1
|
Chen J, Huang Y, Zuo D, Shan R, Li S, Li R, Hua D, Zhan Q, Song X, Chen Y, Ma P, Ma L, Tao G, Shu Y. Dysregulated lipids homeostasis disrupts CHAC1-mediated ferroptosis driving fibroblast growth factor receptor tyrosine kinase inhibitor AZD4547 resistance in gastric cancer. Redox Biol 2025; 84:103693. [PMID: 40460553 PMCID: PMC12167034 DOI: 10.1016/j.redox.2025.103693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/17/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025] Open
Abstract
AIMS This study investigates the mechanisms underlying acquired resistance to FGFR tyrosine kinase inhibitor (FGFR-TKI) in gastric cancer (GC), focusing on the interplay between ferroptosis and lipid metabolism of tumor cells. METHODS We constructed FGFR-TKI-resistant cell lines from GC cells. RNA sequencing was performed to identify differentially expressed genes (DEGs) related to ferroptosis and assess lipid metabolism in resistant cells. GC microenvironment lipid profile was characterized by HPLC-MS/MS lipidomics. The effects of CHAC1 and cholesterol synthesis modulation on ferroptosis and FGFR-TKI resistance were assessed using in vitro and in vivo models. RESULTS We found that FGFR-TKI can induce ferroptosis in FGFR-TKI-sensitive cells, while resistant cells exhibit decreased sensitivity to ferroptosis due to reduced CHAC1 expression, a key glutathione-specific degrading enzyme. Overexpression of CHAC1 enhances FGFR-TKI cytotoxicity. Additionally, cholesterol accumulation in resistant cells, associated with diminished stearic acid (SA) uptake, confers FGFR-TKI-induced ferroptosis resistance. In vivo studies show that CHAC1 overexpression or cholesterol synthesis inhibition can reverse FGFR-TKI resistance, which is dependent on ferroptosis. CONCLUSIONS Dysregulated lipid homeostasis downregulated CHAC1-mediated ferroptosis, leading to FGFR-TKI resistance in gastric cancer. Overexpression of CHAC1 or inhibiting cholesterol synthesis presents promising therapeutic strategies to overcome FGFR-TKI resistance in GC.
Collapse
Affiliation(s)
- Jingwen Chen
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, PR China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, PR China
| | - Yedi Huang
- Department of Immunology, School of Basic Medical Sciences, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, 210000, PR China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, PR China
| | - Daocheng Zuo
- Department of Immunology, School of Basic Medical Sciences, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, 210000, PR China
| | - Ruimin Shan
- Department of Immunology, School of Basic Medical Sciences, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, 210000, PR China
| | - Songmao Li
- Department of Immunology, School of Basic Medical Sciences, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, 210000, PR China
| | - Ran Li
- Department of Immunology, School of Basic Medical Sciences, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, 210000, PR China
| | - Dong Hua
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, School of Medicine, Jiangnan University, Wuxi, 214000, PR China
| | - Qiang Zhan
- Departments of Gastroenterology, the Affiliated Wuxi People's Hospital of Nanjing Medical University & Department of Medical Genetics, Nanjing Medical University, Nanjing, 210000, PR China
| | - Xudong Song
- Department of Gastrointestinal surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, 223300, PR China
| | - Yun Chen
- Department of Immunology, School of Basic Medical Sciences, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, 210000, PR China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, PR China.
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, PR China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, PR China.
| | - Ling Ma
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, PR China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, PR China.
| | - Guoquan Tao
- Gusu School, Nanjing Medical University, Suzhou, 211166, PR China.
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, PR China; Gusu School, Nanjing Medical University, Suzhou, 211166, PR China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, PR China.
| |
Collapse
|
2
|
Satoh T, Barthélémy P, Nogova L, Honda K, Hirano H, Lee KW, Rha SY, Ryu MH, Park JO, Doi T, Ajani J, Hangai N, Kremer J, Mina M, Liu M, Shitara K. Phase 2 study of futibatinib in patients with gastric or gastroesophageal junction cancer harboring FGFR2 amplifications. Eur J Cancer 2025; 218:115262. [PMID: 39919334 DOI: 10.1016/j.ejca.2025.115262] [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: 10/29/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND AND AIMS Aberrant fibroblast growth factor receptor (FGFR)-driven signaling, predominantly arising from FGFR2 amplification, plays a key role in gastric cancer pathogenesis. This open-label, phase 2 study evaluated the efficacy and safety of futibatinib, an irreversible FGFR1-4 inhibitor, in patients with gastric or gastroesophageal junction (GEJ) cancer harboring FGFR2 amplifications. METHODS Patients were treated with futibatinib 20 mg orally once daily in a 28-day cycle. The primary endpoint was objective response rate (ORR) per independent central review. Secondary endpoints included progression-free survival (PFS), overall survival (OS), and safety. RESULTS Among 28 treated patients, the ORR per independent central review was 17.9 %, comprising five patients with a partial response (median duration of response, 3.9 months), and an additional nine patients with stable disease for a disease control rate of 50.0 %. Median PFS per independent central review and median OS were 2.9 and 5.9 months, respectively. The most common treatment-related adverse events (any grade) were hyperphosphatemia (89.3 %), decreased appetite (32.1 %), and increased aspartate aminotransferase (21.4 %). Only one (3.6 %) patient discontinued study treatment due to an adverse event. Futibatinib demonstrated modest antitumor activity with a safety profile consistent with previous reports in patients with gastric or GEJ cancer harboring FGFR2 amplifications, potentially warranting further investigation.
Collapse
Affiliation(s)
| | | | - Lucia Nogova
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | - Keun-Wook Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Oh Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Jaffer Ajani
- University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nanae Hangai
- Taiho Oncology, Inc., Princeton, NJ, United States
| | - Jill Kremer
- Taiho Oncology, Inc., Princeton, NJ, United States
| | - Mark Mina
- Taiho Oncology, Inc., Princeton, NJ, United States
| | - Mei Liu
- Taiho Oncology, Inc., Princeton, NJ, United States
| | - Kohei Shitara
- National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
3
|
Morizane C, Ueno M, Ioka T, Tajika M, Ikeda M, Yamaguchi K, Hara H, Yabusaki H, Miyamoto A, Iwasa S, Muto M, Takashima T, Minashi K, Komatsu Y, Nishina T, Nakajima TE, Takeno A, Moriwaki T, Furukawa M, Sahara T, Ikezawa H, Nomoto M, Takashima S, Uehara T, Funasaka S, Yashiro M, Furuse J. Tasurgratinib in patients with cholangiocarcinoma or gastric cancer: Expansion part of the first-in-human phase I study. Cancer Sci 2025; 116:192-203. [PMID: 39462221 PMCID: PMC11711049 DOI: 10.1111/cas.16354] [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: 03/18/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 10/29/2024] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are a highly conserved family of transmembrane receptor tyrosine kinases with multiple roles in the regulation of key cellular processes. Specific FGFR mutations have been observed in several types of cancers, including gastric carcinoma and cholangiocarcinoma. Dose escalation data of 24 Japanese patients with solid tumors treated with Tasurgratinib (previously known as E7090), a potent, selective FGFR1-3 inhibitor, was reported in a phase I, first-in-human, single-center study. Based on the safety, pharmacokinetic, and pharmacodynamic profiles observed in this study, the recommended dose of 140 mg once daily was selected for the expansion part (Part 2), a multicenter expansion of the dose-finding study restricted to patients with tumors harboring FGFR gene alterations. Safety and preliminary efficacy were assessed in Part 2. Pharmacodynamic pharmacogenomic markers (serum phosphate, FGF23, and 1,25-(OH)2-vitamin D, circulating tumor DNA) and pharmacokinetic profiles were also evaluated. A total of 16 patients were enrolled in Part 2, six with cholangiocarcinoma and 10 with gastric cancer. The most common treatment-emergent adverse events were hyperphosphatemia, palmar-plantar erythrodysesthesia syndrome, and paronychia. Five partial responses (83.3%) in cholangiocarcinoma patients and one partial response (11.1%) in gastric cancer patients were observed; median progression-free survival was 8.26 months (95% confidence interval [CI] 3.84, not evaluable [NE]) and 3.25 months (95% CI 0.95, 4.86), and overall survival was 22.49 months (95% CI 6.37, NE) and 4.27 months (95% CI 2.23, 7.95), respectively, in the two groups. In conclusion, Tasurgratinib 140 mg has a tolerable safety profile with good clinical efficacy in patients with cholangiocarcinoma harboring FGFR2 gene rearrangements.
