251
|
Recent developments and advances of FGFR as a potential target in cancer. Future Med Chem 2018; 10:2109-2126. [DOI: 10.4155/fmc-2018-0103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
FGFs and their receptors (FGFRs) are critical for many biologic processes, including angiogenesis, wound healing and tissue regeneration. Aberrations in FGFR signaling are common in cancer, making FGFRs a promising target in antitumor studies. To date, many FGFR inhibitors are being detected in clinical studies, and resistance to some inhibitors has emerged. Understanding the mechanisms of resistance is a fundamental step for further implementation of targeted therapies. In this review, we will describe the basic knowledge regarding FGF/FGFR signaling and categorize the clinical FGFR inhibitors. The mechanisms of resistance to FGFR inhibitors and corresponding strategies of overcoming drug resistance will also be discussed.
Collapse
|
252
|
Current Status of Fibroblast Growth Factor Receptor-Targeted Therapies in Breast Cancer. Cells 2018; 7:cells7070076. [PMID: 30011957 PMCID: PMC6071019 DOI: 10.3390/cells7070076] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/30/2018] [Accepted: 07/11/2018] [Indexed: 01/08/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy and second only to lung cancer in terms of mortality in women. Despite the incredible progress made in this field, metastatic breast cancer has a poor prognosis. In an era of personalized medicine, there is an urgent need for better knowledge of the biology leading to the disease, which can lead to the design of increasingly accurate drugs against patients' specific molecular aberrations. Among one of the actionable targets is the fibroblast growth factor receptor (FGFR) pathway, triggered by specific ligands. The Fibroblast Growth Factor Receptors/Fibroblast Growth Factors (FGFRs/FGFs) axis offers interesting molecular targets to be pursued in clinical development. This mini-review will focus on the current knowledge of FGFR mutations, which lead to tumor formation and summarizes the state-of-the-art therapeutic strategies for targeted treatments against the FGFRs/FGFs axis in the context of BC.
Collapse
|
253
|
Ghedini GC, Ronca R, Presta M, Giacomini A. Future applications of FGF/FGFR inhibitors in cancer. Expert Rev Anticancer Ther 2018; 18:861-872. [PMID: 29936878 DOI: 10.1080/14737140.2018.1491795] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Deregulation of the fibroblast growth factor (FGF)/FGF receptor (FGFR) network occurs frequently in tumors due to gene amplification, activating mutations, and oncogenic fusions. Thus, the development of FGF/FGFR-targeting therapies is the focus of several basic, preclinical, and clinical studies. Areas covered: This review will recapitulate the status of current FGF/FGFR-targeted drugs. Expert commentary: Non-selective FGF/FGFR inhibitors have been approved for cancer treatment but evidence highlights various complications affecting their use in the clinical practice. It appears mandatory to identify FGF/FGFR alterations and appropriate biomarkers that may predict and monitor response to treatment, to establish the contribution of the FGF/FGFR system to the onset of mechanisms of drug resistance, and to develop effective combinations of FGF/FGFR inhibitors with other targeted therapies.
Collapse
Affiliation(s)
- Gaia Cristina Ghedini
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| | - Roberto Ronca
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| | - Marco Presta
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| | - Arianna Giacomini
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| |
Collapse
|
254
|
Chen L, Zhang YH, Zhang Z, Huang T, Cai YD. Inferring Novel Tumor Suppressor Genes with a Protein-Protein Interaction Network and Network Diffusion Algorithms. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 10:57-67. [PMID: 30069494 PMCID: PMC6068090 DOI: 10.1016/j.omtm.2018.06.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Extensive studies on tumor suppressor genes (TSGs) are helpful to understand the pathogenesis of cancer and design effective treatments. However, identifying TSGs using traditional experiments is quite difficult and time consuming. Developing computational methods to identify possible TSGs is an alternative way. In this study, we proposed two computational methods that integrated two network diffusion algorithms, including Laplacian heat diffusion (LHD) and random walk with restart (RWR), to search possible genes in the whole network. These two computational methods were LHD-based and RWR-based methods. To increase the reliability of the putative genes, three strict screening tests followed to filter genes obtained by these two algorithms. After comparing the putative genes obtained by the two methods, we designated twelve genes (e.g., MAP3K10, RND1, and OTX2) as common genes, 29 genes (e.g., RFC2 and GUCY2F) as genes that were identified only by the LHD-based method, and 128 genes (e.g., SNAI2 and FGF4) as genes that were inferred only by the RWR-based method. Some obtained genes can be confirmed as novel TSGs according to recent publications, suggesting the utility of our two proposed methods. In addition, the reported genes in this study were quite different from those reported in a previous one.
Collapse
Affiliation(s)
- Lei Chen
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People’s Republic of China
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
| | - Zhenghua Zhang
- Department of Clinical Oncology, Jing’an District Centre Hospital of Shanghai (Huashan Hospital Fudan University Jing’An Branch), Shanghai 200040, People’s Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
- Corresponding author: Tao Huang, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, People’s Republic of China
- Corresponding author: Yu-Dong Cai, School of Life Sciences, Shanghai University, Shanghai 200444, People’s Republic of China.
