreno V, Valle JW. Resistance mechanism to fibroblast growth factor receptor (FGFR) inhibitors in cholangiocarcinoma. Cancer Treat Rev 2023; 121:102627. [PMID: 37925878 DOI: 10.1016/j.ctrv.2023.102627] [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: 07/17/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 11/07/2023]
Abstract
Precision medicine is a major achievement that has impacted on management of patients diagnosed with advanced cholangiocarcinoma (CCA) over the last decade. Molecular profiling of CCA has identified targetable alterations, such as fibroblast growth factor receptor-2 (FGFR-2) fusions, and has thus led to the development of a wide spectrum of compounds. Despite favourable response rates, especially with the latest generation FGFRi, there are still a proportion of patients who will not achieve a radiological response to treatment, or who will have disease progression as the best response. In addition, for patients who do respond to treatment, secondary resistance frequently develops and mechanisms of such resistance are not fully understood. This review will summarise the current state of development of FGFR inhibitors in CCA, their mechanism of action, activity, and the hypothesised mechanisms of resistance.
Collapse
Affiliation(s)
- Angela Lamarca
- Department of Medical Oncology - OncoHealth Institute - Instituto de Investigaciones Sanitarias FJD, Fundación Jiménez Díaz University Hospital, Madrid, Spain; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom.
| | - Lorena Ostios
- START-FJD Phase I Unit, Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Mairéad G McNamara
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Carlos Garzon
- Department of Medical Oncology, Infanta Elena University Hospital, Madrid, Spain
| | - Jack P Gleeson
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom; Cancer Res @UCC, University College Cork, Cork, Ireland
| | - Julien Edeline
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Ana Herrero
- Department of Medical Oncology, Villalba University Hospital, Madrid, Spain
| | - Richard A Hubner
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Victor Moreno
- START-FJD Phase I Unit, Department of Medical Oncology, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Juan W Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
16
|
Subbiah V, Sahai V, Maglic D, Bruderek K, Touré BB, Zhao S, Valverde R, O'Hearn PJ, Moustakas DT, Schönherr H, Gerami-Moayed N, Taylor AM, Hudson BM, Houde DJ, Pal D, Foster L, Gunaydin H, Ayaz P, Sharon DA, Goyal L, Schram AM, Kamath S, Sherwin CA, Schmidt-Kittler O, Jen KY, Ricard F, Wolf BB, Shaw DE, Bergstrom DA, Watters J, Casaletto JB. RLY-4008, the First Highly Selective FGFR2 Inhibitor with Activity across FGFR2 Alterations and Resistance Mutations. Cancer Discov 2023; 13:2012-2031. [PMID: 37270847 PMCID: PMC10481131 DOI: 10.1158/2159-8290.cd-23-0475] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Oncogenic activation of fibroblast growth factor receptor 2 (FGFR2) drives multiple cancers and represents a broad therapeutic opportunity, yet selective targeting of FGFR2 has not been achieved. Although the clinical efficacy of pan-FGFR inhibitors (pan-FGFRi) validates FGFR2 driver status in FGFR2 fusion-positive intrahepatic cholangiocarcinoma, their benefit is limited by incomplete target coverage due to FGFR1- and FGFR4-mediated toxicities (hyperphosphatemia and diarrhea, respectively) and the emergence of FGFR2 resistance mutations. RLY-4008 is a highly selective, irreversible FGFR2 inhibitor designed to overcome these limitations. In vitro, RLY-4008 demonstrates >250- and >5,000-fold selectivity over FGFR1 and FGFR4, respectively, and targets primary alterations and resistance mutations. In vivo, RLY-4008 induces regression in multiple xenograft models-including models with FGFR2 resistance mutations that drive clinical progression on current pan-FGFRi-while sparing FGFR1 and FGFR4. In early clinical testing, RLY-4008 induced responses without clinically significant off-isoform FGFR toxicities, confirming the broad therapeutic potential of selective FGFR2 targeting. SIGNIFICANCE Patients with FGFR2-driven cancers derive limited benefit from pan-FGFRi due to multiple FGFR1-4-mediated toxicities and acquired FGFR2 resistance mutations. RLY-4008 is a highly selective FGFR2 inhibitor that targets primary alterations and resistance mutations and induces tumor regression while sparing other FGFRs, suggesting it may have broad therapeutic potential. See related commentary by Tripathi et al., p. 1964. This article is featured in Selected Articles from This Issue, p. 1949.
Collapse
Affiliation(s)
- Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Dejan Maglic
- Relay Therapeutics, Inc., Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | - Debjani Pal
- Relay Therapeutics, Inc., Cambridge, Massachusetts
| | | | | | | | | | - Lipika Goyal
- Massachusetts General Hospital, Boston, Massachusetts
| | | | - Suneel Kamath
- The Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | | | | | - Kai Yu Jen
- Relay Therapeutics, Inc., Cambridge, Massachusetts
| | | | - Beni B. Wolf
- Relay Therapeutics, Inc., Cambridge, Massachusetts
| | - David E. Shaw
- D. E. Shaw Research, New York, New York
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York
| | | | | | | |
Collapse
|
17
|
Valery M, Vasseur D, Fachinetti F, Boilève A, Smolenschi C, Tarabay A, Antoun L, Perret A, Fuerea A, Pudlarz T, Boige V, Hollebecque A, Ducreux M. Targetable Molecular Alterations in the Treatment of Biliary Tract Cancers: An Overview of the Available Treatments. Cancers (Basel) 2023; 15:4446. [PMID: 37760415 PMCID: PMC10526255 DOI: 10.3390/cancers15184446] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Biliary tract cancers (BTCs) are rare tumours, most often diagnosed at an unresectable stage, associated with poor prognosis, with a 5-year survival rate not exceeding 10%. Only first- and second-line treatments are well codified with the combination of cisplatin-gemcitabine chemotherapy and immunotherapy followed by 5-FU and oxaliplatin chemotherapy, respectively. Many studies have shown that BTC, and more particularly intrahepatic cholangiocarcinoma (iCCA), have a high rate of targetable somatic alteration. To date, the FDA has approved several drugs. Ivosidenib targeting IDH1 mutations, as well as futibatinib and pemigatinib targeting FGFR2 fusions, are approved for pre-treated advanced CCA. The combination of dabrafenib and trametinib are approved for BRAFV600E mutated advanced tumours, NTRK inhibitors entrectinib and larotrectinib for tumours bearing NTRK fusion and prembrolizumab for MSI-H advanced tumours, involving a small percentage of BTC in these three settings. Several other potentially targetable alterations are found in BTC, such as HER2 mutations or amplifications or KRASG12C mutations and mutations in genes involved in DNA repair mechanisms. This review aims to clarify the specific diagnostic modalities for gene alterations and to summarize the results of the main trials and developments underway for the management of advanced BTC with targetable alterations.
Collapse
Affiliation(s)
- Marine Valery
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Damien Vasseur
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France;
| | - Francesco Fachinetti
- Dana-Farber Institute, Lowe Center for Thoracic Oncology, Boston, MA 02215, USA;
| | - Alice Boilève
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, F-94805 Villejuif, France
| | - Cristina Smolenschi
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Anthony Tarabay
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Leony Antoun
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Audrey Perret
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Alina Fuerea
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Thomas Pudlarz
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Valérie Boige
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Antoine Hollebecque
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Michel Ducreux
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, F-94805 Villejuif, France
| |
Collapse
|
18
|
Amadeo E, Rossari F, Vitiello F, Burgio V, Persano M, Cascinu S, Casadei-Gardini A, Rimini M. Past, present, and future of FGFR inhibitors in cholangiocarcinoma: from biological mechanisms to clinical applications. Expert Rev Clin Pharmacol 2023; 16:631-642. [PMID: 37387533 DOI: 10.1080/17512433.2023.2232302] [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: 05/09/2023] [Accepted: 06/29/2023] [Indexed: 07/01/2023]
Abstract
INTRODUCTION Biliary tract carcinoma (BTC) is a heterogenous group of aggressive hepatic malignancies, second to hepatocellular carcinoma per prevalence. Despite clinical research advancement, the overall 5-year survival rate is just above 2%. With the identification of somatic core mutations in half of cholangiocarcinomas. In the intrahepatic subtype (iCCA), it is possible to target mutational pathways of pharmacological interest. AREAS COVERED Major attention has been drawn to fibroblast growth factor receptor (FGFR), especially the type 2 (FGFR2), found mutated in 10-15% of iCCAs. FGFR2 fusions became the target of novel tyrosine-kinase inhibitors investigated in clinical studies, showing promising results so as to gain regulatory approval by American and European committees in recent years. Such drugs demonstrated a better impact on the quality of life compared to standard chemotherapy; however, side effects including hyperphosphatemia, gastrointestinal, eye, and nail disorders are common although mostly manageable. EXPERT OPINION As FGFR inhibitors may soon become the new alternative to standard chemotherapy in FGFR-mutated cholangiocarcinoma, accurate molecular testing and monitoring of acquired resistance mechanisms will be essential. The possible application of FGFR inhibitors in first-line treatment, as well as in combination with current standard treatments, remains the next step to be taken.