Collapse
Affiliation(s)
| | | | - Tatsuya Ioka
- Oncology CenterYamaguchi University HospitalUbeJapan
| | | | | | - Kensei Yamaguchi
- The Cancer Institute HospitalJapanese Foundation for Cancer ResearchTokyoJapan
| | | | | | - Atsushi Miyamoto
- National Hospital Organization Osaka National HospitalOsakaJapan
| | | | | | | | | | | | - Tomohiro Nishina
- National Hospital Organization Shikoku Cancer CenterMatsuyamaJapan
| | - Takako Eguchi Nakajima
- St. Marianna University School of MedicineKawasakiJapan
- Department of Early Clinical DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | | | | | | | | | | | | | | | | | | | - Masakazu Yashiro
- Graduate School of MedicineOsaka Metropolitan UniversityOsakaJapan
| | | |
Collapse
|
4
|
Kendziora B, Flaig M, Maurer M. [Hand-foot syndrome after taking fibroblast growth factor receptor 2 inhibitor]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:12-14. [PMID: 39278873 DOI: 10.1007/s00105-024-05339-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 09/18/2024]
Affiliation(s)
- Benjamin Kendziora
- Klinik und Poliklinik für Dermatologie und Allergologie, Klinikum der Universität München, LMU München, Frauenlobstr. 9-11, 80337, München, Deutschland.
- Institut für Versorgungsforschung in der Dermatologie und bei Pflegeberufen (IVDP), Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Deutschland.
| | - Michael Flaig
- Klinik und Poliklinik für Dermatologie und Allergologie, Klinikum der Universität München, LMU München, Frauenlobstr. 9-11, 80337, München, Deutschland
| | - Michaela Maurer
- Klinik und Poliklinik für Dermatologie und Allergologie, Klinikum der Universität München, LMU München, Frauenlobstr. 9-11, 80337, München, Deutschland
| |
Collapse
|
5
|
Lau DK, Collin JP, Mariadason JM. Clinical Developments and Challenges in Treating FGFR2-Driven Gastric Cancer. Biomedicines 2024; 12:1117. [PMID: 38791079 PMCID: PMC11118914 DOI: 10.3390/biomedicines12051117] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Recent advances in the treatment of gastric cancer (GC) with chemotherapy, immunotherapy, anti-angiogenic therapy and targeted therapies have yielded some improvement in survival outcomes; however, metastatic GC remains a lethal malignancy and amongst the leading causes of cancer-related mortality worldwide. Importantly, the ongoing molecular characterisation of GCs continues to uncover potentially actionable molecular targets. Among these, aberrant FGFR2-driven signalling, predominantly arising from FGFR2 amplification, occurs in approximately 3-11% of GCs. However, whilst several inhibitors of FGFR have been clinically tested to-date, there are currently no approved FGFR-directed therapies for GC. In this review, we summarise the significance of FGFR2 as an actionable therapeutic target in GC, examine the recent pre-clinical and clinical data supporting the use of small-molecule inhibitors, antibody-based therapies, as well as novel approaches such as proteolysis-targeting chimeras (PROTACs) for targeting FGFR2 in these tumours, and discuss the ongoing challenges and opportunities associated with their clinical development.
Collapse
Affiliation(s)
- David K. Lau
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Department of Oncology, Monash Health, Clayton, VIC 3168, Australia
| | - Jack P. Collin
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| | - John M. Mariadason
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| |
Collapse
|
6
|
Tsimafeyeu I, Raskin G. Challenges of FGFR2 Testing in Gastric Cancer. Oncol Rev 2023; 17:11790. [PMID: 38155920 PMCID: PMC10752926 DOI: 10.3389/or.2023.11790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Affiliation(s)
- Ilya Tsimafeyeu
- Bureau for Cancer Research, New York Office, New York, NY, United States
| | | |
Collapse
|
7
|
A phase 1b study of the allosteric extracellular FGFR2 inhibitor alofanib in patients with pretreated advanced gastric cancer. Invest New Drugs 2023; 41:324-332. [PMID: 36907947 DOI: 10.1007/s10637-023-01340-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 03/14/2023]
Abstract
Alofanib is a small-molecule allosteric extracellular FGFR2 inhibitor. We report safety and preliminary efficacy from the first-in-human phase 1b study of alofanib in heavily pretreated patients with advanced gastric cancer. The standard dose-escalation design 3+3 aimed to establish the maximum tolerated dose (MTD) or recommended phase 2 dose (RP2D). Alofanib was administered daily intravenously 5 days on, 2 days off. There were five dose levels (50-350 mg/m2). All patients received alofanib until disease progression or unacceptable toxicity. 21 patients were enrolled. Patients were predominantly male (71%), 67% had 2 and more metastatic sites, including liver metastases (43%), 19% had ECOG PS 2, and were heavily pretreated (86% had previous 2 and more treatment lines). During dose escalation, no dose-limiting toxicities were observed, and MTD was not defined. 15 (71.4%) patients had at least one adverse event associated with the treatment (TRAE). Grade 3 or higher TRAEs were observed in 6 patients (28.6%). The most common TRAEs included reactions immediately after administration, diarrhea, thrombocytopenia, arthralgia, and headache. The median progression-free survival and overall survival was 3.63 (95% CI 1.58-5.68) and 7.0 (95% CI 3.82-10.18) months, respectively. The 6- and 12-month overall survival rates were 57.1% and 33.3%. Disease control rate was 68% with one durable partial response. The MTD has not been reached and dose of 350 mg/m2, 5 days on, 2 days off has been declared as RP2D. Alofanib showed acceptable tolerability and preliminary signs of clinical activity in the late-line treatment of metastatic gastric cancer. (ClinicalTrials.gov identifier: NCT04071184).