| |
Collapse
|
255
|
Li Q, Li Y, Liang L, Li J, Luo D, Liu Q, Cai S, Li X. Klotho negatively regulated aerobic glycolysis in colorectal cancer via ERK/HIF1α axis. Cell Commun Signal 2018; 16:26. [PMID: 29884183 PMCID: PMC5994118 DOI: 10.1186/s12964-018-0241-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023] Open
Abstract
Background Klotho (KL) was originally characterized as an aging suppressor gene, and has been identified as a tumor suppressor gene in a variety of cancers, including colorectal cancer. Recent years have witnessed the importance of metabolism transformation in cancer cell malignancies maintenance. Aberrant cancer cell metabolism is considered to be the hallmark of cancer. Our previous studies demonstrated that KL played negative roles in colon cancer cell proliferation and metastasis. However, its role in the cancer cell reprogramming has seldom been reported. The aim of this study was to examine the role of KL in aerobic glycolysis in colorectal cancer. Methods Combining maximum standardized uptake value (SUVmax), which was obtained preoperatively via a PET/CT scan, with immunohistochemistry staining, we analyzed the correlation between SUVmax and KL expression in colorectal cancer tissues. The impact of KL on glucose metabolism and its mechanisms were further validated in vitro and in vivo. Results Patients with lower KL expression exhibited higher 18F-FDG uptake (P < 0.05), indicating that KL might participate in aerobic glycolysis regulation. In vitro assay by using colon cancer cell lines further supported this observation. By overexpressing KL in HTC116 and SW480 cells, we observed that the glycolysis was inhibited and the mitochondrial respiration increased, indicating that KL was a negative regulator of aerobic glycolysis. To seek for the underlying mechanisms, we tried to dig out the relation between KL and HIF1α signaling pathway, and found that KL negatively regulated HIF1α protein level and transcriptional activity. Western blot analysis showed that KL overexpression negatively regulated ERK pathway, and KL regulated aerobic glycolysis in part through its regulation of ERK/ HIF1α axis. Conclusions Taken together, KL is a negative regulator of aerobic glycolysis and KL inhibited glucose metabolism transformation via the ERK/ HIF1α axis. Electronic supplementary material The online version of this article (10.1186/s12964-018-0241-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Yaqi Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Lei Liang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Jing Li
- Departments of CyberKnife, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Dakui Luo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Qi Liu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong'an Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
256
|
DeSmet M, Kanginakudru S, Jose L, Xie F, Gilson T, Androphy EJ. Papillomavirus E2 protein is regulated by specific fibroblast growth factor receptors. Virology 2018; 521:62-68. [PMID: 29885490 DOI: 10.1016/j.virol.2018.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 02/01/2023]
Abstract
The papillomavirus (PV) E2 protein activates transcription and replication by recruiting cellular proteins and the E1 DNA helicase to their binding sites in the viral genome. We recently demonstrated that phosphorylation of tyrosine 102 in the bovine papillomavirus (BPV-1) E2 protein restricts these activities and that fibroblast growth factor receptor-3 (FGFR3) tyrosine kinase binds PV E2. Expression of FGFR3 decreased viral replication with both wild-type and the phenylalanine substitution at position 102, inferring that another kinase targets Y102. Here we tested FGFR- 1, -2 and -4 for association with PV E2 proteins. FGFR2 but not FGFR1 or FGFR4 co-immunoprecipitated with BPV-1 E2. We found that FGFR2 suppressed replication but did not depend on phosphorylation of BPV-1 Y102. HPV-16 and -31 E2 interacted with FGFR1, -2, and -4. These results imply that the expression and activity of FGF receptors in epithelial cells can regulate the function of E2 in viral replication.
Collapse
Affiliation(s)
- Marsha DeSmet
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Leny Jose
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fang Xie
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Dermatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Timra Gilson
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elliot J Androphy
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
257
|
Kabashima A, Hirsova P, Bronk SF, Hernandez MC, Truty MJ, Ilyas SI, Kaufmann SH, Gores GJ. Fibroblast growth factor receptor inhibition induces loss of matrix MCL1 and necrosis in cholangiocarcinoma. J Hepatol 2018; 68:1228-1238. [PMID: 29408314 PMCID: PMC5960421 DOI: 10.1016/j.jhep.2018.01.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 01/18/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Myeloid cell leukemia 1 (MCL1), a prosurvival member of the BCL2 protein family, has a pivotal role in human cholangiocarcinoma (CCA) cell survival. We previously reported that fibroblast growth factor receptor (FGFR) signalling mediates MCL1-dependent survival of CCA cells in vitro and in vivo. However, the mode and mechanisms of cell death in this model were not delineated. METHODS Human CCA cell lines were treated with the pan-FGFR inhibitor LY2874455 and the mode of cell death examined by several complementary assays. Mitochondrial oxidative metabolism was examined using a XF24 extracellular flux analyser. The efficiency of FGFR inhibition in patient-derived xenografts (PDX) was also assessed. RESULTS CCA cells expressed two species of MCL1, a full-length form localised to the outer mitochondrial membrane, and an N terminus-truncated species compartmentalised within the mitochondrial matrix. The pan-FGFR inhibitor LY2874455 induced non-apoptotic cell death in the CCA cell lines associated with cellular depletion of both MCL1 species. The cell death was accompanied by failure of mitochondrial oxidative metabolism and was most consistent with necrosis. Enforced expression of N terminus-truncated MCL1 targeted to the mitochondrial matrix, but not full-length MCL1 targeted to the outer mitochondrial membrane, rescued cell death and mitochondrial function. LY2874455 treatment of PDX-bearing mice was associated with tumour cell loss of MCL1 and cell necrosis. CONCLUSIONS FGFR inhibition induces loss of matrix MCL1, resulting in cell necrosis. These observations support a heretofore unidentified, alternative MCL1 survival function, namely prevention of cell necrosis, and have implications for treatment of human CCA. LAY SUMMARY Herein, we report that therapeutic inhibition of a cell receptor expressed by bile duct cancer cells resulted in the loss of a critical survival protein termed MCL1. Cellular depletion of MCL1 resulted in the death of the cancer cells by a process characterised by cell rupture. Cell death by this process can stimulate the immune system and has implications for combination therapy using receptor inhibition with immunotherapy.