Collapse
Affiliation(s)
- Elisabeth Amadeo
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Federico Rossari
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Francesco Vitiello
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Valentina Burgio
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Mara Persano
- Medical Oncology, University and University Hospital of Cagliari, Cagliari, Italy
| | - Stefano Cascinu
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Andrea Casadei-Gardini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Margherita Rimini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| |
Collapse
|
19
|
Ilyas SI, Affo S, Goyal L, Lamarca A, Sapisochin G, Yang JD, Gores GJ. Cholangiocarcinoma - novel biological insights and therapeutic strategies. Nat Rev Clin Oncol 2023; 20:470-486. [PMID: 37188899 PMCID: PMC10601496 DOI: 10.1038/s41571-023-00770-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2023] [Indexed: 05/17/2023]
Abstract
In the past 5 years, important advances have been made in the scientific understanding and clinical management of cholangiocarcinoma (CCA). The cellular immune landscape of CCA has been characterized and tumour subsets with distinct immune microenvironments have been defined using molecular approaches. Among these subsets, the identification of 'immune-desert' tumours that are relatively devoid of immune cells emphasizes the need to consider the tumour immune microenvironment in the development of immunotherapy approaches. Progress has also made in identifying the complex heterogeneity and diverse functions of cancer-associated fibroblasts in this desmoplastic cancer. Assays measuring circulating cell-free DNA and cell-free tumour DNA are emerging as clinical tools for detection and monitoring of the disease. Molecularly targeted therapy for CCA has now become a reality, with three drugs targeting oncogenic fibroblast growth factor receptor 2 (FGFR2) fusions and one targeting neomorphic, gain-of-function variants of isocitrate dehydrogenase 1 (IDH1) obtaining regulatory approval. By contrast, immunotherapy using immune-checkpoint inhibitors has produced disappointing results in patients with CCA, underscoring the requirement for novel immune-based treatment strategies. Finally, liver transplantation for early stage intrahepatic CCA under research protocols is emerging as a viable therapeutic option in selected patients. This Review highlights and provides in-depth information on these advances.
Collapse
Affiliation(s)
- Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Silvia Affo
- Liver, Digestive System and Metabolism Research, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lipika Goyal
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Angela Lamarca
- Department of Oncology, OncoHealth Institute, Fundación Jiménez Díaz University Hospital, Madrid, Spain
- Department of Medical Oncology, The Christie NHS Foundation, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Gonzalo Sapisochin
- Ajmera Transplant Program and HPB Surgical Oncology, Toronto General Hospital, University of Toronto, Toronto, Canada
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Uson PLS, Bearss J, Babiker HM, Borad M. A drug safety evaluation of pemigatinib for advanced cholangiocarcinoma. Expert Opin Drug Saf 2023; 22:637-641. [PMID: 37363820 DOI: 10.1080/14740338.2023.2227561] [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: 02/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION Pemigatinib is a selective small-molecule inhibitor of the fibroblast growth factor receptor (FGFR) 1-3. FGFR is associated with increased cell division, proliferation, and survival. Inhibition of this receptor is an effective treatment against tumors driven by activated fusions in FGFR2. AREAS COVERED The drug was first evaluated in patients with advanced solid tumors and demonstrated a manageable safety profile, with the most common adverse events being oscillations in blood phosphate levels, fatigue, gastrointestinal symptoms, and skin and ocular toxicities. Pemigatinib was further evaluated in a phase II cohort study of patients with previously treated locally advanced or metastatic cholangiocarcinoma harboring FGFR2 genomic alterations. After a median follow-up of 17.8 months, the objective response rate in patients with tumors harboring FGFR2 fusions or rearrangements was 35.5% (95% CI, 26.5-45.4). Based on these results, the FDA granted accelerated approval on 17 April 2020, to pemigatinib, for the treatment of adults with previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with an FGFR2 fusion or another rearrangement. Articles selected for this review were based on reported studies indexed in PubMed (2010-2023). EXPERT OPINION Future perspectives in the treatment of FGFR2 fused cholangiocarcinoma include the evaluation of pemigatinib in previously untreated patients and possible active combinations or sequencing strategies with other drugs.
Collapse
Affiliation(s)
- Pedro Luiz Serrano Uson
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
| | | | - Hani M Babiker
- Department of Oncology, Mayo Clinic Jacksonville, Florida, USA
| | - Mitesh Borad
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
21
|
Karasic TB, Eads JR, Goyal L. Precision Medicine and Immunotherapy Have Arrived for Cholangiocarcinoma: An Overview of Recent Approvals and Ongoing Clinical Trials. JCO Precis Oncol 2023; 7:e2200573. [PMID: 37053534 PMCID: PMC10309532 DOI: 10.1200/po.22.00573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/31/2023] [Indexed: 04/15/2023] Open
Affiliation(s)
- Thomas B. Karasic
- Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, PA
| | - Jennifer R. Eads
- Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, PA
| | - Lipika Goyal
- Department of Medicine, Division of Hematology and Oncology, Stanford Cancer Center, Palo Alto, CA
| |
Collapse
|
22
|
Brown ZJ, Ruff SM, Pawlik TM. Developments in FGFR and IDH inhibitors for cholangiocarcinoma therapy. Expert Rev Anticancer Ther 2023; 23:257-264. [PMID: 36744395 DOI: 10.1080/14737140.2023.2176846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Cholangiocarcinoma (CCA) is an uncommon malignancy originating from epithelial cells of the biliary tract. Regardless of the site of origin within the biliary tree, CCAs are generally aggressive with a poor survival. Surgical resection remains the only chance for cure, yet a majority of patients are not surgical candidates at presentation. Unfortunately, systemic therapies are often ineffective and complicated by side effects. As such, more effective targeted therapies are required in order to improve survival. AREA COVERED Genetic analysis of CCA has allowed for a better understanding of the genomic landscape of CCA. Isocitrate dehydrogenase (IDH) and fibroblast growth factor receptor (FGFR) mutations have emerged as the most promising molecular targets for CCA. Inhibitors of IDH and FGFR have proven to have therapeutic benefit with an acceptable safety profile. However, patients often develop resistance rendering the therapy ineffective. EXPERT OPINION Understanding the molecular pathways of IDH and FGFR may lead to a better understanding of the mechanisms of resistance. Thus, novel therapies may be developed to improve the efficacy of these therapies. Developing novel biomarkers may improve patient selection and further enhance effectiveness of targeted therapies.
Collapse
Affiliation(s)
- Zachary J Brown
- Department of Surgery, Summit Health, Berkeley Heights, NJ, USA
| | - Samantha M Ruff
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
23
|
Lin M, Zhang XL, You R, Liu YP, Cai HM, Liu LZ, Liu XF, Zou X, Xie YL, Zou RH, Zhang YN, Sun R, Feng WY, Wang HY, Tao GH, Li HJ, Huang WJ, Zhang C, Huang PY, Wang J, Zhao Q, Yang Q, Zhang HW, Liu T, Li HF, Jiang XB, Tang J, Gu YK, Yu T, Wang ZQ, Feng L, Kang TB, Zuo ZX, Chen MY. Evolutionary route of nasopharyngeal carcinoma metastasis and its clinical significance. Nat Commun 2023; 14:610. [PMID: 36739462 PMCID: PMC9899247 DOI: 10.1038/s41467-023-35995-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/04/2023] [Indexed: 02/06/2023] Open
Abstract
It is critical to understand factors associated with nasopharyngeal carcinoma (NPC) metastasis. To track the evolutionary route of metastasis, here we perform an integrative genomic analysis of 163 matched blood and primary, regional lymph node metastasis and distant metastasis tumour samples, combined with single-cell RNA-seq on 11 samples from two patients. The mutation burden, gene mutation frequency, mutation signature, and copy number frequency are similar between metastatic tumours and primary and regional lymph node tumours. There are two distinct evolutionary routes of metastasis, including metastases evolved from regional lymph nodes (lymphatic route, 61.5%, 8/13) and from primary tumours (hematogenous route, 38.5%, 5/13). The hematogenous route is characterised by higher IFN-γ response gene expression and a higher fraction of exhausted CD8+ T cells. Based on a radiomics model, we find that the hematogenous group has significantly better progression-free survival and PD-1 immunotherapy response, while the lymphatic group has a better response to locoregional radiotherapy.
Collapse
Affiliation(s)
- Mei Lin
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Zhongshan 2nd Road, Guangzhou, 510080, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Xiao-Long Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Take2 Health (Shenzhen) Limited, Shenzhen, 518066, P. R. China
| | - Rui You
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - You-Ping Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Hong-Min Cai
- School of Computer Science and Engineering, South China University of Technology, 382 East Waihuan Road, Guangzhou, 510006, P. R. China
| | - Li-Zhi Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Imaging Diagnosis and Interventional Center, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Xue-Fei Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Xiong Zou
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Yu-Long Xie
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Ru-Hai Zou
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Department of Ultrasound, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Yi-Nuan Zhang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Wei-Yi Feng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Hai-Yan Wang
- College of Mathematics and Informatics, South China Agricultural University, 483 Wushan Road, Guangzhou, 510642, P. R. China
| | - Gui-Hua Tao
- School of Computer Science and Engineering, South China University of Technology, 382 East Waihuan Road, Guangzhou, 510006, P. R. China
| | - Hao-Jiang Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Imaging Diagnosis and Interventional Center, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Wen-Jie Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Imaging Diagnosis and Interventional Center, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Chao Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Pei-Yu Huang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Jin Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Qi Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Qi Yang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Hong-Wan Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Ting Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Hui-Feng Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Xiao-Bing Jiang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Jun Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, 51 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Yang-Kui Gu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Department of Minimally Invasive Interventional Radiology, Sun Yat-sen University Cancer Center, 51 Dongfeng East Road, Guangzhou, 510060, P. R. China
| | - Tao Yu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Zhi-Qiang Wang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China
| | - Lin Feng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Tie-Bang Kang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China
| | - Zhi-Xiang Zuo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China.
| | - Ming-Yuan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, P. R. China.
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China.