Collapse
|
8
|
Kleo K, Jovanovic VM, Arndold A, Lehmann A, Lammert H, Berg E, Harloff H, Treese C, Hummel M, Daum S. Response prediction in patients with gastric and esophagogastric adenocarcinoma under neoadjuvant chemotherapy using targeted gene expression analysis and next-generation sequencing in pre-therapeutic biopsies. J Cancer Res Clin Oncol 2023; 149:1049-1061. [PMID: 35246724 PMCID: PMC9984352 DOI: 10.1007/s00432-022-03944-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/02/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Perioperative chemo-(radio-) therapy is the accepted standard in European patients with locally advanced adenocarcinoma of the esophagogastric junction or stomach (AEG/AS). However, 30-85% of patients do not respond to this treatment. The aim of our study was the identification of predictive biomarkers in pre-therapeutic endoscopic tumor biopsies from patients with histopathologic response (Becker-1) versus non-response (Becker-2/3) to preoperative chemotherapy. METHODS Formalin-fixed paraffin-embedded biopsies from 36 Caucasian patients (Becker-1 n = 11, Becker-2 n = 7, Becker-3 n = 18) with AEG/AS, taken prior to neoadjuvant chemotherapy were selected. For RNA expression analysis, we employed the NanoString nCounter System. To identify genomic alterations like single nucleotide variants (SNV), copy number variation (CNV) and fusion events, we used Illumina TST170 gene panel. For HER2 and FGFR2 protein expression, immunostaining was performed. Furthermore, we analyzed the microsatellite instability (MSI) and Epstein-Barr virus (EBV) infection status by EBER in situ hybridization. RESULTS Heat map and principal component analyses showed no clustering by means of gene expression according to regression grade. Concerning two recently proposed predictive markers, our data showed equal distribution for MSI (Becker-1: 2; Becker-2: 1; Becker-3: 3; out of 29 tested) and EBV infection was rare (1/32). We could not reveal discriminating target genes concerning SNV, but found a higher mutational burden in non-responders versus responders and fusion (in 6/14) and CNV events (in 5/14) exclusively in Becker-3. CONCLUSIONS Although we could not identify discriminating target genes, our data suggest that molecular alterations are in general more prevalent in patients with AEG/AS belonging to the non-responding Becker group 3.
Collapse
Affiliation(s)
- Karsten Kleo
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Vladimir M Jovanovic
- Institute of Informatics, Bioinformatics Solution Center, Freie Universität (FU), Takustr. 9, 14195, Berlin, Germany
| | - Alexander Arndold
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Annika Lehmann
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Hedwig Lammert
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Erika Berg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Hannah Harloff
- Medical Department, Division of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Christoph Treese
- Medical Department, Division of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
- Experimental and Clinical Research Center, Charité University Medicine, Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Chariteplatz 1, 10117, Berlin, Germany
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Michael Hummel
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Severin Daum
- Medical Department, Division of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany.
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
9
|
Yu X, He S, Shen J, Huang Q, Yang P, Huang L, Pu D, Wang L, Li L, Liu J, Liu Z, Zhu L. Tumor vessel normalization and immunotherapy in gastric cancer. Ther Adv Med Oncol 2022; 14:17588359221110176. [PMID: 35872968 PMCID: PMC9297465 DOI: 10.1177/17588359221110176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a common malignant tumor, and patients with GC have a low survival rate due to limited effective treatment methods. Angiogenesis and immune evasion are two key processes in GC progression, and they act synergistically to promote tumor progression. Tumor vascular normalization has been shown to improve the efficacy of cancer immunotherapy, which in turn may be improved through enhanced immune stimulation. Therefore, it may be interesting to identify synergies between immunomodulatory agents and anti-angiogenic therapies in GC. This strategy aims to normalize the tumor microenvironment through the action of the anti-vascular endothelial growth factor while stimulating the immune response through immunotherapy and prolonging the survival of GC patients.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Shan He
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jian Shen
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Qiushi Huang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Peng Yang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Lin Huang
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dan Pu
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Wang
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Lu Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Jinghua Liu
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, Shandong 276000, People's Republic of China
| | - Zelong Liu
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, No. 37, Guo Xue Xiang, Wuhou District, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
10
|
Clinical Response to Futibatinib in Patients with High-Level FGFR2-Amplified Advanced Gastric Cancer: Two Case Reports. Clin Drug Investig 2022; 42:697-701. [PMID: 35854171 DOI: 10.1007/s40261-022-01183-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
|
11
|
Yan YH, Lei XY, Hu WP. Comparative effectiveness and tolerability of targeted agents combined with chemotherapy in patients with HER2-positive gastroesophageal cancer: A network meta-analysis. Saudi J Gastroenterol 2022; 28:175-185. [PMID: 34747874 PMCID: PMC9212117 DOI: 10.4103/sjg.sjg_367_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/14/2021] [Indexed: 12/07/2022] Open
Abstract
Background Gastric cancer (GC) or gastroesophageal junction (GEJ) cancer with HER2 overexpression is highly invasive, with a poor prognosis. With the development of new targeted agents, which agents have ideal therapeutic effects must be determined. This network meta-analysis analyzed the effectiveness and tolerability of targeted agents combined with chemotherapy in HER2-positive GC/GEJ cancer. Methods Public databases were searched from the date of inception to October 22, 2020. Randomized controlled trials (RCTs) on targeted agent-related regimens for HER2-positive advanced GC or GEJ cancer were included. Subgroup analyses based on publication language, first-line treatment, second/third-line treatment, and HER2 staining intensity were performed. Results In total, 13 articles were included. The trastuzumabderuxtecan (TraD) and pertuzumab plus trastuzumab and chemotherapy (PerTraChemo) regimens were considered to have high effectiveness but low tolerability. In the subgroup analysis, PerTraChemo still had high effectiveness with low tolerability as the first-line therapy. As the second- or third-line therapy, TraD and lapatinib plus chemotherapy (LapChemo) had high effectiveness and moderate tolerability. In terms of overall survival (OS) time, PerTraChemo had a relative advantage in the immunohistochemistry (IHC) 2+/in situ hybridization (ISH)+ population, whereas TraD, PerTraChemo, and trastuzumab plus chemotherapy (TraChemo) had a relative advantage in the IHC3+ population. Conclusion TraD had relative advantages as the second- or third-line therapy and in the IHC3 + population. PerTraChemo is a potential first-line therapy, but it requires further confirmation because the JACOB phase III clinical trial failed to confirm the superiority of PerTraChemo over TraChemo with regard to OS.
Collapse
Affiliation(s)
- Yin-Hong Yan
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiao-Yi Lei
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wei-Ping Hu
- Department of Nephrological, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Department of Nephrological Department, The Fifth Hospital of Xiamen, Xiamen, China
| |
Collapse
|
12
|
Rozen EJ, Shohet JM. Systematic review of the receptor tyrosine kinase superfamily in neuroblastoma pathophysiology. Cancer Metastasis Rev 2022; 41:33-52. [PMID: 34716856 PMCID: PMC8924100 DOI: 10.1007/s10555-021-10001-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Neuroblastoma is a devastating disease accounting for 15% of all childhood cancer deaths. Yet, our understanding of key molecular drivers such as receptor tyrosine kinases (RTKs) in this pathology remains poorly clarified. Here, we provide a systematic analysis of the RTK superfamily in the context of neuroblastoma pathogenesis. METHODS Statistical correlations for all RTK family members' expression to neuroblastoma patient survival across 10 independent patient cohorts were annotated, synthesized, and ranked using the R2: Genomics Analysis and Visualization Platform. Gene expression of selected members across different cancer cell lines was further analyzed in the Cancer Cell Line Encyclopedia, part of the Cancer Dependency Map portal (depmap portal ( http://depmap.org )). Finally, we provide a detailed literature review for highly ranked candidates. RESULTS Our analysis defined two subsets of RTKs showing robust associations with either better or worse survival, constituting potential novel players in neuroblastoma pathophysiology, diagnosis, and therapy. We review the available literature regarding the oncogenic functions of these RTKs, their roles in neuroblastoma pathophysiology, and potential utility as therapeutic targets. CONCLUSIONS Our systematic analysis and review of the RTK superfamily in neuroblastoma pathogenesis provides a new resource to guide the research community towards focused efforts investigating signaling pathways that contribute to neuroblastoma tumor establishment, growth, and/or aggressiveness and targeting these druggable molecules in novel therapeutic strategies.