Collapse
Affiliation(s)
- Ayano Kabashima
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA; Institute of Clinical Biochemistry and Diagnostics, Charles University, Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic; Department of Pharmacology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Steven F Bronk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Mark J Truty
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
258
|
Design, synthesis and biological evaluation of a series of novel 2-benzamide-4-(6-oxy-N-methyl-1-naphthamide)-pyridine derivatives as potent fibroblast growth factor receptor (FGFR) inhibitors. Eur J Med Chem 2018; 154:9-28. [PMID: 29775937 DOI: 10.1016/j.ejmech.2018.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/08/2018] [Accepted: 05/04/2018] [Indexed: 02/08/2023]
Abstract
Starting from the phase II clinical FGFR inhibitor lucitanib (2), we conducted a medicinal chemistry approach by opening the central quinoline skeleton coupled with a scaffold hopping process thus leading to a series of novel 2-benzamide-4-(6-oxy-N-methyl-1-naphthamide)-pyridine derivatives. Compound 25a was identified to show selective and equally high potency against FGFR1/2 and VEGFR2 with IC50 values less than 5.0 nM. Significant antiproliferative effects on both FGFR1/2 and VEGFR2 aberrant cancer cells were observed. In the SNU-16 xenograft model, compound 25a showed tumor growth inhibition rates of 25.0% and 81.0% at doses of 10 mg/kg and 50 mg/kg, respectively, with 5% and 10%body weight loss. In view of the synergistic potential of FGFs and VEGFs in tumor angiogenesis observed in preclinical studies, the FGFR/VEGFR2 dual inhibitor 25a may achieve better clinical benefits.
Collapse
|
259
|
Kolenda T, Przybyła W, Kapałczyńska M, Teresiak A, Zajączkowska M, Bliźniak R, Lamperska KM. Tumor microenvironment - Unknown niche with powerful therapeutic potential. Rep Pract Oncol Radiother 2018; 23:143-153. [PMID: 29760589 PMCID: PMC5948324 DOI: 10.1016/j.rpor.2018.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 11/20/2017] [Accepted: 01/20/2018] [Indexed: 12/25/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are in a group of cancers that are the most resistant to treatment. The survival rate of HNSCC patients has been still very low since last 20 years. The existence of relationship between oncogenic and surrounding cells is probably the reason for a poor response to treatment. Fibroblasts are an important element of tumor stroma which increases tumor cells ability to proliferate. Another highly resistance, tumorigenic and metastatic cell population in tumor microenvironment are cancer initiating cells (CICs). The population of cancer initiating cells can be found regardless of differentiation status of cancer and they seem to be crucial for HNSCC development. In this review, we describe the current state of knowledge about HNSCC biological and physiological tumor microenvironment.
Collapse
Affiliation(s)
- Tomasz Kolenda
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Weronika Przybyła
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marta Kapałczyńska
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Gastroenterology and Hepatology, Charite University Medicine Berlin, Berlin, Germany
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anna Teresiak
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
| | - Maria Zajączkowska
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Renata Bliźniak
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
| | | |
Collapse
|
260
|
Xu JM, Wang Y, Chen YL, Jia R, Li J, Gong JF, Li J, Qi C, Hua Y, Tan CR, Wang J, Li K, Sai Y, Zhou F, Ren YX, Qing WG, Jia H, Su WG, Shen L. Sulfatinib, a novel kinase inhibitor, in patients with advanced solid tumors: results from a phase I study. Oncotarget 2018; 8:42076-42086. [PMID: 28159938 PMCID: PMC5522050 DOI: 10.18632/oncotarget.14942] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/05/2016] [Indexed: 12/17/2022] Open
Abstract
Sulfatinib is a small molecule kinase inhibitor that targets tumor angiogenesis and immune modulation. This phase I study (NCT02133157) investigated the safety, pharmacokinetic characteristics, and preliminary anti-tumor activity of sulfatinib in patients with advanced solid tumors. The study included a dose-escalation phase (50-350 mg/day, 28-day cycle) with a Fibonacci (3+3) design, and a tumor-specific expansion phase investigating the tumor response to treatment. Two sulfatinib formulations were assessed: formulation 1 (5, 25, and 50 mg capsules) and formulation 2 (50 and 200 mg capsules). Seventy-seven Chinese patients received oral sulfatinib; the maximum tolerated dose was not reached. Dose-limiting toxicities included abnormal hepatic function and coagulation tests, and upper gastrointestinal hemorrhage. The most common treatment-related adverse events were proteinuria, hypertension and diarrhea. Among 34 patients receiving sulfatinib formulation 2, one patient with hepatocellular carcinoma and eight with neuroendocrine tumors exhibited a partial response; 15 had stable disease. The objective response rate was 26.5% (9/34) and the disease control rate was 70.6% (24/34). Pharmacokinetic, safety, and efficacy data supported continuous oral administration of sulfatinib at 300 mg as the recommended phase II dose. Sulfatinib exhibited an acceptable safety profile and encouraging antitumor activity in patients with advanced solid tumors, particularly neuroendocrine tumors.