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, China.
| |
Collapse
|
24
|
Testa U, Pelosi E, Castelli G. The clinical value of identifying genetic abnormalities that can be targeted in cholangiocarcinomas. Expert Rev Anticancer Ther 2023; 23:147-162. [PMID: 36654529 DOI: 10.1080/14737140.2023.2170878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Cholangiocarcinomas (CCAs) are a heterogenous group of epithelial malignancies originating at any level of the biliary tree and are subdivided according to their location into intrahepatic (iCCA) and extrahepatic (eCCA). AREAS COVERED This review provides an updated analysis of studies of genetic characterization of CCA at the level of gene mutation profiling, copy number alterations and gene expression, with definition of molecular subgroups and identification of some molecular biomarkers and therapeutic targets. EXPERT OPINION With the development of genetic sequencing, several driver mutations have been identified and targeted as novel therapeutic approaches, including FGFR2, IDH1, BRAF, NTRK, HER2, ROS, and RET. Furthermore, identification of the cellular and molecular structure of the tumor microenvironment has contributed to the development of novel therapies, such as tumor immunotherapy. Combination therapies of chemotherapy plus targeted molecules or immunotherapy are under evaluation and offer the unique opportunity to improve the outcomes of CCA patients with advanced disease.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore Di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore Di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore Di Sanità, Rome, Italy
| |
Collapse
|
25
|
Abstract
Biliary tract cancer (BTC) is the second most common primary liver cancer after hepatocellular carcinoma and accounts for 2% of cancer-related deaths. BTCs are classified according to their anatomical origin into intrahepatic (iCCA), perihilar, or distal cholangiocarcinoma, as well as gall bladder carcinoma. While the mutational profiles in these anatomical BTC subtypes overlap to a large extent, iCCA is notable for the high frequency of IDH1/2 mutations (10-22%) and the nearly exclusive occurrence of FGFR2 fusions in 10-15% of patients. In recent years, FGFR2 fusions have become one of the most promising targets for precision oncology targeting BTC, with FGFR inhibitors already approved in Europe and the United States for patients with advanced, pretreated iCCA. While the therapeutic potential of nonfusion alterations is still under debate, it is expected that the field of FGFR2-directed therapies will be subject to rapid further evolution and optimization. The scope of this review is to provide an overview of oncogenic FGFR signaling in iCCA cells and highlight the pathophysiology, diagnostic testing strategies, and therapeutic promises and challenges associated with FGFR2-altered iCCA.
Collapse
Affiliation(s)
- Arndt Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany; .,Center for Personalized Medicine (ZPM), Hannover Medical School, Hannover, Germany
| | - Oreste Segatto
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Anna Saborowski
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany; .,Center for Personalized Medicine (ZPM), Hannover Medical School, Hannover, Germany
| |
Collapse
|
26
|
Goyal L, Meric-Bernstam F, Hollebecque A, Valle JW, Morizane C, Karasic TB, Abrams TA, Furuse J, Kelley RK, Cassier PA, Klümpen HJ, Chang HM, Chen LT, Tabernero J, Oh DY, Mahipal A, Moehler M, Mitchell EP, Komatsu Y, Masuda K, Ahn D, Epstein RS, Halim AB, Fu Y, Salimi T, Wacheck V, He Y, Liu M, Benhadji KA, Bridgewater JA. Futibatinib for FGFR2-Rearranged Intrahepatic Cholangiocarcinoma. N Engl J Med 2023; 388:228-239. [PMID: 36652354 DOI: 10.1056/nejmoa2206834] [Citation(s) in RCA: 220] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Alterations in fibroblast growth factor receptor 2 (FGFR2) have emerged as promising drug targets for intrahepatic cholangiocarcinoma, a rare cancer with a poor prognosis. Futibatinib, a next-generation, covalently binding FGFR1-4 inhibitor, has been shown to have both antitumor activity in patients with FGFR-altered tumors and strong preclinical activity against acquired resistance mutations associated with ATP-competitive FGFR inhibitors. METHODS In this multinational, open-label, single-group, phase 2 study, we enrolled patients with unresectable or metastatic FGFR2 fusion-positive or FGFR2 rearrangement-positive intrahepatic cholangiocarcinoma and disease progression after one or more previous lines of systemic therapy (excluding FGFR inhibitors). The patients received oral futibatinib at a dose of 20 mg once daily in a continuous regimen. The primary end point was objective response (partial or complete response), as assessed by independent central review. Secondary end points included the response duration, progression-free and overall survival, safety, and patient-reported outcomes. RESULTS Between April 16, 2018, and November 29, 2019, a total of 103 patients were enrolled and received futibatinib. A total of 43 of 103 patients (42%; 95% confidence interval, 32 to 52) had a response, and the median duration of response was 9.7 months. Responses were consistent across patient subgroups, including patients with heavily pretreated disease, older adults, and patients who had co-occurring TP53 mutations. At a median follow-up of 17.1 months, the median progression-free survival was 9.0 months and overall survival was 21.7 months. Common treatment-related grade 3 adverse events were hyperphosphatemia (in 30% of the patients), an increased aspartate aminotransferase level (in 7%), stomatitis (in 6%), and fatigue (in 6%). Treatment-related adverse events led to permanent discontinuation of futibatinib in 2% of the patients. No treatment-related deaths occurred. Quality of life was maintained throughout treatment. CONCLUSIONS In previously treated patients with FGFR2 fusion or rearrangement-positive intrahepatic cholangiocarcinoma, the use of futibatinib, a covalent FGFR inhibitor, led to measurable clinical benefit. (Funded by Taiho Oncology and Taiho Pharmaceutical; FOENIX-CCA2 ClinicalTrials.gov number, NCT02052778.).
Collapse
Affiliation(s)
- Lipika Goyal
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Funda Meric-Bernstam
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Antoine Hollebecque
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Juan W Valle
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Chigusa Morizane
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Thomas B Karasic
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Thomas A Abrams
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Junji Furuse
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Robin K Kelley
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Philippe A Cassier
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Heinz-Josef Klümpen
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Heung-Moon Chang
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Li-Tzong Chen
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Josep Tabernero
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Do-Youn Oh
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Amit Mahipal
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Markus Moehler
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Edith P Mitchell
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Yoshito Komatsu
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Kunihiro Masuda
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Daniel Ahn
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Robert S Epstein
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Abdel-Baset Halim
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Yao Fu
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Tehseen Salimi
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Volker Wacheck
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Yaohua He
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Mei Liu
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - Karim A Benhadji
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| | - John A Bridgewater
- From the Department of Medicine, Stanford University School of Medicine, and the Stanford Cancer Center, Palo Alto (L.G.), and the University of California, San Francisco, San Francisco (R.K.K.) - both in California; the Mass General Cancer Center, Harvard Medical School (L.G.), and Dana-Farber Cancer Institute (T.A.A.) - both in Boston; the University of Texas M.D. Anderson Cancer Center, Houston (F.M.-B.); the Drug Development Department, Gustave Roussy, Villejuif (A.H.), and Centre Léon Bérard, Lyon (P.A.C.) - both in France; the University of Manchester and the Christie NHS Foundation Trust, Manchester (J.W.V.), and University College London Cancer Institute, London (J.A.B.) - both in the United Kingdom; National Cancer Center Hospital, Tokyo (C.M.), Kanagawa Cancer Center, Yokohama (J.F.), Hokkaido University Hospital Cancer Center, Sapporo (Y.K.), and Tohoku University Graduate School of Medicine, Sendai (K.M.) - all in Japan; the Hospital of the University of Pennsylvania (T.B.K.) and Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital (E.P.M.) - both in Philadelphia; Amsterdam University Medical Center, University of Amsterdam, Amsterdam (H.-J.K.); Asan Medical Center, University of Ulsan College of Medicine (H.-M.C.), and Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine (D.-Y.O.) - both in Seoul, South Korea; the National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan (L.-T.C.); Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, University of Vic-Central University of Catalonia, Baselga Oncologic Institute, Hospital Quiron, Barcelona (J.T.); Mayo Clinic, Rochester, MN (A.M.); Johannes Gutenberg-Mainz University Medical Center, Mainz, Germany (M.M.); Mayo Clinic, Phoenix, AZ (D.A.); Epstein Health, Woodcliff Lake, NJ (R.S.E.); Taiho Oncology, Princeton, NJ (A.-B.H., T.S., V.W., Y.H., M.L., K.A.B.); and Ilumina, San Diego, CA (Y.F.)
| |
Collapse
|
27
|
Wu Q, Qian W, Sun X, Jiang S. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol 2022; 15:143. [PMID: 36209184 PMCID: PMC9548212 DOI: 10.1186/s13045-022-01362-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/02/2022] [Indexed: 11/10/2022] Open
Abstract
The United States Food and Drug Administration (US FDA) has always been a forerunner in drug evaluation and supervision. Over the past 31 years, 1050 drugs (excluding vaccines, cell-based therapies, and gene therapy products) have been approved as new molecular entities (NMEs) or biologics license applications (BLAs). A total of 228 of these 1050 drugs were identified as cancer therapeutics or cancer-related drugs, and 120 of them were classified as therapeutic drugs for solid tumors according to their initial indications. These drugs have evolved from small molecules with broad-spectrum antitumor properties in the early stage to monoclonal antibodies (mAbs) and antibody‒drug conjugates (ADCs) with a more precise targeting effect during the most recent decade. These drugs have extended indications for other malignancies, constituting a cancer treatment system for monotherapy or combined therapy. However, the available targets are still mainly limited to receptor tyrosine kinases (RTKs), restricting the development of antitumor drugs. In this review, these 120 drugs are summarized and classified according to the initial indications, characteristics, or functions. Additionally, RTK-targeted therapies and immune checkpoint-based immunotherapies are also discussed. Our analysis of existing challenges and potential opportunities in drug development may advance solid tumor treatment in the future.