Collapse
Affiliation(s)
- Esteban Javier Rozen
- Department of Pediatrics, UMass Chan Medical School, Lazare Research Building LRB603, 364 Plantation Street, Worcester, MA, 01605, USA.
| | - Jason Matthew Shohet
- Division of Hematology/Oncology, Department of Pediatrics, UMass Chan Medical School, Lazare Research Building LRB603, 364 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
13
|
Schrumpf T, Behrens HM, Haag J, Krüger S, Röcken C. FGFR2 overexpression and compromised survival in diffuse-type gastric cancer in a large central European cohort. PLoS One 2022; 17:e0264011. [PMID: 35167603 PMCID: PMC8846517 DOI: 10.1371/journal.pone.0264011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/01/2022] [Indexed: 12/24/2022] Open
Abstract
The significance of fibroblast growth factor receptor 2 (FGFR2) in gastric cancer (GC) has been studied predominantly in Asian patient cohorts. Data on White patients are scarce. Here, we aimed to independently validate the expression and putative tumor biological significance of FGFR2 in a large non-Asian GC cohort. Immunohistochemistry (IHC) was performed on large-area tissue sections from 493 patients with GC and evaluated using the HScore. GCs with moderate and strong FGFR2 expression were studied for Fgfr2 amplification using chromogenic in situ hybridization (CISH). Median overall survival was determined using the Kaplan–Meier method. The majority [240 (99.1%)] of FGFR2-positive GCs showed a variable combination of staining intensities with marked intratumoral heterogeneity, including weak [198 (40.2%) cases], moderate [145 (29.4%)], and strong [108 (21.9%)] staining in diverse combinations. 250 (50.9%) GCs expressed no FGFR2. Fgfr2 gene amplification was found in 40% of selected cases with high protein expression and was also heterogeneous at the cell level. FGFR2 protein expression did not correlate with patient survival in the entire cohort However, using different cutoff values, a negative correlation between FGFR2-expression and patient outcome was found for diffuse-type GC. FGFR2 expression was associated with a lower tumor grade and intestinal phenotype (p≤0.0001). FGFR2–positive diffuse-type GCs classify a small subset of patients with a poor tumor specific survival (5.29±1.3 vs. 14.67±1.9 months; p = 0.004).
Collapse
Affiliation(s)
- Thorben Schrumpf
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hans-Michael Behrens
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jochen Haag
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sandra Krüger
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christoph Röcken
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
- * E-mail:
| |
Collapse
|
14
|
Meric-Bernstam F, Bahleda R, Hierro C, Sanson M, Bridgewater J, Arkenau HT, Tran B, Kelley RK, Park JO, Javle M, He Y, Benhadji KA, Goyal L. Futibatinib, an Irreversible FGFR1-4 Inhibitor, in Patients with Advanced Solid Tumors Harboring FGF/ FGFR Aberrations: A Phase I Dose-Expansion Study. Cancer Discov 2022; 12:402-415. [PMID: 34551969 PMCID: PMC9762334 DOI: 10.1158/2159-8290.cd-21-0697] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/16/2021] [Accepted: 09/20/2021] [Indexed: 01/07/2023]
Abstract
Futibatinib, a highly selective, irreversible FGFR1-4 inhibitor, was evaluated in a large multihistology phase I dose-expansion trial that enrolled 197 patients with advanced solid tumors. Futibatinib demonstrated an objective response rate (ORR) of 13.7%, with responses in a broad spectrum of tumors (cholangiocarcinoma and gastric, urothelial, central nervous system, head and neck, and breast cancer) bearing both known and previously uncharacterized FGFR1-3 aberrations. The greatest activity was observed in FGFR2 fusion/rearrangement-positive intrahepatic cholangiocarcinoma (ORR, 25.4%). Some patients with acquired resistance to a prior FGFR inhibitor also experienced responses with futibatinib. Futibatinib demonstrated a manageable safety profile. The most common treatment-emergent adverse events were hyperphosphatemia (81.2%), diarrhea (33.5%), and nausea (30.4%). These results formed the basis for ongoing futibatinib phase II/III trials and demonstrate the potential of genomically selected early-phase trials to help identify molecular subsets likely to benefit from targeted therapy. SIGNIFICANCE: This phase I dose-expansion trial demonstrated clinical activity and tolerability of the irreversible FGFR1-4 inhibitor futibatinib across a broad spectrum of FGFR-aberrant tumors. These results formed the rationale for ongoing phase II/III futibatinib trials in cholangiocarcinoma, breast cancer, gastroesophageal cancer, and a genomically selected disease-agnostic population.This article is highlighted in the In This Issue feature, p. 275.
Collapse
Affiliation(s)
- Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Rastislav Bahleda
- Early Drug Development Department (DITEP), Gustave Roussy Cancer Center, Villejuif, France
| | - Cinta Hierro
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH) and Institute of Oncology (VHIO), Barcelona, Spain
| | - Marc Sanson
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle Epinière, and AP-HP Hôpitaux Universitaires La Pitié Salpêtrière Charles Foix, Service de Neurologie 2 Mazarin, Paris, France
| | | | | | - Ben Tran
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Joon Oh Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Milind Javle
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yaohua He
- Taiho Oncology, Inc., Princeton, New Jersey
| | | | - Lipika Goyal
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| |
Collapse
|
15
|
Li J, Hu K, Huang J, Zhou L, Yan Y, Xu Z. A Pancancer Analysis of the Expression Landscape and Clinical Relevance of Fibroblast Growth Factor Receptor 2 in Human Cancers. Front Oncol 2021; 11:644854. [PMID: 33968743 PMCID: PMC8097147 DOI: 10.3389/fonc.2021.644854] [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: 12/23/2020] [Accepted: 02/23/2021] [Indexed: 02/05/2023] Open
Abstract
Background: Fibroblast growth factor receptor 2 (FGFR2) is frequently altered in tumors and one of the top therapeutic targets in cholangiocarcinoma (CHOL) with FGFR2 fusions. Although there have been several studies on individual tumors, a comprehensive analysis of FGFR2 genetic aberrations and their simultaneous clinical implications across different tumors have not been reported. Methods: In this study, we used the large comprehensive datasets available, covering over 10,000 tumor samples across more than 30 cancer types, to analyze FGFR2 abnormal expression, methylation, alteration (mutations/fusions and amplification/deletion), and their clinical associations. Results: Alteration frequency, mutation location distribution, oncogenic effects, and therapeutic implications varied among different cancers. The overall mutation rate of FGFR2 is low in pancancer. CHOL had the highest mutation frequency, and fusion accounted for the major proportion. All these fusion aberrations in CHOL were targetable, and an FDA-approved drug was approved recently. Uterine corpus endometrial carcinoma (UCEC) had the highest number of FGFR2 mutations, and the most frequently mutated positions were S252W and N549K, where the functional impact was oncogenic, but targeted therapy was less effective. Additionally, DNA methylation was associated with FGFR2 expression in several cancers. Moreover, FGFG2 expression and genetic aberrations showed clinical associations with patient survival in several cancers, indicating their potential for application as new tumor markers and therapeutic targets. Conclusions: This study showed the full FGFR2 alteration spectrum and provided a broad molecular perspective of FGFR2 in a comprehensive manner, suggesting some new directions for clinical targeted therapy of cancers.