Collapse
Affiliation(s)
- Jian Ming Xu
- Department of Gastrointestinal Oncology, The Affiliated Hospital Cancer Center (The 307th Hospital of Chinese People's Liberation Army), Academy of Military Medical Sciences, Beijing, China
| | - Yan Wang
- Department of Gastrointestinal Oncology, The Affiliated Hospital Cancer Center (The 307th Hospital of Chinese People's Liberation Army), Academy of Military Medical Sciences, Beijing, China
| | - Yu Ling Chen
- Department of Gastrointestinal Oncology, The Affiliated Hospital Cancer Center (The 307th Hospital of Chinese People's Liberation Army), Academy of Military Medical Sciences, Beijing, China
| | - Ru Jia
- Department of Gastrointestinal Oncology, The Affiliated Hospital Cancer Center (The 307th Hospital of Chinese People's Liberation Army), Academy of Military Medical Sciences, Beijing, China
| | - Jie Li
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| | - Ji Fang Gong
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| | - Jing Li
- Clinical and Regulatory Department, Hutchison MediPharma Limited, Shanghai, China
| | - Chuan Qi
- Clinical and Regulatory Department, Hutchison MediPharma Limited, Shanghai, China
| | - Ye Hua
- Clinical and Regulatory Department, Hutchison MediPharma Limited, Shanghai, China
| | - Cui Rong Tan
- Clinical and Regulatory Department, Hutchison MediPharma Limited, Shanghai, China
| | - Jian Wang
- Drug Metabolism and Pharmacokinetic Department, Hutchison MediPharma Limited, Shanghai, China
| | - Ke Li
- Drug Metabolism and Pharmacokinetic Department, Hutchison MediPharma Limited, Shanghai, China
| | - Yang Sai
- Drug Metabolism and Pharmacokinetic Department, Hutchison MediPharma Limited, Shanghai, China
| | - Feng Zhou
- Oncology Department, Hutchison MediPharma Limited, Shanghai, China
| | - Yong Xin Ren
- Oncology Department, Hutchison MediPharma Limited, Shanghai, China
| | - Wei Guo Qing
- Oncology Department, Hutchison MediPharma Limited, Shanghai, China
| | - Hong Jia
- Chemistry Department, Hutchison MediPharma Limited, Shanghai, China
| | - Wei Guo Su
- Chemistry Department, Hutchison MediPharma Limited, Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| |
Collapse
|
261
|
Computational Simulation Studies on the Binding Selectivity of 1-(1H-Benzimidazol-5-yl)-5-aminopyrazoles in Complexes with FGFR1 and FGFR4. Molecules 2018; 23:molecules23040767. [PMID: 29584670 PMCID: PMC6017917 DOI: 10.3390/molecules23040767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/12/2018] [Accepted: 03/20/2018] [Indexed: 11/16/2022] Open
Abstract
Fibroblast growth factor receptor 1 (FGFR1) has become a potential target for the treatment of cancer. Designing FGFR1-selective inhibitors remains fundamental to the development of anti-cancer drugs because of highly sequential homology among FGFR subtypes. In present work, four inhibitors were examined with intermolecular interaction patterns with FGFR1 and FGFR4, respectively, for the exploration of binding mechanisms by applying a combined approach of computational techniques, including flexible docking, binding site analyses, electronic structure computations, molecular dynamic simulations, and binding free energy predictions. Molecular simulation-predicted binding conformations and pharmacophoric features of these molecules in the active pocket of either FGFR1 or FGFR4. MMPB(GB)SA-calculated binding free energies were accordant with the ordering of their tested potency values. Furthermore, in silico mutations of two residues (FGFR1: Tyr563 and Ser565) were also performed to check their impact on ligand binding by applying MD simulations and binding free energy calculations. The present studies may provide a structural understanding of the FGFR1-selective mechanism. The viewpoints from computational simulations would be valuable guidelines for the development of novel FGFR1-selective inhibitors.
Collapse
|
262
|
Bartels S, Adisa A, Aladelusi T, Lemound J, Stucki-Koch A, Hussein S, Kreipe H, Hartmann C, Lehmann U, Hussein K. Molecular defects in BRAF wild-type ameloblastomas and craniopharyngiomas-differences in mutation profiles in epithelial-derived oropharyngeal neoplasms. Virchows Arch 2018; 472:1055-1059. [PMID: 29546640 DOI: 10.1007/s00428-018-2323-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/26/2018] [Accepted: 02/13/2018] [Indexed: 12/18/2022]
Abstract
The aim of this study was to evaluate the mutation profile of BRAF wild-type craniopharyngiomas and ameloblastomas. Pre-screening by immunohistochemistry and pyrosequencing for identifying BRAF wild-type tumors was performed on archived specimens of ameloblastic tumors (n = 20) and craniopharyngiomas (n = 62). Subsequently, 19 BRAF wild-type tumors (nine ameloblastic tumors and ten craniopharyngiomas) were analyzed further using next-generation sequencing (NGS) targeting hot spot mutations of 22 cancer-related genes. Thereby, we found craniopharyngiomas mainly CTNNB1 mutated (8/10), including two FGFR3/CTNNB1-double mutated tumors. Ameloblastic tumors were often FGFR2 mutated (4/9; including one FGFR2/TP53/PTEN-triple mutated case) and rarely CTNNB1/TP53-double mutated (1/9) and KRAS-mutated (1/9). In the remaining samples, no mutation could be detected in the 22 genes under investigation. In conclusion, mutation profiles of BRAF wild-type craniopharyngiomas and ameloblastomas share mutations of FGFR genes and have additional mutations with potential for targeted therapy.