Collapse
Affiliation(s)
- Qing Wu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Wei Qian
- Department of Radiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009 Zhejiang China
| | - Xiaoli Sun
- Department of Radiation Oncology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310003 Zhejiang China
| | - Shaojie Jiang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| |
Collapse
|
28
|
Berchuck JE, Facchinetti F, DiToro DF, Baiev I, Majeed U, Reyes S, Chen C, Zhang K, Sharman R, Junior PLSU, Maurer J, Shroff RT, Pritchard CC, Wu MJ, Catenacci DVT, Javle M, Friboulet L, Hollebecque A, Bardeesy N, Zhu AX, Lennerz JK, Tan B, Borad M, Parikh AR, Kiedrowski LA, Kelley RK, Mody K, Juric D, Goyal L. The Clinical Landscape of Cell-Free DNA Alterations in 1,671 Patients with Advanced Biliary Tract Cancer. Ann Oncol 2022; 33:1269-1283. [PMID: 36089135 DOI: 10.1016/j.annonc.2022.09.150] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/18/2022] [Accepted: 09/01/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Targeted therapies have transformed clinical management of advanced biliary tract cancer (BTC). Cell-free DNA (cfDNA) analysis is an attractive approach for cancer genomic profiling that overcomes many limitations of traditional tissue-based analysis. We examined cfDNA as a tool to inform clinical management of patients with advanced BTC and generate novel insights into BTC tumor biology. PATIENTS AND METHODS We analyzed next-generation sequencing data of 2,068 cfDNA samples from 1,671 patients with advanced BTC generated with Guardant360. We performed clinical annotation on a multi-institutional subset (n=225) to assess intra-patient cfDNA-tumor concordance and the association of cfDNA variant allele fraction (VAF) with clinical outcomes. RESULTS Genetic alterations were detected in cfDNA in 84% of patients, with targetable alterations detected in 44% of patients. FGFR2 fusions, IDH1 mutations, and BRAF V600E were clonal in majority of cases, affirming these targetable alterations as early driver events in BTC. Concordance between cfDNA and tissue for mutation detection was high for IDH1 mutations (87%) and BRAF V600E (100%), and low for FGFR2 fusions (18%). cfDNA analysis uncovered novel putative mechanisms of resistance to targeted therapies, including mutation of the cysteine residue (FGFR2 C492F) to which covalent FGFR inhibitors bind. High pre-treatment cfDNA VAF associated with poor prognosis and shorter response to chemotherapy and targeted therapy. Finally, we report the frequency of promising targets in advanced BTC currently under investigation in other advanced solid tumors, including KRAS G12C (1.0%), KRAS G12D (5.1%), PIK3CA mutations (6.8%), and ERBB2 amplifications (4.9%). CONCLUSIONS These findings from the largest and most comprehensive study to date of cfDNA from patients with advanced BTC highlight the utility of cfDNA analysis in current management of this disease. Characterization of oncogenic drivers and mechanisms of therapeutic resistance in this study will inform drug development efforts to reduce mortality for patients with BTC.
Collapse
Affiliation(s)
- Jacob E Berchuck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Francesco Facchinetti
- Université Paris-Saclay, Institut Gustave Roussy, Inserm U981, Biomarqueurs Prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, Villejuif, France
| | - Daniel F DiToro
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Islam Baiev
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA
| | - Umair Majeed
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| | | | - Christopher Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Karen Zhang
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - Reya Sharman
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | | | - Jordan Maurer
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA
| | - Rachna T Shroff
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Meng-Ju Wu
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA
| | | | - Milind Javle
- Division of Cancer Medicine, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luc Friboulet
- Université Paris-Saclay, Institut Gustave Roussy, Inserm U981, Biomarqueurs Prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, Villejuif, France
| | - Antoine Hollebecque
- Université Paris-Saclay, Institut Gustave Roussy, Inserm U981, Biomarqueurs Prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, Villejuif, France
| | - Nabeel Bardeesy
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA
| | - Andrew X Zhu
- Jiahui International Cancer Center, Jihaui Health, Shanghai, China; I-Mab Biopharma, Shanghai, China
| | - Jochen K Lennerz
- Center for Integrated Diagnostics, Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Benjamin Tan
- Department of Medicine, Washington University, St. Louis, MO
| | - Mitesh Borad
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ
| | - Aparna R Parikh
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA
| | | | - Robin Kate Kelley
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - Kabir Mody
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| | - Dejan Juric
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA
| | - Lipika Goyal
- Department of Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
29
|
FGFR2-IIIb Expression by Immunohistochemistry Has High Specificity in Cholangiocarcinoma with FGFR2 Genomic Alterations. Dig Dis Sci 2022; 67:3797-3805. [PMID: 34773565 DOI: 10.1007/s10620-021-07303-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/21/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND FGFR2 genomic alterations are observed in 10-20% of cholangiocarcinoma (CCA). Although FGFR2 fusions are an important actionable target, FGFR2 protein expression has not been thoroughly characterized. AIMS To evaluate FGFR2 protein expression in cholangiocarcinoma harboring FGFR2 genomic alterations. METHODS FGFR2 protein expression was evaluated in 99 CCA cases with two different antibodies. FGFR2 genomic alterations were confirmed via next-generating sequencing (NGS) or FISH. Primary objective was to determine the specificity and sensitivity of FGFR2 immunohistochemistry staining for detecting FGFR2 genomic alterations. Secondary objectives included overall FGFR2 immunohistochemistry staining in CCA patients, and evaluation of whether FGFR2 expression correlates with clinical outcomes including overall survival (OS), progression-free survival (PFS), and time-to-tumor recurrence (TTR). RESULTS Immunohistochemistry staining with two antibodies against FGFR2, FPR2-D, and clone 98706 showed high accuracy (78.7% and 91.9%) and specificity (82.9% and 97.7%), and moderate sensitivity (53.9% and 57.1%), respectively, when compared with the standard methods for detecting FGFR2 genomic alterations. In a median follow-up of 72 months, there were no statistically significant differences in OS, PFS, and TTR, for patients with positive or negative FGFR2 staining. CONCLUSION FGFR2 protein expression by immunohistochemistry has high specificity and therefore could be used to imply the presence of FGFR2 genomic alterations in the context of a positive test. In the case of a negative test, NGS or FISH would be necessary to ascertain cases with FGFR2 genomic alterations.
Collapse
|
30
|
Wu Q, Zhen Y, Shi L, Vu P, Greninger P, Adil R, Merritt J, Egan R, Wu MJ, Yin X, Ferrone CR, Deshpande V, Baiev I, Pinto CJ, McLoughlin DE, Walmsley CS, Stone JR, Gordan JD, Zhu AX, Juric D, Goyal L, Benes CH, Bardeesy N. EGFR Inhibition Potentiates FGFR Inhibitor Therapy and Overcomes Resistance in FGFR2 Fusion-Positive Cholangiocarcinoma. Cancer Discov 2022; 12:1378-1395. [PMID: 35420673 PMCID: PMC9064956 DOI: 10.1158/2159-8290.cd-21-1168] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/10/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022]
Abstract
FGFR inhibitors are approved for the treatment of advanced cholangiocarcinoma harboring FGFR2 fusions. However, the response rate is moderate, and resistance emerges rapidly due to acquired secondary FGFR2 mutations or due to other less-defined mechanisms. Here, we conducted high-throughput combination drug screens, biochemical analysis, and therapeutic studies using patient-derived models of FGFR2 fusion-positive cholangiocarcinoma to gain insight into these clinical profiles and uncover improved treatment strategies. We found that feedback activation of EGFR signaling limits FGFR inhibitor efficacy, restricting cell death induction in sensitive models and causing resistance in insensitive models lacking secondary FGFR2 mutations. Inhibition of wild-type EGFR potentiated responses to FGFR inhibitors in both contexts, durably suppressing MEK/ERK and mTOR signaling, increasing apoptosis, and causing marked tumor regressions in vivo. Our findings reveal EGFR-dependent adaptive signaling as an important mechanism limiting FGFR inhibitor efficacy and driving resistance and support clinical testing of FGFR/EGFR inhibitor therapy for FGFR2 fusion-positive cholangiocarcinoma. SIGNIFICANCE We demonstrate that feedback activation of EGFR signaling limits the effectiveness of FGFR inhibitor therapy and drives adaptive resistance in patient-derived models of FGFR2 fusion-positive cholangiocarcinoma. These studies support the potential of combination treatment with FGFR and EGFR inhibitors as an improved treatment for patients with FGFR2-driven cholangiocarcinoma.
Collapse
Affiliation(s)
- Qibiao Wu
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yuanli Zhen
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lei Shi
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Phuong Vu
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Patricia Greninger
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ramzi Adil
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joshua Merritt
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Regina Egan
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Meng-Ju Wu
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xunqin Yin
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Cristina R Ferrone
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vikram Deshpande
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Islam Baiev
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christopher J Pinto
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel E McLoughlin
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Charlotte S Walmsley
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - John D Gordan
- Helen Diller Family Comprehensive Cancer Center and Quantitative Biosciences Institute, University of California, San Francisco
| | - Andrew X Zhu
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Jiahui International Cancer Center, Jiahui Health, Shanghai, China
| | - Dejan Juric
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lipika Goyal
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Cyril H Benes
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nabeel Bardeesy
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
31
|
Rengan AK, Denlinger CS. Robust Response to Futibatinib in a Patient With Metastatic FGFR-Addicted Cholangiocarcinoma Previously Treated Using Pemigatinib. J Natl Compr Canc Netw 2022; 20:430-435. [PMID: 35378504 DOI: 10.6004/jnccn.2021.7121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/14/2021] [Indexed: 01/21/2023]
Abstract
Futibatinib is a novel FGFR inhibitor currently under investigation as a second-line treatment for locally advanced or metastatic cholangiocarcinoma harboring FGFR2 gene fusions and rearrangements. As FGFR-targeted therapies move into the frontline setting, sequencing of these drugs remains undetermined. To date, no study has investigated the use of futibatinib in the context of pemigatinib resistance. We describe a 50-year-old woman with metastatic FGFR-aberrant intrahepatic cholangiocarcinoma who showed a robust response to futibatinib for 23.6 months, having previously benefited from pemigatinib. Futibatinib was safely used despite her history of decompensated cirrhosis and significant cytopenias. We observed a reduction in CA 19-9 level and a partial radiographic response on futibatinib. Serial next-generation sequencing and cell-free DNA testing proved crucial to making appropriate treatment decisions.