Collapse
Affiliation(s)
- Juanni Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Kuan Hu
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jinzhou Huang
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Lei Zhou
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Integrated analysis of DNA methylation and mRNA expression profiles to identify key genes in head and neck squamous cell carcinoma. Biosci Rep 2021; 40:221746. [PMID: 31894857 PMCID: PMC6981101 DOI: 10.1042/bsr20193349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023] Open
Abstract
DNA methylation has been demonstrated to play significant roles in the etiology and pathogenesis of head and neck squamous cell carcinoma (HNSCC). In the present study, methylation microarray dataset (GSE87053) and gene expression microarray dataset (GSE23558) were downloaded from GEO database and analyzed through R language. A total of 255 hypermethylated-downregulated genes and 114 hypomethylated-upregulated genes were finally identified. Functional enrichment analyses were performed and a comprehensive protein–protein interaction (PPI) network was constructed. Subsequently, the top ten hub genes selected by Cytoscape software were subjected to further analyses. It was illustrated that the expression level of CSF2, CTLA4, ETS1, PIK3CD, and CFTR was intimately associated with HNSCC. Survival analysis suggested that CTLA4 and FGFR2 could serve as effective independent prognostic biomarkers for HNSCC patients. Overall, our study lay a groundwork for further investigation into the underlying molecular mechanisms in HNSCC carcinogenesis, providing potential biomarkers and therapeutic targets for HNSCC.
Collapse
|
17
|
Hu K, Yu W, Ajayi OE, Li L, Huang Z, Rong Q, Wang S, Wu QF. Integrated analysis of whole genome and transcriptome sequencing in a young patient with gastric cancer provides insights for precision therapy. Oncol Lett 2020; 20:115. [PMID: 32863928 PMCID: PMC7448559 DOI: 10.3892/ol.2020.11976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/17/2020] [Indexed: 11/25/2022] Open
Abstract
Gastric cancer is a leading cause of cancer-associated deaths worldwide and is considered to be an age-related disease. In younger patients, gastric cancer is biologically more aggressive, and prognosis is worse compared with that in elderly patients. In the present case report, the whole genome and transcriptome was sequenced in a 26-year-old patient with gastric cancer who presented with gastric cancer-related symptoms and was admitted to the First Affiliated Anhui Medical Hospital (Hefei, China) in December 2016. In total, 9 germline and 4 somatic mutations were identified in the patient, and there were more deleterious sites in the germline mutated genes. Genes with somatic mutations, such as MUC2, MUC4, SLC8A2, and with structural variations, including CCND3, FGFR2 and FGFR3, were found to be differentially expressed. Cancer-associated pathways, such as the 'calcium signaling pathway', 'cGMP-PKG signaling pathway' and 'transcriptional mis-regulation' were also enriched at both the genomic and transcriptomic levels. The genes found to have germline (SFRP4), somatic (MUC2, MUC4, SLC8A2) mutations, or structural variations (CCND3, FGFR2 and FGFR3) were differentially expressed in the patient and could be promising precision therapy targets.
Collapse
Affiliation(s)
- Kongwang Hu
- Division of Gastrointestinal Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Weiqiang Yu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| | - Olugbenga Emmanuel Ajayi
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| | - Longlong Li
- Division of Gastrointestinal Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Zhiguo Huang
- Division of Gastrointestinal Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Qiqi Rong
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| | - Shuaili Wang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| | - Qing-Fa Wu
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| |
Collapse
|
18
|
Kuroda K, Yashiro M, Miki Y, Sera T, Yamamoto Y, Sugimoto A, Nishimura S, Kushiyama S, Togano S, Okuno T, Ohira M. Circulating tumor cells with FGFR2 expression might be useful to identify patients with existing FGFR2-overexpressing tumor. Cancer Sci 2020; 111:4500-4509. [PMID: 32946655 PMCID: PMC7734156 DOI: 10.1111/cas.14654] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
Fibroblast growth factor receptor (FGFR) is associated with proliferation, migration, and angiogenesis of carcinomas, and FGFR signaling inhibitors are considered a key drug for the treatment of solid tumors with FGFR overexpression. Amplification of FGFR2 is reportedly identified in 3%‐10% of gastric cancers (GCs). The aim of this study is to clarify whether the identification of the circulating tumor cells (CTCs) with FGFR2 overexpression is useful to detect patients with FGFR2‐overexpressing GC. One hundred GC patients who underwent gastrectomy were enrolled. A total volume of 8 mL of peripheral blood was collected from each patient just before gastrectomy, and mononuclear cells were enriched by Ficol density gradient centrifugation. These cells were immunostained with PI/CD45/EpCAM/FGFR2. The number of CTCs with FGFR2 expression in each sample was enumerated by FACScan. The FGFR2 expression level of the resected primary tumor was assessed by immunohistochemistry. The number of FGFR2‐positive CTCs in the GC patients' peripheral blood was significantly correlated with the FGFR2 expression level of the primary GC. The relapse‐free survival of the patients with FGFR2‐positive CTCs (≥5 cells/10 mL blood) was significantly poorer (P = .018, log‐rank) than that of the patients without FGFR2‐positive CTCs (<5 cell/10 mL blood). These findings suggested that the determination of FGFR2‐positive CTCs might help identify an existing tumor with FGFR2 overexpression.
Collapse
Affiliation(s)
- Kenji Kuroda
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuichiro Miki
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tomohiro Sera
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yurie Yamamoto
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Sugimoto
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Sadaaki Nishimura
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shuhei Kushiyama
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shingo Togano
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tomohisa Okuno
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
19
|
Takahashi K, Ishibashi E, Kubo T, Harada Y, Hayashi H, Kano M, Shimizu Y, Shirota H, Mori Y, Muto M, Ishioka C, Dosaka-Akita H, Matsubara H, Nishihara H, Sueoka-Aragane N, Toyooka S, Hirakawa A, Tateishi U, Miyake S, Ikeda S. A phase 2 basket trial of combination therapy with trastuzumab and pertuzumab in patients with solid cancers harboring human epidermal growth factor receptor 2 amplification (JUPITER trial). Medicine (Baltimore) 2020; 99:e21457. [PMID: 32769873 PMCID: PMC7592999 DOI: 10.1097/md.0000000000021457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Human epidermal growth factor receptor 2 (HER2) gene amplification and mutations have emerged as oncogenic drivers and therapeutic targets not limited to breast and gastric cancers, but also in a variety of cancers. However, even if an actionable gene alteration is found, the incidence of HER2 amplification in these cancers is less than 5%. It is too difficult to conduct a conventional randomized, controlled trial in a rare fraction. Therefore, we have designed a organ-agnostic basket study, which covers a variety of solid cancers harboring HER2 amplification, in 1 study protocol. METHODS/DESIGN This trial is a multicenter, single-arm, basket phase 2 study in Japan. Patients with solid cancers harboring HER2 amplification that have progressed with standard treatment, or rare cancers for which there is no standard treatment, will be eligible. Target cancers include bile duct, urothelial, uterine, ovarian, and other solid cancers where HER2 amplification is detected by comprehensive genomic profiling using next-generation sequencing technology. A total of 38 patients will be treated with combination therapy with trastuzumab and pertuzumab every 3 weeks until disease progression, unmanageable toxicity, death, or patient refusal. The primary endpoint is the objective response rate, and secondary endpoints are progression-free survival, overall survival, and duration of response. DISCUSSION The aim of this trial is to evaluate the safety and efficacy of combination therapy with trastuzumab and pertuzumab in patients with locally advanced or metastatic, solid cancers harboring HER2 amplification. Instead of focusing on 1 organ type, our trial design uses a basket study focusing on HER2 amplification, regardless of the site or origin of the cancer. The results of our study will advance clinical and scientific knowledge concerning the treatment of locally advanced, rare solid cancers harboring HER2 amplification, using the combination of trastuzumab and pertuzumab. TRIAL REGISTRATION This trial was registered in Japan Registry of Clinical Trials (jCRT) on February 25, 2019, as jRCT2031180150.