Collapse
Affiliation(s)
- Stephan Bartels
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Akinyele Adisa
- Oral Pathology Department University College Hospital Ibadan, University of Ibadan, Ibadan, Nigeria
| | - Timothy Aladelusi
- Oral and Maxillofacial Surgery Department University College Hospital Ibadan, University of Ibadan, Ibadan, Nigeria
| | - Juliana Lemound
- Department of Cranio-Maxillofacial Surgery, Hannover Medical School, Hannover, Germany
| | - Angelika Stucki-Koch
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Sami Hussein
- Department of Neurosurgery, Al-Makassed-Hospital, Al-Quds School of Medicine, Jerusalem, Israel
| | - Hans Kreipe
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Christian Hartmann
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Department of Neuropathology, Hannover Medical School, Hannover, Germany
| | - Ulrich Lehmann
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Kais Hussein
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
263
|
Liu H, Murphy CJ, Karreth FA, Emdal KB, White FM, Elemento O, Toker A, Wulf GM, Cantley LC. Identifying and Targeting Sporadic Oncogenic Genetic Aberrations in Mouse Models of Triple-Negative Breast Cancer. Cancer Discov 2018; 8:354-369. [PMID: 29203461 PMCID: PMC5907916 DOI: 10.1158/2159-8290.cd-17-0679] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/11/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancers (TNBC) are genetically characterized by aberrations in TP53 and a low rate of activating point mutations in common oncogenes, rendering it challenging in applying targeted therapies. We performed whole-exome sequencing (WES) and RNA sequencing (RNA-seq) to identify somatic genetic alterations in mouse models of TNBCs driven by loss of Trp53 alone or in combination with Brca1 Amplifications or translocations that resulted in elevated oncoprotein expression or oncoprotein-containing fusions, respectively, as well as frameshift mutations of tumor suppressors were identified in approximately 50% of the tumors evaluated. Although the spectrum of sporadic genetic alterations was diverse, the majority had in common the ability to activate the MAPK/PI3K pathways. Importantly, we demonstrated that approved or experimental drugs efficiently induce tumor regression specifically in tumors harboring somatic aberrations of the drug target. Our study suggests that the combination of WES and RNA-seq on human TNBC will lead to the identification of actionable therapeutic targets for precision medicine-guided TNBC treatment.Significance: Using combined WES and RNA-seq analyses, we identified sporadic oncogenic events in TNBC mouse models that share the capacity to activate the MAPK and/or PI3K pathways. Our data support a treatment tailored to the genetics of individual tumors that parallels the approaches being investigated in the ongoing NCI-MATCH, My Pathway Trial, and ESMART clinical trials. Cancer Discov; 8(3); 354-69. ©2017 AACR.See related commentary by Natrajan et al., p. 272See related article by Matissek et al., p. 336This article is highlighted in the In This Issue feature, p. 253.
Collapse
Affiliation(s)
- Hui Liu
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Charles J Murphy
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Florian A Karreth
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kristina B Emdal
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Forest M White
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Alex Toker
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, and Ludwig Center at Harvard, Boston, Massachusetts
| | - Gerburg M Wulf
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
264
|
Li YX, Yu ZW, Jiang T, Shao LW, Liu Y, Li N, Wu YF, Zheng C, Wu XY, Zhang M, Zheng DF, Qi XL, Ding M, Zhang J, Chang Q. SNCA, a novel biomarker for Group 4 medulloblastomas, can inhibit tumor invasion and induce apoptosis. Cancer Sci 2018; 109:1263-1275. [PMID: 29369502 PMCID: PMC5891175 DOI: 10.1111/cas.13515] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/24/2022] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in childhood. It contains at least four distinct molecular subgroups. The aim of this study is to explore novel diagnostic and potential therapeutic markers within each subgroup of MB, in particular within Group 4, the largest subgroup, to facilitate diagnosis together with gene therapy. One hundred and six MB samples were examined. Tumor subtype was evaluated with the NanoString assay. Several novel tumor related genes were shown to have high subgroup sensitivity and specificity, including PDGFRA,FGFR1, and ALK in the WNT group, CCND1 in the SHH group, and α‐synuclein (SNCA) in Group 4. Knockdown and overexpression assays of SNCA revealed the ability of this gene to inhibit tumor invasion and induce apoptosis. Methylation‐specific PCR and pyrosequencing analysis showed that epigenetic mechanisms, rather than DNA hypermethylation, might play the key role in the regulation of SNCA expression in MB tumors. In conclusion, we identify SNCA as a novel diagnostic biomarker for Group 4 MB. Some other subgroup signature genes have also been found as candidate therapeutic targets for this tumor.