Collapse
Affiliation(s)
- Anil K Rengan
- 1Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, and
| | - Crystal S Denlinger
- 1Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, and.,2National Comprehensive Cancer Network, Plymouth Meeting, Pennsylvania
| |
Collapse
|
32
|
Zugman M, Botrus G, Pestana RC, Uson Junior PLS. Precision Medicine Targeting FGFR2 Genomic Alterations in Advanced Cholangiocarcinoma: Current State and Future Perspectives. Front Oncol 2022; 12:860453. [PMID: 35444941 PMCID: PMC9013963 DOI: 10.3389/fonc.2022.860453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/14/2022] [Indexed: 01/06/2023] Open
Abstract
Although a relatively uncommon tumor, cholangiocarcinoma is on the rise globally. Of note, most patients are diagnosed with metastatic disease, and the prognosis is poor with cytotoxic chemotherapy. Strategies targeting specific genomic alterations have demonstrated promising activity in recent years and could represent a new therapeutic avenue for these patients. In this review, we will address the biology and clinical results of FGFR inhibition in intrahepatic cholangiocarcinoma, highlighting limitations associated with treatment and discussing the use of circulating tumor DNA to detect mechanisms of resistance.
Collapse
Affiliation(s)
- Miguel Zugman
- Department of Oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Gehan Botrus
- Medical Oncology Phase I Clinical Trials, HonorHealth Research Institute, Phoenix, AZ, United States
| | | | - Pedro Luiz Serrano Uson Junior
- Department of Oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
33
|
Silverman IM, Li M, Murugesan K, Krook MA, Javle MM, Kelley RK, Borad MJ, Roychowdhury S, Meng W, Yilmazel B, Milbury C, Shewale S, Feliz L, Burn TC, Albacker LA. Validation and Characterization of FGFR2 Rearrangements in Cholangiocarcinoma with Comprehensive Genomic Profiling. J Mol Diagn 2022; 24:351-364. [PMID: 35176488 DOI: 10.1016/j.jmoldx.2021.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/26/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a heterogeneous biliary tract cancer with a poor prognosis. Approximately 30% to 50% of patients harbor actionable alterations, including FGFR2 rearrangements. Pemigatinib, a potent, selective fibroblast growth factor receptor (FGFR) FGFR1-3 inhibitor, is approved for previously treated, unresectable, locally advanced or metastatic CCA harboring FGFR2 fusions/rearrangements, as detected by a US Food and Drug Administration-approved test. The next-generation sequencing (NGS)-based FoundationOneCDx (F1CDx) was US Food and Drug Administration approved for detecting FGFR2 fusions or rearrangements. The precision and reproducibility of F1CDx in detecting FGFR2 rearrangements in CCA were examined. Analytical concordance between F1CDx and an externally validated RNA-based NGS (evNGS) test was performed. Identification of FGFR2 rearrangements in the screening population from the pivotal FIGHT-202 study (NCT02924376) was compared with F1CDx. The reproducibility and repeatability of F1CDx were 90% to 100%. Adjusted positive, negative, and overall percentage agreements were 87.1%, 99.6%, and 98.3%, respectively, between F1CDx and evNGS. Compared with evNGS, F1CDx had a positive predictive value of 96.2% and a negative predictive value of 98.5%. The positive percentage agreement, negative percentage agreement, overall percentage agreement, positive predictive value, and negative predictive value were 100% for F1CDx versus the FIbroblast Growth factor receptor inhibitor in oncology and Hematology Trial-202 (FIGHT-202) clinical trial assay. Of 6802 CCA samples interrogated, 9.2% had FGFR2 rearrangements. Cell lines expressing diverse FGFR2 fusions were sensitive to pemigatinib. F1CDx demonstrated sensitivity, reproducibility, and high concordance with clinical utility in identifying patients with FGFR2 rearrangements who may benefit from pemigatinib treatment.
Collapse
Affiliation(s)
- Ian M Silverman
- Translational Sciences, Incyte Research Institute, Wilmington, Delaware
| | - Meijuan Li
- Research and Development, Foundation Medicine, Cambridge, Massachusetts
| | | | - Melanie A Krook
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Milind M Javle
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robin K Kelley
- University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | | | | | - Wei Meng
- Research and Development, Foundation Medicine, Cambridge, Massachusetts
| | - Bahar Yilmazel
- Research and Development, Foundation Medicine, Cambridge, Massachusetts
| | - Coren Milbury
- Research and Development, Foundation Medicine, Cambridge, Massachusetts
| | - Shantanu Shewale
- Research and Development, Foundation Medicine, Cambridge, Massachusetts
| | - Luis Feliz
- Clinical Development, Incyte Biosciences International Sàrl, Morges, Switzerland
| | - Timothy C Burn
- Translational Sciences, Incyte Research Institute, Wilmington, Delaware.
| | - Lee A Albacker
- Research and Development, Foundation Medicine, Cambridge, Massachusetts.
| |
Collapse
|
34
|
An J, Kim D, Oh B, Oh YJ, Song J, Park N, Kim HI, Kang HJ, Oh JH, Kim W, Lee E, Sung CO, Song GW, Kim DG, Yu E, Letouzé E, Zucman-Rossi J, Lee HC, Shim JH. Comprehensive characterization of viral integrations and genomic aberrations in HBV-infected intrahepatic cholangiocarcinomas. Hepatology 2022; 75:997-1011. [PMID: 34478159 DOI: 10.1002/hep.32135] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/08/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Despite the epidemiological association between intrahepatic cholangiocarcinoma (iCCA) and HBV infection, little is known about the relevant oncogenic effects. We sought to identify the landscape and mechanism of HBV integration, along with the genomic architecture of HBV-infected iCCA (HBV-iCCA) tumors. APPROACH AND RESULTS We profiled a cohort of 108 HBV-iCCAs using whole-genome sequencing, deep sequencing, and RNA sequencing, together with preconstructed data sets of HBV-infected HCC (HBV-HCC; n = 167) and combined hepatocellular cholangiocarcinoma (HBV-cHCC/CCA; n = 59), and conventional (n = 154) and fluke-related iCCAs (n = 16). Platforms based on primary iCCA cell lines to evaluate the functional effects of chimeric transcripts were also used. We found that HBV had inserted at multiple sites in the iCCA genomes in 45 (41.7%) of the tumors. Recurrent viral integration breakpoints were found at nine different sites. The most common insertional hotspot (7 tumors) was in the TERT (telomerase reverse transcriptase) promoter, where insertions and mutations (11 tumors) were mutually exclusive, and were accompanied by promoter hyperactivity. Recurrent HBV integration events (5 tumors) were also detected in FAT2 (FAT atypical cadherin 2), and were associated with enrichment of epithelial-mesenchymal transition-related genes. A distinctive intergenic insertion (chr9p21.3), between DMRTA1 (DMRT like family A1) and LINC01239 (long intergenic non-protein coding RNA 1239), had oncogenic effects through activation of the mammalian target of rapamycin (mTOR)/4EBP/S6K pathway. Regarding the mutational profiles of primary liver cancers, the overall landscape of HBV-iCCA was closer to that of nonviral conventional iCCA, than to HBV-HCC and HBV-cHCC/CCA. CONCLUSIONS Our findings provide insight into the behavior of iCCAs driven by various pathogenic mechanisms involving HBV integration events and associated genomic aberrations. This knowledge should be of use in managing HBV carriers.