Collapse
Affiliation(s)
| | - Eri Ishibashi
- Medical Innovation Promotion Center, Tokyo Medical and Dental University, Tokyo
| | - Toshio Kubo
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama
| | - Yohei Harada
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga
| | - Hideyuki Hayashi
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo
| | - Masayuki Kano
- Department of Frontier Surgery, Chiba University, Chiba
| | - Yasushi Shimizu
- Department of Medical Oncology, Faculty of Medicine & Graduate School of Medicine, Hokkaido University, Hokkaido
| | - Hidekazu Shirota
- Department of Clinical Oncology, Tohoku University Hospital, Sendai
| | - Yukiko Mori
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto
| | - Chikashi Ishioka
- Department of Clinical Oncology, Tohoku University Hospital, Sendai
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Faculty of Medicine & Graduate School of Medicine, Hokkaido University, Hokkaido
| | | | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama
| | | | - Ukihide Tateishi
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Sadakatsu Ikeda
- Center for Innovative Cancer Treatment
- Moores Cancer Center, University of California, San Diego, CA
| |
Collapse
|
20
|
Hur JY, Chao J, Kim K, Kim ST, Kim KM, Klempner SJ, Lee J. High-level FGFR2 amplification is associated with poor prognosis and Lower response to chemotherapy in gastric cancers. Pathol Res Pract 2020; 216:152878. [PMID: 32089408 DOI: 10.1016/j.prp.2020.152878] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/22/2020] [Accepted: 02/11/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recurrent FGFR2 amplification is observed in gastroesophageal cancers but clinical implications are unknown. We investigated the association of FGFR2 amplification with cytotoxic chemotherapy outcome in gastroesophageal cancer (GC) patients. METHODS Between 2016 and 2018, we identified 1045 metastatic GC patients who received palliative fluoropyrimidine/platinum-based chemotherapy and underwent tumor genomic profiling at a tertiary hospital in Korea and two US cancer centers. We retrospectively identified FGFR2-amplified cases and abstracted clinicopathologic features and treatment outcomes. Cox proportional hazard regression model was used to evaluate the variables that demonstrated effects on progression free and overall survival PFS and OS. Descriptive statistics were used to correlate level of FGFR2 copy number amplification CNA and clinicopathological parameters. RESULTS The incidence of FGFR2-amplified GC was 4.0 %. A total of 42 FGFR2-amplified GC patients were included and divided into high and lower FGFR2 amplification values. Fifteen patients had an FGFR2 CNA greater than 30, and 27 had a CNA of 30 or less. There was no significant differences between age, sex, tumor localization, Lauren classification, or tumor staging. After a median follow-up duration of 11.4 months, patients with high FGFR2 amplification had significantly poorer median PFS (3.2 vs. 4.8 months, hazard ratio (HR) 2.08, 95 % CI, 1.03-4.22, P = 0.042) and significantly shorter median OS (10.1 vs. 26.3 months, HR 2.99, 95 % CI = 1.05-8.49, P = 0.040) than the low FGFR2 amplification group. CONCLUSION Recurrent FGFR2 amplification was observed in roughly 4.0 % of GC patients. High FGFR2 amplification was significantly associated with poor progression free survival and overall survival in GC patients. Clinical studies of FGFR2-directed therapies are warranted and should consider stratification by FGFR2 CNA.
Collapse
Affiliation(s)
- Joon Young Hur
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Division of Hematology and Oncology, Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea.
| | - Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Kyung Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Samuel J Klempner
- Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
21
|
Effects of Huaier Polysaccharide SP1 on Gastric Cancer Cell Proliferation, Apoptosis, Migration, and Invasion by Regulating TGF- β/SMAD Signaling Pathway. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/8486039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objective. To study the effects of Huaier polysaccharide SP1 on the proliferation, apoptosis, migration, and invasion of gastric cancer cell line MGC-803 and the underlying mechanism. Methods. MGC-803 cells were cultured in vitro and treated with SP1. The effects of SP1 on the proliferation, apoptosis, migration, and invasion of MGC-803 cells were detected by CCK-8 assay, flow cytometry analysis, and Transwell assay, respectively. Western blot and qRT-PCR were used to detect the expression of related genes. Results. Our study showed that Huaier polysaccharide SP1 could inhibit proliferation, migration, invasion, and promote the apoptosis of MGC-803 cells in vitro in a dose-dependent manner. Huaier polysaccharide SP1 could inhibit the activation of TGF-β/SMAD signal pathway by upregulating SMAD7 expression, thereby downregulating the expression of SOX4, ZEB2, MMP9, Snail, and Slug. Conclusion. Huaier polysaccharide SP1 can regulate the proliferation, apoptosis, migration, and invasion of gastric cancer cells by promoting the expression of SMAD7 and inhibiting the activation of TGF-β/SMAD signal pathway as well as the expression of the downstream oncogenes.
Collapse
|
22
|
Futami T, Kawase T, Mori K, Asaumi M, Kihara R, Shindoh N, Kuromitsu S. Identification of a novel oncogenic mutation of FGFR4 in gastric cancer. Sci Rep 2019; 9:14627. [PMID: 31601997 PMCID: PMC6787178 DOI: 10.1038/s41598-019-51217-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer remains one of the leading causes of cancer death worldwide. Despite intensive investigations of treatments over the past three decades, the poor prognosis of patients with unresectable advanced or recurrent gastric cancer has not significantly changed, and improved therapies are required. Here, we report the identification of an oncogenic mutation in FGFR4 in a human gastric tumour that leads to constitutive activation of its product, FGFR4. The G636C-FGFR4 tyrosine kinase domain mutation was found in 1 of 83 primary human gastric tumours. The G636C mutation increased FGFR4 autophosphorylation, and activated FGFR4 downstream signalling molecules and enhanced anchorage-independent cell growth when expressed in NIH/3T3 cells. 3D-structural analysis and modelling of FGFR4 suggest that G636C destabilizes an auto-inhibitory conformation and stabilizes an active conformation, leading to increased kinase activation. Ba/F3 cell lines expressing the G636C-FGFR4 mutant were significantly more sensitive to ASP5878, a selective FGFR inhibitor, than the control. Oral administration of ASP5878 significantly inhibited the growth of tumours in mice engrafted with G636C-FGFR4/3T3 cells. Together, our results demonstrate that mutationally activated FGFR4 acts as an oncoprotein. These findings support the therapeutic targeting of FGFR4 in gastric cancer.
Collapse
Affiliation(s)
- Takashi Futami
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan.
| | - Tatsuya Kawase
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| | - Kenichi Mori
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| | - Makoto Asaumi
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| | - Rumi Kihara
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| | - Nobuaki Shindoh
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| | - Sadao Kuromitsu
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| |
Collapse
|
23
|
Zhang J, Tang PMK, Zhou Y, Cheng ASL, Yu J, Kang W, To KF. Targeting the Oncogenic FGF-FGFR Axis in Gastric Carcinogenesis. Cells 2019; 8:cells8060637. [PMID: 31242658 PMCID: PMC6627225 DOI: 10.3390/cells8060637] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/14/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most wide-spread malignancies in the world. The oncogenic role of signaling of fibroblast growing factors (FGFs) and their receptors (FGFRs) in gastric tumorigenesis has been gradually elucidated by recent studies. The expression pattern and clinical correlations of FGF and FGFR family members have been comprehensively delineated. Among them, FGF18 and FGFR2 demonstrate the most prominent driving role in gastric tumorigenesis with gene amplification or somatic mutations and serve as prognostic biomarkers. FGF-FGFR promotes tumor progression by crosstalking with multiple oncogenic pathways and this provides a rational therapeutic strategy by co-targeting the crosstalks to achieve synergistic effects. In this review, we comprehensively summarize the pathogenic mechanisms of FGF-FGFR signaling in gastric adenocarcinoma together with the current targeted strategies in aberrant FGF-FGFR activated GC cases.