Collapse
Affiliation(s)
- Yong-Xiao Li
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Zhen-Wei Yu
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China
| | - Li-Wei Shao
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Yan Liu
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Na Li
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Yu-Feng Wu
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Chen Zheng
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Xiao-Yu Wu
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Ming Zhang
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Dan-Feng Zheng
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| | - Xue-Ling Qi
- Department of Pathology, Beijing Sanbo Brain Hospital, Beijing, China
| | - Min Ding
- Department of Pathology, Anhui Provincial Hospital, Hefei, China
| | - Jing Zhang
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Qing Chang
- Department of Pathology, Peking University School of Basic Medical Science, Peking University Third Hospital, Peking Univeristy Health Science Center, Beijing, China
| |
Collapse
|
265
|
Fonseca FP, Monteiro Benites B, Soares CD, de Lima Morais TM, do Amaral-Silva GK, de Almeida OP, Soares FA, Fregnani ER. Prognostic importance of FGF2 and FGFR1 expression for patients affected by ameloblastoma. J Oral Pathol Med 2018; 47:417-424. [PMID: 29453876 DOI: 10.1111/jop.12695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Fibroblast growth factor 2 (FGF2) and FGF receptor 1 (FGFR1) have been investigated in different human neoplasms and were shown to play important roles in the pathogenesis of these diseases; however, very few are known regarding their prognostic importance in the context of ameloblastoma. Therefore, the aim of this study was to investigate whether the expression of FGF2 and FGFR1 is associated with ameloblastoma clinical behavior. METHODS Fifty-eight cases of ameloblastoma arranged in tissue microarray were submitted to immunohistochemistry against FGF2 and FGFR1. Clinicopathological parameters regarding sex, age, tumor size, duration and location, treatment, recurrences, radiographic features, cortical disruptions, and follow-up data were obtained from patients' medical records and correlated with the molecules expression. Univariate and multivariate Cox regression analyses were used to investigate the prognostic potential of the biomarkers. RESULTS Forty-four cases (75.9%) exhibited cytoplasmic positivity for FGF2 in central and peripheral epithelial cells, 46 of 58 (79.3%) showed FGFR1 cytoplasmic positivity predominantly in the columnar peripheral cells, and 43 cases (74.1%) were positive for both. Expression of FGF2 and FGF2 + FGFR1 was associated with tumor recurrences (P = .05). However, univariate and multivariate analyses did not demonstrate a significant influence of FGF2, FGFR1, or FGF2 + FGFR1 in the 5-year disease-free survival (DFS) rate (P = .27, P = .33, and P = .25, respectively). CONCLUSION Cytoplasmic expression of FGF2 and FGF2 + FGFR1 is associated with ameloblastoma recurrence, but FGF2 and FGFR1 are not determinants of a lower DFS.
Collapse
Affiliation(s)
- Felipe Paiva Fonseca
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ciro Dantas Soares
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas Piracicaba, Piracicaba, Brazil
| | - Thayná Melo de Lima Morais
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas Piracicaba, Piracicaba, Brazil
| | | | - Oslei Paes de Almeida
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas Piracicaba, Piracicaba, Brazil
| | | | | |
Collapse
|
266
|
Cooper SL, Sandhu H, Hussain A, Mee C, Maddock H. Involvement of mitogen activated kinase kinase 7 intracellular signalling pathway in Sunitinib-induced cardiotoxicity. Toxicology 2018; 394:72-83. [DOI: 10.1016/j.tox.2017.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 12/30/2022]
|
267
|
Hu P, Chen H, McGowan EM, Ren N, Xu M, Lin Y. Assessment of FGFR1 Over-Expression and Over-Activity in Lung Cancer Cells: A Toolkit for Anti-FGFR1 Drug Screening. Hum Gene Ther Methods 2018; 29:30-43. [PMID: 29281903 DOI: 10.1089/hgtb.2017.104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Penghui Hu
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hongjie Chen
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Eileen M McGowan
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Nina Ren
- Guangdong Online Hospital Clinic, Guangdong 2nd Provincial People's Hospital, Guangzhou, China
| | - Meng Xu
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
268
|
Fibroblast growth factor receptor 1 and 3 expression is associated with regulatory PI3K/AKT kinase activity, as well as invasion and prognosis, in human laryngeal cancer. Cell Oncol (Dordr) 2018; 41:253-268. [DOI: 10.1007/s13402-017-0367-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2017] [Indexed: 12/11/2022] Open
|
269
|
Farran B, Müller S, Montenegro RC. Gastric cancer management: Kinases as a target therapy. Clin Exp Pharmacol Physiol 2018; 44:613-622. [PMID: 28271563 DOI: 10.1111/1440-1681.12743] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/19/2017] [Accepted: 02/20/2017] [Indexed: 12/16/2022]
Abstract
The molecular diagnostics revolution has reshaped the practice of oncology by facilitating the identification of genetic, epigenetic and proteomic modifications correlated with cancer, thus delineating 'oncomaps' for various cancer types. These advances have enhanced our understanding of gastric cancer, one of the most fatal diseases worldwide, and culminated in the approval of novel molecular therapies such as trastuzumab. Gastric tumours display recurrent aberrations in key kinase oncogenes such as Her2, epidermal growth factor receptor (EGFR), PI3K, mTOR or c-Met, suggesting that these receptors are amenable to inhibition using specific drug agents. In this review, we examine the mutational landscape of gastric cancer, the use of kinase inhibitors as targeted therapies in gastric tumours and the clinical trials underway for novel inhibitors, highlighting successes, failures and future directions.