Collapse
Affiliation(s)
- Jihyun An
- Gastroenterology and HepatologyHanyang University College of MedicineGuri, GyeonggiRepublic of Korea
| | - Deokhoon Kim
- PathologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea.,Center for Cancer Genome DiscoveryAsan Institute for Life ScienceUniversity of Ulsan College of MedicineAsan Medical CenterSeoulRepublic of Korea
| | - Bora Oh
- Asan Institute for Life ScienceAsan Medical CenterSeoulRepublic of Korea
| | - Yoo-Jin Oh
- Asan Institute for Life ScienceAsan Medical CenterSeoulRepublic of Korea
| | - Jihyun Song
- Asan Institute for Life ScienceAsan Medical CenterSeoulRepublic of Korea
| | - Naomi Park
- Asan Institute for Life ScienceAsan Medical CenterSeoulRepublic of Korea
| | - Ha Il Kim
- GastroenterologyKyung Hee University Hospital at GangdongSeoulRepublic of Korea
| | - Hyo Jeong Kang
- PathologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Ji-Hye Oh
- Center for Cancer Genome DiscoveryAsan Institute for Life ScienceUniversity of Ulsan College of MedicineAsan Medical CenterSeoulRepublic of Korea.,Asan Institute for Life ScienceAsan Medical CenterSeoulRepublic of Korea
| | - Wonkyung Kim
- Center for Cancer Genome DiscoveryAsan Institute for Life ScienceUniversity of Ulsan College of MedicineAsan Medical CenterSeoulRepublic of Korea.,Asan Institute for Life ScienceAsan Medical CenterSeoulRepublic of Korea
| | - Eunjung Lee
- Medical ScienceAsan Medical Institute of Convergence Science and TechnologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Chang Ohk Sung
- PathologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea.,Center for Cancer Genome DiscoveryAsan Institute for Life ScienceUniversity of Ulsan College of MedicineAsan Medical CenterSeoulRepublic of Korea
| | - Gi-Won Song
- SurgeryAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea.,Asan Liver CenterAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Dae-Ghon Kim
- Gastroenterology and HepatologyChonbuk National University Medical SchoolJeonjuJeonbukRepublic of Korea
| | - Eunsil Yu
- PathologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea.,Asan Liver CenterAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Eric Letouzé
- Centre de Recherche des CordeliersSorbonne UniversitéINSERMUniversité de ParisParisFrance.,Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale Contre le CancerLabex OncoImmunologyParisFrance
| | - Jessica Zucman-Rossi
- Centre de Recherche des CordeliersSorbonne UniversitéINSERMUniversité de ParisParisFrance.,Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale Contre le CancerLabex OncoImmunologyParisFrance.,Hôpital Européen Georges PompidouParisFrance
| | - Han Chu Lee
- Asan Liver CenterAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea.,GastroenterologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Ju Hyun Shim
- Asan Liver CenterAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea.,GastroenterologyAsan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| |
Collapse
|
35
|
Deciphering Tumour Heterogeneity: From Tissue to Liquid Biopsy. Cancers (Basel) 2022; 14:cancers14061384. [PMID: 35326534 PMCID: PMC8946040 DOI: 10.3390/cancers14061384] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Most malignant tumours are highly heterogeneous at molecular and phenotypic levels. Tumour variability poses challenges for the management of patients, as it arises between patients and even evolves in space and time within a single patient. Currently, treatment-decision making usually relies on the molecular characteristics of a limited tumour tissue sample at the time of diagnosis or disease progression but does not take into account the complexity of the bulk tumours and their constant evolution over time. In this review, we explore the extent of tumour heterogeneity and report the mechanisms that promote and sustain this diversity in cancers. We summarise the clinical strikes of tumour diversity in the management of patients with cancer. Finally, we discuss the current material and technological approaches that are relevant to adequately appreciate tumour heterogeneity. Abstract Human solid malignancies harbour a heterogeneous set of cells with distinct genotypes and phenotypes. This heterogeneity is installed at multiple levels. A biological diversity is commonly observed between tumours from different patients (inter-tumour heterogeneity) and cannot be fully captured by the current consensus molecular classifications for specific cancers. To extend the complexity in cancer, there are substantial differences from cell to cell within an individual tumour (intra-tumour heterogeneity, ITH) and the features of cancer cells evolve in space and time. Currently, treatment-decision making usually relies on the molecular characteristics of a limited tumour tissue sample at the time of diagnosis or disease progression but does not take into account the complexity of the bulk tumours and their constant evolution over time. In this review, we explore the extent of tumour heterogeneity with an emphasis on ITH and report the mechanisms that promote and sustain this diversity in cancers. We summarise the clinical strikes of ITH in the management of patients with cancer. Finally, we discuss the current material and technological approaches that are relevant to adequately appreciate ITH.
Collapse
|
36
|
Chakrabarti S, Finnes HD, Mahipal A. Fibroblast growth factor receptor (FGFR) inhibitors in cholangiocarcinoma: current status, insight on resistance mechanisms and toxicity management. Expert Opin Drug Metab Toxicol 2022; 18:85-98. [DOI: 10.1080/17425255.2022.2039118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sakti Chakrabarti
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226
| | - Heidi D. Finnes
- Pharmacy Cancer Research, Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Amit Mahipal
- Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| |
Collapse
|
37
|
Ghosh A, Myacheva K, Riester M, Schmidt C, Diederichs S. Chimeric oligonucleotides combining guide RNA and single-stranded DNA repair template effectively induce precision gene editing. RNA Biol 2021; 19:588-593. [PMID: 35465826 PMCID: PMC9128553 DOI: 10.1080/15476286.2022.2067713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/11/2022] [Indexed: 10/30/2022] Open
Abstract
The ability to precisely alter the genome holds immense potential for molecular biology, medicine and biotechnology. The development of the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) into a genomic editing tool has vastly simplified genome engineering. Here, we explored the use of chemically synthesized chimeric oligonucleotides encoding a target-specific crRNA (CRISPR RNA) fused to a single-stranded DNA repair template for RNP-mediated precision genome editing. By generating three clinically relevant oncogenic driver mutations, two non-stop extension mutations, an FGFRi resistance mutation and a single nucleotide change, we demonstrate the ability of chimeric oligos to form RNPs and direct Cas9 to effectively induce genome editing. Further, we demonstrate that the polarity of the chimeric oligos is crucial: only chimeric oligos with the single-stranded DNA repair template fused to the 3'-end of the crRNA are functional for accurate editing, while templates fused to the 5'-end are ineffective. We also find that chimeras can perform editing with both symmetric and asymmetric single-stranded DNA repair templates. Depending on the target locus, the editing efficiency using chimeric RNPs is similar to or less than the efficiency of editing using the bipartite standard RNPs. Our results indicate that chimeric RNPs comprising RNA-DNA oligos formed from fusing the crRNA and DNA repair templates can successfully induce precise edits. While chimeric RNPs do not display an advantage over standard RNPs, they nonetheless represent a viable approach for one-molecule precision genome editing.
Collapse
Affiliation(s)
- Avantika Ghosh
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Freiburg, Germany
| | - Ksenia Myacheva
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Freiburg, Germany
- Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marisa Riester
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Freiburg, Germany
| | - Carla Schmidt
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Freiburg, Germany
| | - Sven Diederichs
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Freiburg, Germany
- Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
38
|
Uson Junior PLS, Borad MJ. Precision approaches for cholangiocarcinoma: Progress in clinical trials and beyond. Expert Opin Investig Drugs 2021; 31:125-131. [PMID: 34904492 DOI: 10.1080/13543784.2022.2017882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Although a relatively uncommon tumor, the incidence of intrahepatic cholangiocarcinoma is rising globally. Unfortunately, most patients are diagnosed with locally advanced or metastatic disease with poor prognosis. Strategies targeting genomic alterations and incorporation of precision medicine will represent a new therapeutic avenue for these patients. AREAS COVERED In this review article we addressed clinical trials in cholangiocarcinoma with FGFR, IDH, BRAF and ErbB2 targeted therapies. We also reviewed mechanisms of resistance to precision medicine and possible future strategies to overcome clonal evolution. Articles selected for this review were based on reported studies indexed in PubMed (2010-2021). EXPERT OPINION Pemigatinib, infigratinib and futibatinib could eventually be incorporated in the landscape of first-line systemic treatment for advanced cholangiocarcinoma with FGFR2 fusions or rearrangements after the ongoing phase III trials. Circulating tumor DNA could be used as a dynamic tool for evaluating mechanisms of resistance and prediction of response in patients treated with directed therapy.
Collapse
Affiliation(s)
- Pedro Luiz Serrano Uson Junior
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA.,Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Mitesh J Borad
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Medicine, Rochester, MN, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
39
|
Combination therapies for targeting FGFR2 fusions in cholangiocarcinoma. Trends Cancer 2021; 8:83-86. [PMID: 34840108 DOI: 10.1016/j.trecan.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
Fibroblast growth factor receptor 2 (FGFR2) fusion proteins (FFs) are oncogenic drivers in 10-15% of intrahepatic cholangiocarcinoma (iCCA). FGFR-specific inhibitors provide temporary benefit in FF+ unresectable patients. Recent work with mouse iCCA models has documented the necessary role of RAS-ERK downstream to FFs and provided examples of preclinical experimentation aimed at improving FF targeting in iCCA.
Collapse
|
40
|
FGF/FGFR-Dependent Molecular Mechanisms Underlying Anti-Cancer Drug Resistance. Cancers (Basel) 2021; 13:cancers13225796. [PMID: 34830951 PMCID: PMC8616288 DOI: 10.3390/cancers13225796] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Deregulation of the FGF/FGFR axis is associated with many types of cancer and contributes to the development of chemoresistance, limiting the effectiveness of current treatment strategies. There are several mechanisms involved in this phenomenon, including cross-talks with other signaling pathways, avoidance of apoptosis, stimulation of angiogenesis, and initiation of EMT. Here, we provide an overview of current research and approaches focusing on targeting components of the FGFR/FGF signaling module to overcome drug resistance during anti-cancer therapy. Abstract Increased expression of both FGF proteins and their receptors observed in many cancers is often associated with the development of chemoresistance, limiting the effectiveness of currently used anti-cancer therapies. Malfunctioning of the FGF/FGFR axis in cancer cells generates a number of molecular mechanisms that may affect the sensitivity of tumors to the applied drugs. Of key importance is the deregulation of cell signaling, which can lead to increased cell proliferation, survival, and motility, and ultimately to malignancy. Signaling pathways activated by FGFRs inhibit apoptosis, reducing the cytotoxic effect of some anti-cancer drugs. FGFRs-dependent signaling may also initiate angiogenesis and EMT, which facilitates metastasis and also correlates with drug resistance. Therefore, treatment strategies based on FGF/FGFR inhibition (using receptor inhibitors, ligand traps, monoclonal antibodies, or microRNAs) appear to be extremely promising. However, this approach may lead to further development of resistance through acquisition of specific mutations, metabolism switching, and molecular cross-talks. This review brings together information on the mechanisms underlying the involvement of the FGF/FGFR axis in the generation of drug resistance in cancer and highlights the need for further research to overcome this serious problem with novel therapeutic strategies.
Collapse
|
41
|
Wu Y, Chen Q, Zhang Q, Li M, Li H, Jia L, Huang Y, Zhang J. Analysis of whole-exome data of cfDNA and the tumor tissue of non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1453. [PMID: 34734005 PMCID: PMC8506706 DOI: 10.21037/atm-21-4117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022]
Abstract
Background Non-small cell lung cancer (NSCLC) has the highest cancer mortality rate in the world, but currently there is no effective method of dynamic monitoring. Gene mutation is an important factor in tumorigenesis and can be detected using high-throughput sequencing technology. This study aimed to analyze the driving genes in the tumor of NSCLC patients by whole exon sequencing, and to compare and analyze the subclones of the tumor at different time points. Methods We collected 87 cases of NSCLC tumor tissues, para-cancer tissues, and peripheral blood samples for detecting cell-free DNAs (cfDNAs) from January 2016 to December 2018, and whole-exome sequencing was performed. The gene mutation map of NSCLC was drawn in detail by second-generation sequencing data analysis and new driver genes were found. In addition, we performed a subclonal analysis of tumors from different stages of the same patient to further describe the tumor heterogeneity. Results We found that the clonal analysis obtained by cfDNA detection was similar to the clonal analysis of the tissue samples, so real-time monitoring of tumor changes can be carried out through monitoring cfDNA. Conclusions This study provides evidence for studying the gene mutation information of NSCLC and shows the importance of cfDNA in the analysis of tumor subcloning information.