Collapse
Affiliation(s)
- Jinglin Zhang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| | - Patrick M K Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| | - Yuhang Zhou
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| | - Alfred S L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Jun Yu
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
24
|
Kim HS, Kim JH, Jang HJ. Pathologic and prognostic impacts of FGFR2 amplification in gastric cancer: a meta-analysis and systemic review. J Cancer 2019; 10:2560-2567. [PMID: 31258762 PMCID: PMC6584337 DOI: 10.7150/jca.29184] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 04/12/2019] [Indexed: 01/10/2023] Open
Abstract
Fibroblast growth factor receptor-2 (FGFR2) gene is amplified in up to 15% of patients with gastric cancer (GC). However, the prognostic significance of FGFR2 amplification has been controversial. This meta-analysis was conducted to evaluate the clinicopathological impacts of FGFR2 amplification in patients with GC. We performed a systematic computerized search of the electronic databases of PubMed, PMC, EMBASE, Web of Science, and Google Scholar and selected studies assessing the correlation of FGFR2 amplification with pathologic features and/or prognosis in gastric adenocarcinoma. From eight studies, 2,377 patients were included in the pooled analysis of odds ratios (ORs) with 95% confidence intervals (CIs) for pathologic findings and hazard ratios (HRs) with 95% CIs for overall survival. FGFR2 amplification was significantly associated with LN metastasis (OR = 3.93, 95% CI: 2.22-6.96, p < 0.00001) and poorly differentiated adenocarcinoma (OR = 2.36, 95% CI: 1.03-5.39, p = 0.04). In addition, patients with GC harboring FGFR2 amplification showed significantly worse survival (HR = 2.09, 95% CI: 1.68-2.59, p < 0.00001), compared with patients with FGFR2-unamplified GC. In conclusion, this meta-analysis indicates that FGFR2 amplification is an adverse prognostic factor in patients with GC.
Collapse
Affiliation(s)
- Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Hwasung, Republic of Korea
| |
Collapse
|
25
|
Kim HS, Kim JH, Jang HJ, Han B, Zang DY. Pathological and Prognostic Impacts of FGFR2 Overexpression in Gastric Cancer: A Meta-Analysis. J Cancer 2019; 10:20-27. [PMID: 30662521 PMCID: PMC6329854 DOI: 10.7150/jca.28204] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/15/2018] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor receptor-2 (FGFR2) protein expression by immunohistochemistry has been reported in up to 60% of patients with gastric cancer (GC). However, the clicopathological impacts of high FGFR2 expression have not been consistent among studies. We conducted this meta-analysis to evaluate the pathological and prognostic significance of FGFR2 overexpression in patients with GC. A systematic search of the electronic databases including PubMed, PMC, EMBASE, and Google Scholar was performed. From ten studies, 4,294 patients were included in the pooled analyses of odds ratios (ORs) with 95% confidence intervals (CIs) for pathological features and hazard ratios (HRs) with 95% CIs for overall survival according to the FGFR2 expression status. Compared with tumors showing low FGFR2 expression, GCs with FGFR2 overexpression revealed deeper depth of invasion (pT3-4) (OR = 2.63, 95% CI: 1.70-4.06, p < 0.0001), higher rate of lymph node metastasis (OR = 1.87, 95% CI: 1.31-2.67, p < 0.0001), and more advanced stage (III-IV) (OR = 1.78, 95% CI: 1.07-2.96, p = 0.03). In addition, patients with FGFR2-overexpressed GC showed significantly worse survival than those with FGFR2-low tumor (HR = 1.40, 95% CI: 1.25-1.58, p < 0.00001). In conclusion, this meta-analysis indicates that FGFR2 overexpression is associated with poor pathological features and prognosis in patients with GC.
Collapse
Affiliation(s)
- Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Hwasung, Republic of Korea
| | - Boram Han
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Dae Young Zang
- Division of Hemato-Oncology, Department of Internal Medicine, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
26
|
Tirino G, Pompella L, Petrillo A, Laterza MM, Pappalardo A, Caterino M, Orditura M, Ciardiello F, Galizia G, De Vita F. What's New in Gastric Cancer: The Therapeutic Implications of Molecular Classifications and Future Perspectives. Int J Mol Sci 2018; 19:E2659. [PMID: 30205505 PMCID: PMC6165492 DOI: 10.3390/ijms19092659] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 02/05/2023] Open
Abstract
Despite some remarkable innovations and the advent of novel molecular classifications the prognosis of patients with advanced gastric cancer (GC) remains overall poor and current clinical application of new advances is disappointing. During the last years only Trastuzumab and Ramucirumab have been approved and currently used as standard of care targeted therapies, but the systemic management of advanced disease did not radically change in contrast with the high number of molecular drivers identified. The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group (ACRG) classifications paved the way, also for GC, to that more contemporary therapeutic approach called "precision medicine" even if tumor heterogeneity and a complex genetic landscape still represent a strong barrier. The identification of specific cancer subgroups is also making possible a better selection of patients that are most likely to respond to immunotherapy. This review aims to critically overview the available molecular classifications summarizing the main druggable molecular drivers and their possible therapeutic implications also taking advantage of new technologies and acquisitions.
Collapse
Affiliation(s)
- Giuseppe Tirino
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Luca Pompella
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Angelica Petrillo
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Maria Maddalena Laterza
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Annalisa Pappalardo
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Marianna Caterino
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Michele Orditura
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Fortunato Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Gennaro Galizia
- Division of GI Tract Surgical Oncology, Department of Cardio-Thoracic and Respiratory Sciences, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini n.5, 80131 Naples, Italy.
| |
Collapse
|
27
|
Lee SY, Na YJ, Jeong YA, Kim JL, Oh SC, Lee DH. Upregulation of EphB3 in gastric cancer with acquired resistance to a FGFR inhibitor. Int J Biochem Cell Biol 2018; 102:128-137. [PMID: 30044964 DOI: 10.1016/j.biocel.2018.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/20/2018] [Accepted: 07/21/2018] [Indexed: 11/16/2022]
Abstract
Amplification of fibroblast growth factor receptor2 (FGFR2) has been regarded as a druggable target in gastric cancer (GC). Despite known potential of AZD4547, a selective inhibitor of FGFR 1-3, to suppress tumorigenic effects of activated FGFR2, resistance to the targeted agent has been an unresolved issue. This study was performed to elucidate the mechanism of AZD4547 resistance in GC cells. SNU-16 cells were used to establish an AZD4547-resistant GC cell line, SNU-16R. Elevated phosphorylation of EphB3 was confirmed using the Human Phospho-Receptor Tyrosine Kinase Array kit. A tyrosine kinase inhibitor (TKI) of EphB3 was used to investigate the effects of suppressed EphB3 activity in the SNU-16R cell line. SNU-16R cells exhibited upregulated phosphorylation of EphB3. Treatment of SNU-16R cells with the EphB3 TKI resulted in induction of apoptosis, decreased cellular viability, and cell cycle arrest at sub-G1 phase. SNU-16R cells expressed upregulated levels of N-cadherin, vimentin, Snail, matrix metalloproteinase 2 (MMP-2), and MMP-9, and reduced levels of E-cadherin, characteristic of epithelial to mesenchymal transition (EMT). Matrigel invasion assay also demonstrated the increased invasiveness of SNU-16R cells. EphB3 TKI treatment inhibited EMT of SNU-16R cells. Activation of mammalian target of rapamycin (mTOR) through the Ras-ERK1/2 pathway was suggested as the signal transduction mechanism downstream EphB3 by showing enhanced phosphorylation of Raf-1, MEK1/2, ERK1/2, mTOR and its downstream substrates in SNU-16R cells. As expected, EphB3 TKI decreased phosphorylation of these proteins. Our data suggest phosphorylation of mTOR through signaling by EphB3 is a potential mechanism of AZD4547 resistance in GC cells.