Collapse
Affiliation(s)
- Batoul Farran
- Department of Structural and Molecular Biology, University College London, London, UK
| | - Susanne Müller
- Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Frankfurt am Main, DE, Germany
| | - Raquel C Montenegro
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
270
|
Hilberg F, Tontsch-Grunt U, Baum A, Le AT, Doebele RC, Lieb S, Gianni D, Voss T, Garin-Chesa P, Haslinger C, Kraut N. Triple Angiokinase Inhibitor Nintedanib Directly Inhibits Tumor Cell Growth and Induces Tumor Shrinkage via Blocking Oncogenic Receptor Tyrosine Kinases. J Pharmacol Exp Ther 2017; 364:494-503. [PMID: 29263244 DOI: 10.1124/jpet.117.244129] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 12/11/2017] [Indexed: 12/11/2022] Open
Abstract
The triple-angiokinase inhibitor nintedanib is an orally available, potent, and selective inhibitor of tumor angiogenesis by blocking the tyrosine kinase activities of vascular endothelial growth factor receptor (VEGFR) 1-3, platelet-derived growth factor receptor (PDGFR)-α and -β, and fibroblast growth factor receptor (FGFR) 1-3. Nintedanib has received regulatory approval as second-line treatment of adenocarcinoma non-small cell lung cancer (NSCLC), in combination with docetaxel. In addition, nintedanib has been approved for the treatment of idiopathic lung fibrosis. Here we report the results from a broad kinase screen that identified additional kinases as targets for nintedanib in the low nanomolar range. Several of these kinases are known to be mutated or overexpressed and are involved in tumor development (discoidin domain receptor family, member 1 and 2, tropomyosin receptor kinase A (TRKA) and C, rearranged during transfection proto-oncogene [RET proto oncogene]), as well as in fibrotic diseases (e.g., DDRs). In tumor cell lines displaying molecular alterations in potential nintedanib targets, the inhibitor demonstrates direct antiproliferative effects: in the NSCLC cell line NCI-H1703 carrying a PDGFRα amplification (ampl.); the gastric cancer cell line KatoIII and the breast cancer cell line MFM223, both driven by a FGFR2 amplification; AN3CA (endometrial carcinoma) bearing a mutated FGFR2; the acute myeloid leukemia cell lines MOLM-13 and MV-4-11-B with FLT3 mutations; and the NSCLC adenocarcinoma LC-2/ad harboring a CCDC6-RET fusion. Potent kinase inhibition does not, however, strictly translate into antiproliferative activity, as demonstrated in the TRKA-dependent cell lines CUTO-3 and KM-12. Importantly, nintedanib treatment of NCI-H1703 tumor xenografts triggered effective tumor shrinkage, indicating a direct effect on the tumor cells in addition to the antiangiogenic effect on the tumor stroma. These findings will be instructive in guiding future genome-based clinical trials of nintedanib.
Collapse
Affiliation(s)
- Frank Hilberg
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Ulrike Tontsch-Grunt
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Anke Baum
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Anh T Le
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Robert C Doebele
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Simone Lieb
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Davide Gianni
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Tilman Voss
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Pilar Garin-Chesa
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Christian Haslinger
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH Co KG, Vienna, Austria (F.H., U.T.-G., A.B., S.L., D.G., T.V., P.G.-C., C.H., N.K.); University of Colorado, School of Medicine, Division of Medical Oncology, Aurora, Colorado (A.T.L., R.C.D.); and AstraZeneca - Innovative Medicines and Early Development, Discovery Sciences, Cambridge Science Park, Milton, Cambridge (D.G.)
| |
Collapse
|
271
|
Katoh M. Antibody-drug conjugate targeting protein tyrosine kinase 7, a receptor tyrosine kinase-like molecule involved in WNT and vascular endothelial growth factor signaling: effects on cancer stem cells, tumor microenvironment and whole-body homeostasis. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:462. [PMID: 29285495 DOI: 10.21037/atm.2017.09.11] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo, Japan
| |
Collapse
|
272
|
Strati P, Tang G, Duose DY, Mallampati S, Luthra R, Patel KP, Hussaini M, Mirza AS, Komrokji RS, Oh S, Mascarenhas J, Najfeld V, Subbiah V, Kantarjian H, Garcia-Manero G, Verstovsek S, Daver N. Myeloid/lymphoid neoplasms with FGFR1 rearrangement. Leuk Lymphoma 2017; 59:1672-1676. [PMID: 29119847 DOI: 10.1080/10428194.2017.1397663] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Myeloid/lymphoid neoplasms with FGFR1 rearrangement are a rare entity. We present a multicenter experience of 17 patients with FISH-confirmed FGFR1 rearrangement. The clinical presentation at diagnosis included myeloproliferative neoplasm (MPN) in 4 (24%) patients, acute leukemia (AL) in 7 (41%), and concomitant MPN with AL in 6 (35%). The two most frequently observed cytogenetic abnormalities were t(8;13)(p11.2;q12)(partner gene ZMYM2) and t(8;22)(p11.2; q11.2)(BCR). Seventy-eight percent of tested patients had a RUNX1 mutation, of whom all had AL. Overall response rate to frontline therapy was 69%, and 76% of patients subsequently received allogeneic stem cell transplant (ASCT). After a median follow-up of 11 months, median progression-free survival was 15 months and median overall survival was not reached. In conclusion, FGFR1-rearranged hematologic malignancies present with features of MPN and/or AL. FGFR1 and RUNX1 are therapeutic targets for ongoing and future clinical trials.