Collapse
Affiliation(s)
- Yuanzhou Wu
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qunqing Chen
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | | | - Man Li
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Li
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Longfei Jia
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Huang
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Vignone A, Biancaniello F, Casadio M, Pesci L, Cardinale V, Ridola L, Alvaro D. Emerging Therapies for Advanced Cholangiocarcinoma: An Updated Literature Review. J Clin Med 2021; 10:4901. [PMID: 34768421 PMCID: PMC8584870 DOI: 10.3390/jcm10214901] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinoma is a group of malignancies with poor prognosis. Treatments for the management of advanced-stage cholangiocarcinoma are limited, and the 5-year survival rate is estimated to be approximately 5-15%, considering all tumor stages. There is a significant unmet need for effective new treatment approaches. The present review is provided with the aim of summarizing the current evidence and future perspectives concerning new therapeutic strategies for cholangiocarcinoma. The role of targeted therapies and immunotherapies is currently investigational in cholangiocarcinoma. These therapeutic options might improve survival outcomes, as shown by the promising results of several clinical trials illustrated in the present review. The co-presence of driver mutations and markers of susceptibility to immunotherapy may lead to rational combination strategies and clinical trial development. A better understanding of immunologically based therapeutic weapons is needed, which will lead to a form of a precision medicine strategy capable of alleviating the clinical aggressiveness and to improve the prognosis of cholangiocarcinoma.
Collapse
Affiliation(s)
- Anthony Vignone
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (L.P.); (L.R.); (D.A.)
| | - Francesca Biancaniello
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (L.P.); (L.R.); (D.A.)
| | - Marco Casadio
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (L.P.); (L.R.); (D.A.)
| | - Ludovica Pesci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (L.P.); (L.R.); (D.A.)
| | - Vincenzo Cardinale
- Department of Medical-Surgical and Biotechnologies Sciences, Polo Pontino, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| | - Lorenzo Ridola
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (L.P.); (L.R.); (D.A.)
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (L.P.); (L.R.); (D.A.)
| |
Collapse
|
43
|
Repetto M, Crimini E, Giugliano F, Morganti S, Belli C, Curigliano G. Selective FGFR/FGF pathway inhibitors: inhibition strategies, clinical activities, resistance mutations, and future directions. Expert Rev Clin Pharmacol 2021; 14:1233-1252. [PMID: 34591728 DOI: 10.1080/17512433.2021.1947246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Fibroblast growth factor receptor (FGFR)/fibroblast growth factor (FGF) is a pathway characterized by recurring alterations in cancer. Its dysregulations enhance cancer cell proliferation, survival, migration and invasion, as well as angiogenesis and immune evasion.Areas covered: FGFR/FGF selective inhibitors belong to a broad class of drugs with some being approved for specific indications and others under investigation in ongoing phase I and II clinical trials. In this review, all available clinical data from trials on selective FGFR/FGF inhibitors as well as described resistance mutations and mechanisms are presented. FGFR/FGF pathway inhibitors are classified according to the mechanism they employ to dampen/suppress signaling and to the preferred FGFR binding mode when X-ray crystal structure is available.Expert opinion: Data presented suggests the general actionability of FGFR1,2,3 mutations and fusions across histologies, whereas FGFR1,2,3 amplifications alone are poor predictors of response to tyrosine kinase inhibitors. Overexpression on immunohistochemistry (IHC) of FGF19, the stimulatory ligand of FGFR4, can predict response to FGFR selective inhibitors in hepatocellular carcinoma. Whereas IHC overexpression of FGFR1,2,3 is not sufficient to predict benefit from FGFR inhibitors across solid tumors. FGFR1,2,3 mRNA overexpression can predict response even in absence of structural alteration. Data on resistance mutations suggests the need for new inhibitors to overcome gatekeeper mutations.
Collapse
Affiliation(s)
- Matteo Repetto
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Edoardo Crimini
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Federica Giugliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefania Morganti
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmen Belli
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
44
|
Genomic and Transcriptomic Characterization of Relapsed SCLC Through Rapid Research Autopsy. JTO Clin Res Rep 2021; 2:100164. [PMID: 34590014 PMCID: PMC8474405 DOI: 10.1016/j.jtocrr.2021.100164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 01/22/2023] Open
Abstract
Introduction Relapsed SCLC is characterized by therapeutic resistance and high mortality rate. Despite decades of research, mechanisms responsible for therapeutic resistance have remained elusive owing to limited tissues available for molecular studies. Thus, an unmet need remains for molecular characterization of relapsed SCLC to facilitate development of effective therapies. Methods We performed whole-exome and transcriptome sequencing of metastatic tumor samples procured from research autopsies of five patients with relapsed SCLC. We implemented bioinformatics tools to infer subclonal phylogeny and identify recurrent genomic alterations. We implemented immune cell signature and single-sample gene set enrichment analyses on tumor and normal transcriptome data from autopsy and additional primary and relapsed SCLC data sets. Furthermore, we evaluated T cell-inflamed gene expression profiles in neuroendocrine (ASCL1, NEUROD1) and non-neuroendocrine (YAP1, POU2F3) SCLC subtypes. Results Exome sequencing revealed clonal heterogeneity (intertumor and intratumor) arising from branched evolution and identified resistance-associated truncal and subclonal alterations in relapsed SCLC. Transcriptome analyses further revealed a noninflamed phenotype in neuroendocrine SCLC subtypes (ASCL1, NEUROD1) associated with decreased expression of genes involved in adaptive antitumor immunity whereas non-neuroendocrine subtypes (YAP1, POU2F3) revealed a more inflamed phenotype. Conclusions Our results reveal substantial tumor heterogeneity and complex clonal evolution in relapsed SCLC. Furthermore, we report that neuroendocrine SCLC subtypes are immunologically cold, thus explaining decreased responsiveness to immune checkpoint blockade. These results suggest that the mechanisms of innate and acquired therapeutic resistances are subtype-specific in SCLC and highlight the need for continued investigation to bolster therapy selection and development for this cancer.
Collapse
|
45
|
Vendramin R, Litchfield K, Swanton C. Cancer evolution: Darwin and beyond. EMBO J 2021; 40:e108389. [PMID: 34459009 PMCID: PMC8441388 DOI: 10.15252/embj.2021108389] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/04/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
Clinical and laboratory studies over recent decades have established branched evolution as a feature of cancer. However, while grounded in somatic selection, several lines of evidence suggest a Darwinian model alone is insufficient to fully explain cancer evolution. First, the role of macroevolutionary events in tumour initiation and progression contradicts Darwin's central thesis of gradualism. Whole-genome doubling, chromosomal chromoplexy and chromothripsis represent examples of single catastrophic events which can drive tumour evolution. Second, neutral evolution can play a role in some tumours, indicating that selection is not always driving evolution. Third, increasing appreciation of the role of the ageing soma has led to recent generalised theories of age-dependent carcinogenesis. Here, we review these concepts and others, which collectively argue for a model of cancer evolution which extends beyond Darwin. We also highlight clinical opportunities which can be grasped through targeting cancer vulnerabilities arising from non-Darwinian patterns of evolution.
Collapse
Affiliation(s)
- Roberto Vendramin
- Cancer Research UK Lung Cancer Centre of ExcellenceUniversity College London Cancer InstituteLondonUK
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of ExcellenceUniversity College London Cancer InstituteLondonUK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of ExcellenceUniversity College London Cancer InstituteLondonUK
- Cancer Evolution and Genome Instability LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
46
|
Cristinziano G, Porru M, Lamberti D, Buglioni S, Rollo F, Amoreo CA, Manni I, Giannarelli D, Cristofoletti C, Russo G, Borad MJ, Grazi GL, Diodoro MG, Giordano S, Sacconi A, Forcato M, Anastasi S, Leonetti C, Segatto O. FGFR2 fusion proteins drive oncogenic transformation of mouse liver organoids towards cholangiocarcinoma. J Hepatol 2021; 75:351-362. [PMID: 33741397 DOI: 10.1016/j.jhep.2021.02.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS About 15% of intrahepatic cholangiocarcinomas (iCCAs) express fibroblast growth factor receptor 2 (FGFR2) fusion proteins (FFs), usually alongside mutational inactivation of TP53, CDKN2A or BAP1. In FFs, FGFR2 residues 1-768 fuse to sequences encoded by a diverse array of partner genes (>60) causing oncogenic FF activation. While FGFR-specific tyrosine kinase inhibitors (F-TKI) provide clinical benefit in FF+ iCCA, responses are partial and/or limited by resistance mechanisms, such as the V565F substitution in the FGFR2 gatekeeper residue. Improving on FF targeting in iCCA therefore remains a critical unmet need. Herein, we aimed to generate a murine model of FF-driven iCCA and use this to uncover actionable FF-associated dependencies. METHODS Four iCCA FFs carrying different fusion sequences were expressed in Tp53-/- mouse liver organoids. Tumorigenic properties of genetically modified liver organoids were assessed by transplantation into immuno-deficient mice. Cellular models derived from neoplastic lesions were exploited for pre-clinical studies. RESULTS Transplantation of FF-expressing liver organoids yielded tumors diagnosed as CCA based on histological, phenotypic and transcriptomic analyses. The penetrance of this tumorigenic phenotype was influenced by FF identity. Tumor organoids and 2D cell lines derived from CCA lesions were addicted to FF signaling via Ras-Erk, regardless of FF identity or V565F mutation. Dual blockade of FF and the Ras-Erk pathway by concomitant pharmacological inhibition of FFs and Mek1/2 provided greater therapeutic efficacy than single agent F-TKI in vitro and in vivo. CONCLUSIONS FF-driven iCCA pathogenesis was successfully modeled on a Tp53-/- murine background, revealing biological heterogeneity among structurally different FFs. Double blockade of FF-ERK signaling deserves consideration for precision-based approaches against human FF+ iCCA. LAY SUMMARY Intrahepatic cholangiocarcinoma (iCCA) is a rare cancer that is difficult to treat. A subtype of iCCA is caused by genomic alterations that generate oncogenic drivers known as FGFR2 fusions. Patients with FGFR2 fusions respond to FGFR inhibitors, but clinical responses are often of modest duration. We used animal and cellular models to show that FGFR2 fusions require the activity of a downstream effector named Mek1/2. We found that dual blockade of FGFR2 fusions and Mek1/2 was more effective than isolated inhibition of FGFR2 fusions, pointing to the potential clinical utility of dual FGFR2-MEK1/2 blockade in patients with iCCA.