Collapse
Affiliation(s)
- Suk-Young Lee
- Division of Oncology/Hematology, Department of Internal Medicine, College of Medicine, Korea University, 148 Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea.
| | - Yoo Jin Na
- Division of Oncology/Hematology, Department of Internal Medicine, College of Medicine, Korea University, 148 Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea.
| | - Yoon A Jeong
- Division of Oncology/Hematology, Department of Internal Medicine, College of Medicine, Korea University, 148 Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea.
| | - Jung Lim Kim
- Division of Oncology/Hematology, Department of Internal Medicine, College of Medicine, Korea University, 148 Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea.
| | - Sang Cheul Oh
- Division of Oncology/Hematology, Department of Internal Medicine, College of Medicine, Korea University, 148 Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea.
| | - Dae-Hee Lee
- Division of Oncology/Hematology, Department of Internal Medicine, College of Medicine, Korea University, 148 Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea; Brain Korea 21 Program for Biomedicine Science, College of Medicine, Korea University, 148 Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea.
| |
Collapse
|
28
|
De Mello RA, Castelo-Branco L, Castelo-Branco P, Pozza DH, Vermeulen L, Palacio S, Salzberg M, Lockhart AC. What Will We Expect From Novel Therapies to Esophageal and Gastric Malignancies? Am Soc Clin Oncol Educ Book 2018; 38:249-261. [PMID: 30231398 DOI: 10.1200/edbk_198805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Esophageal cancer and gastric cancer are aggressive diseases for which treatment approaches are facing a new era. Some molecular pathways, such as VEGF, EGFR, fibroblast growth factor receptor, PIK3CA, and PARP-1, have been studied, and novel targeted drugs are presumed to be developed in the near future. From The Cancer Genome Atlas report, 80% of Epstein-Barr virus tumors and 42% of tumors with microsatellite instability have PIK3CA mutations, suggesting that this pathway could be reevaluated as a possible target for new systemic treatment of gastric cancer. Notably, higher PARP-1 expression can be found in gastric cancer, which might be related to more advanced disease and worse prognosis. In addition, PD-L1 expression, high microsatellite instability, and mismatch repair deficiency can be found in gastric cancer, thus suggesting that immunotherapy may also play a role in those patients. We discuss trends related to the potential of novel therapies for patients with esophageal and gastric cancers in the near future.
Collapse
Affiliation(s)
- Ramon Andrade De Mello
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Luis Castelo-Branco
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Pedro Castelo-Branco
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Daniel Humberto Pozza
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Louis Vermeulen
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Sofia Palacio
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Matthew Salzberg
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - A Craig Lockhart
- From the Department of Biomedical Sciences and Medicine, Division of Oncology, University of Algarve, Faro, Portugal; Algarve Biomedical Center, Campus Gambelas, Faro, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal; Research Centre, Division of Medical Oncology, Hospital São Mateus, NOHC Clinic, Fortaleza, CE, Brazil; Algarve Hospital and University Center, Department of Oncology, Faro, Portugal; Portuguese Public Health School, Nova University, Lisbon, Portugal; Centre for Biomedical Research, University of Algarve, Faro, Portugal; Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal; Academic Medical Center Amsterdam, Center for Experimental Molecular Medicine, Amsterdam, The Netherlands; and the Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| |
Collapse
|
29
|
Volpi CC, Gualeni AV, Pietrantonio F, Vaccher E, Carbone A, Gloghini A. Bright-field in situ hybridization detects gene alterations and viral infections useful for personalized management of cancer patients. Expert Rev Mol Diagn 2018; 18:259-277. [PMID: 29431533 DOI: 10.1080/14737159.2018.1440210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Bright-field in situ hybridization (ISH) methods detect gene alterations that may improve diagnostic precision and personalized management of cancer patients. Areas covered: This review focuses on some bright-field ISH techniques for detection of gene amplification or viral infection that have already been introduced in tumor pathology, research and diagnostic practice. Other emerging ISH methods, for the detection of translocation, mRNA and microRNA have recently been developed and need both an optimization and analytical validation. The review also deals with their clinical applications and implications on the management of cancer patients. Expert commentary: The technology of bright-field ISH applications has advanced significantly in the last decade. For example, an automated dual-color assay was developed as a clinical test for selecting cancer patients that are candidates for personalized therapy. Recently an emerging bright-field gene-protein assay has been developed. This method simultaneously detects the protein, gene and centromeric targets in the context of tissue morphology, and might be useful in assessing the HER2 status particularly in equivocal cases or samples with heterogeneous tumors. The application of bright-field ISH methods has become the gold standard for the detection of tumor-associated viral infection as diagnostic or prognostic factors.
Collapse
Affiliation(s)
- Chiara C Volpi
- a Department of Pathology and Laboratory Medicine , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| | - Ambra V Gualeni
- a Department of Pathology and Laboratory Medicine , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| | - Filippo Pietrantonio
- b Department of Medical Oncology , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| | - Emanuela Vaccher
- c Department of Medical Oncology , Centro di Riferimento Oncologico, IRCCS, National Cancer Institute , Aviano , Italy
| | - Antonino Carbone
- d Department of Pathology , Centro di Riferimento Oncologico, IRCCS, National Cancer Institute , Aviano , Italy
| | - Annunziata Gloghini
- a Department of Pathology and Laboratory Medicine , Fondazione IRCCS, Istituto Nazionale dei Tumori , Milano , Italy
| |
Collapse
|
30
|
Multicenter phase II study of apatinib treatment for metastatic gastric cancer after failure of second-line chemotherapy. Oncotarget 2017; 8:104552-104559. [PMID: 29262660 PMCID: PMC5732826 DOI: 10.18632/oncotarget.21053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/17/2017] [Indexed: 12/31/2022] Open
Abstract
Apatinib is a tyrosine kinase inhibitor and vascular endothelial growth factor receptor 2 (VEGFR-2) targeted drug. A phase I clinical trial showed that this agent has antitumor activity in Chinese patients with metastatic gastric cancer (mGC). The aim of this study was to investigate the safety and efficacy of apatinib treatment in patients with mGC. This was an open-label, multicenter, single-arm study involving four institutions in China. We enrolled 42 patients from March 2015 to October 2015 who experienced tumor progression after second-line chemotherapy and had no other treatment options that clearly conferred a survival benefit. Oral apatinib (850 mg daily) was administered within 30 min of eating breakfast, lunch, or dinner on days 1 through 28 of each 4-week cycle. The median progression-free survival (PFS) time and median overall survival (OS) time were 4.0 months (95% CI, 2.85-5.15) and 4.50 months (95% CI, 4.03-4.97), respectively. The disease control rate (DCR) and objective response rate (ORR) were, respectively, 78.57% and 9.52% after 2 cycles and 57.14% and 19.05% after 4 cycles. The main adverse events (AEs) were secondary hypertension, elevated aminotransferase, and hand-foot syndrome, with incidences of 35.71%, 45.24%, and 40.48%, respectively. The most common grade 3 to 4 AEs were secondary hypertension and elevated aminotransferase, with incidences of 7.14% each. Apatinib is effective and safe in heavily pretreated patients with mGC who fail to respond to two or more prior chemotherapy regimens. Toxicities were tolerable or could be clinically managed.
Collapse
|