Collapse
Affiliation(s)
- Giulia Cristinziano
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Manuela Porru
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Dante Lamberti
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Rollo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carla Azzurra Amoreo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Manni
- SAFU, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Diana Giannarelli
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | | | - Mitesh J Borad
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, USA
| | - Gian Luca Grazi
- Division of Hepatobiliary Pancreatic Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Grazia Diodoro
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Andrea Sacconi
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mattia Forcato
- Center for Genome Research, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sergio Anastasi
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Carlo Leonetti
- SAFU, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Oreste Segatto
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
47
|
FGFR Inhibitors in Oncology: Insight on the Management of Toxicities in Clinical Practice. Cancers (Basel) 2021; 13:cancers13122968. [PMID: 34199304 PMCID: PMC8231807 DOI: 10.3390/cancers13122968] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary FGFR inhibitors evolved as therapeutic options in cholangiocarcinoma and urothelial malignancies. Given the implications of FGFR pathway in various physiological functions, FGFR inhibitors are known to cause unique toxicities. In this review, we summarized the physiology of FGF/FGFR signaling and briefly discussed the possible mechanisms that could lead to FGFR inhibitor resistance and side effects. In addition, we proposed treatment guidelines for the management of FGFR-inhibitor-associated toxicities. Abstract Fibroblast Growth Factor receptor (FGFR) pathway aberrations have been implicated in approximately 7% of the malignancies. As our knowledge of FGFR aberrations in cancer continues to evolve, FGFR inhibitors emerged as potential targeted therapeutic agents. The promising results of pemigatinib and infigratinib in advanced unresectable cholangiocarcinoma harboring FGFR2 fusions or rearrangement, and erdafitinib in metastatic urothelial carcinoma with FGFR2 and FGFR3 genetic aberrations, lead to their accelerated approval by the United States (USA) FDA. Along with these agents, many phase II/III clinical trials are currently evaluating the use of derazantinib, infigratinib, and futibatinib either alone or in combination with immunotherapy. Despite the encouraging results seen with FGFR inhibitors, resistance mechanisms and side effect profile may limit their clinical utility. A better understanding of the unique FGFR-inhibitor-related toxicities would invariably help us in the prevention and effective management of FGFR-inhibitor-induced adverse events thereby enhancing their clinical benefit. Herein, we summarized the physiology of FGF/FGFR signaling and briefly discussed the possible mechanisms that could lead to FGFR inhibitor resistance and side effects. In addition, we proposed treatment guidelines for the management of FGFR-inhibitor-associated toxicities. This work would invariably help practicing oncologists to effectively manage the unique toxicities of FGFR inhibitors.
Collapse
|
48
|
Panicker S, Venkatabalasubramanian S, Pathak S, Ramalingam S. The impact of fusion genes on cancer stem cells and drug resistance. Mol Cell Biochem 2021; 476:3771-3783. [PMID: 34095988 DOI: 10.1007/s11010-021-04203-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022]
Abstract
With ever increasing evidences on the role of fusion genes as the oncogenic protagonists in myriad cancers, it's time to explore if fusion genes can be the next generational drug targets in meeting the current demands of higher drug efficacy. Eliminating cancer stem cells (CSC) has become the current focus; however, we have reached a standstill in drug development owing to the lack of effective strategies to eradicate CSC. We believe that fusion genes could be the novel targets to overcome this limitation. The intriguing feature of fusion genes is that it dominantly impacts every aspect of CSC including self-renewal, differentiation, lineage commitment, tumorigenicity and stemness. Given the clinical success of fusion gene-based drugs in hematological cancers, our attempt to target fusion genes in eradicating CSC can be rewarding. As fusion genes are expressed explicitly in cancer cells, eradicating CSC by targeting fusion genes provides yet an another advantage of negligible patient side effects since normal cells remain unaffected by the drug. We hereby delineate the latest evidences on how fusion genes regulate CSC and drug resistance.
Collapse
Affiliation(s)
- Saurav Panicker
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram, 603203, Tamil Nadu, India
| | | | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Chennai, 603103, Tamil Nadu, India
| | - Satish Ramalingam
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram, 603203, Tamil Nadu, India.
| |
Collapse
|
49
|
Casadio M, Biancaniello F, Overi D, Venere R, Carpino G, Gaudio E, Alvaro D, Cardinale V. Molecular Landscape and Therapeutic Strategies in Cholangiocarcinoma: An Integrated Translational Approach towards Precision Medicine. Int J Mol Sci 2021; 22:5613. [PMID: 34070643 PMCID: PMC8199244 DOI: 10.3390/ijms22115613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cholangiocarcinomas (CCAs) are heterogeneous biliary tract malignancies with dismal prognosis, mainly due to tumor aggressiveness, late diagnosis, and poor response to current therapeutic options. High-throughput technologies have been used as a fundamental tool in unveiling CCA molecular landscape, and several molecular classifications have been proposed, leading to various targeted therapy trials. In this review, we aim to analyze the critical issues concerning the status of precision medicine in CCA, discussing molecular signatures and clusters, related to both anatomical classification and different etiopathogenesis, and the latest therapeutic strategies. Furthermore, we propose an integrated approach comprising the CCA molecular mechanism, pathobiology, clinical and histological findings, and treatment perspectives for the ultimate purpose of improving the methods of patient allocations in clinical trials and the response to personalized therapies.
Collapse
Affiliation(s)
- Marco Casadio
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (R.V.); (D.A.)
| | - Francesca Biancaniello
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (R.V.); (D.A.)
| | - Diletta Overi
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Via Borelli 50, 00161 Rome, Italy; (D.O.); (E.G.)
| | - Rosanna Venere
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (R.V.); (D.A.)
| | - Guido Carpino
- Department of Movement, Human and Health Sciences, Division of Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Rome, Italy;
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Via Borelli 50, 00161 Rome, Italy; (D.O.); (E.G.)
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell’Università 37, 00185 Rome, Italy; (M.C.); (R.V.); (D.A.)
| | - Vincenzo Cardinale
- Medical-Surgical and Biotechnologies Sciences, Polo Pontino, Sapienza University of Rome, Corso della Repubblica 79, 04100 Latina, Italy;
| |
Collapse
|
50
|
Bekaii-Saab TS, Bridgewater J, Normanno N. Practical considerations in screening for genetic alterations in cholangiocarcinoma. Ann Oncol 2021; 32:1111-1126. [PMID: 33932504 DOI: 10.1016/j.annonc.2021.04.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/06/2021] [Accepted: 04/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinoma (CCA) encompasses diverse epithelial tumors historically associated with poor outcomes due to an aggressive disease course, late diagnosis, and limited benefit of standard chemotherapy for advanced disease. Comprehensive molecular profiling has revealed a diverse landscape of genomic alterations as oncogenic drivers in CCA. TP53 mutations, CDKN2A/B loss, and KRAS mutations are the most common genetic alterations in CCA. However, intrahepatic CCA (iCCA) and extrahepatic CCA (eCCA) differ substantially in the frequency of many alterations. This includes actionable alterations, such as isocitrate dehydrogenase 1 (IDH1) mutations and a large variety of FGFR2 rearrangements, which are found in up to 29% and ∼10% of patients with iCCA, respectively, but are rare in eCCA. FGFR2 rearrangements are currently the only genetic alteration in CCA for which a targeted therapy, the fibroblast growth factor receptor 1-3 inhibitor pemigatinib, has been approved. However, favorable phase III results for IDH1-targeted therapy with ivosidenib in iCCA have been published, and numerous other alterations are actionable by targeted therapies approved in other indications. Recent advances in next-generation sequencing (NGS) have led to the development of assays that allow comprehensive genomic profiling of large gene panels within 2-3 weeks, including in vitro diagnostic tests approved in the United States. These assays vary regarding acceptable source material (tumor tissue or peripheral whole blood), genetic source for library construction (DNA or RNA), target selection technology, gene panel size, and type of detectable genomic alterations. While some large commercial laboratories offer rapid and comprehensive genomic profiling services based on proprietary assay platforms, clinical centers may use commercial genomic profiling kits designed for clinical research to develop their own customized laboratory-developed tests. Large-scale genomic profiling based on NGS allows for a detailed and precise molecular diagnosis of CCA and provides an important opportunity for improved targeted treatment plans tailored to the individual patient's genetic signature.
Collapse
Affiliation(s)
| | - J Bridgewater
- University College London Cancer Institute, London, UK
| | - N Normanno
- Istituto Nazionale Tumori 'Fondazione Giovanni Pascale' IRCCS, Naples, Italy
| |
Collapse
|