Collapse
Affiliation(s)
- Paolo Strati
- a Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Guilin Tang
- b Department of Hematopathology The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Dzifa Y Duose
- c Department of Malignant Hematology , H Lee Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| | - Saradhi Mallampati
- b Department of Hematopathology The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Rajyalakshmi Luthra
- b Department of Hematopathology The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Keyur P Patel
- b Department of Hematopathology The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Mohammad Hussaini
- c Department of Malignant Hematology , H Lee Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| | - Abu-Sayeef Mirza
- c Department of Malignant Hematology , H Lee Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| | - Rami S Komrokji
- c Department of Malignant Hematology , H Lee Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| | - Stephen Oh
- d Department of Hematology , Washington University School of Medicine , St Louis , MO , USA
| | - John Mascarenhas
- e Department of Hematology/Oncology , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Vesna Najfeld
- f Department of Pathology Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Vivek Subbiah
- g Department of Investigational Therapeutics , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Hagop Kantarjian
- a Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Guillermo Garcia-Manero
- a Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Srdan Verstovsek
- a Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Naval Daver
- a Department of Leukemia , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
273
|
Collateral Damage Intended-Cancer-Associated Fibroblasts and Vasculature Are Potential Targets in Cancer Therapy. Int J Mol Sci 2017; 18:ijms18112355. [PMID: 29112161 PMCID: PMC5713324 DOI: 10.3390/ijms18112355] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/25/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023] Open
Abstract
After oncogenic transformation, tumor cells rewire their metabolism to obtain sufficient energy and biochemical building blocks for cell proliferation, even under hypoxic conditions. Glucose and glutamine become their major limiting nutritional demands. Instead of being autonomous, tumor cells change their immediate environment not only by their metabolites but also by mediators, such as juxtacrine cell contacts, chemokines and other cytokines. Thus, the tumor cells shape their microenvironment as well as induce resident cells, such as fibroblasts and endothelial cells (ECs), to support them. Fibroblasts differentiate into cancer-associated fibroblasts (CAFs), which produce a qualitatively and quantitatively different extracellular matrix (ECM). By their contractile power, they exert tensile forces onto this ECM, leading to increased intratumoral pressure. Moreover, along with enhanced cross-linkage of the ECM components, CAFs thus stiffen the ECM. Attracted by tumor cell- and CAF-secreted vascular endothelial growth factor (VEGF), ECs sprout from pre-existing blood vessels during tumor-induced angiogenesis. Tumor vessels are distinct from EC-lined vessels, because tumor cells integrate into the endothelium or even mimic and replace it in vasculogenic mimicry (VM) vessels. Not only the VM vessels but also the characteristically malformed EC-lined tumor vessels are typical for tumor tissue and may represent promising targets in cancer therapy.
Collapse
|
274
|
The integration of genomics testing and functional proteomics in the era of personalized medicine. Expert Rev Proteomics 2017; 14:1055-1058. [PMID: 29039218 DOI: 10.1080/14789450.2017.1392245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
275
|
Shivapurkar N, Vietsch EE, Carney E, Isaacs C, Wellstein A. Circulating microRNAs in patients with hormone receptor-positive, metastatic breast cancer treated with dovitinib. Clin Transl Med 2017; 6:37. [PMID: 28980224 PMCID: PMC5628092 DOI: 10.1186/s40169-017-0169-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Serial analysis of biomarkers in the circulation of patients undergoing treatment ("liquid biopsies") can provide new insights into drug effects. In particular the analysis of cell-free, circulating nucleic acids such as microRNAs (miRs) can reveal altered expression patterns indicative of mechanism of drug action, cancer growth, and tumor-stroma interactions. RESULTS Here we analyzed plasma miRs in patients with hormone receptor positive, metastatic breast cancer with prior disease progression during aromatase inhibitor therapy (n = 8) in a phase I/II trial with the multiple tyrosine kinase inhibitor dovitinib (TKI258). Plasma miR levels were measured by quantitative RT-qPCR before and after treatment with dovitinib. A candidate miR signature of drug response was established from a 379 miR screen for detectable plasma miRs as well as from the published literature. Changes in miR expression patterns and tumor sizes were compared. In this analysis we identified miR-21-5p, miR-100-5p, miR-125b-5p, miR-126-3p, miR-375 and miR-424-5p as potential indicators of a response to dovitinib. The altered expression patterns observed for the six circulating miRs separated patients with resistant disease from those with drug responsive disease. There was no relationship between adverse effects of dovitinib treatment and identifiable changes in miR patterns. CONCLUSION We conclude that changes in the expression patterns of circulating miRs can be indicators of drug responses that merit prospective studies for validation.
Collapse
Affiliation(s)
- Narayan Shivapurkar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Eveline E Vietsch
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Erin Carney
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road NW, Washington, DC, 20057, USA
| | - Claudine Isaacs
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road NW, Washington, DC, 20057, USA.
| | - Anton Wellstein
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road NW, Washington, DC, 20057, USA.
| |
Collapse
|
276
|
Papadopoulos KP, El-Rayes BF, Tolcher AW, Patnaik A, Rasco DW, Harvey RD, LoRusso PM, Sachdev JC, Abbadessa G, Savage RE, Hall T, Schwartz B, Wang Y, Kazakin J, Shaib WL. A Phase 1 study of ARQ 087, an oral pan-FGFR inhibitor in patients with advanced solid tumours. Br J Cancer 2017; 117:1592-1599. [PMID: 28972963 PMCID: PMC5729432 DOI: 10.1038/bjc.2017.330] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 12/15/2022] Open
Abstract
Background: ARQ 087 is an orally administered pan-FGFR inhibitor with multi-kinase activity. This Phase 1 study evaluated safety, pharmacokinetics, and pharmacodynamics of ARQ 087 and defined the recommended Phase 2 dose (RP2D). Methods: Patients with advanced solid tumours received ARQ 087 administered initially at 25 mg every other day and dose-escalated from 25 to 425 mg daily (QD) continuous dosing. FGF19, 21, 23, and serum phosphate were assessed as potential biomarkers of target engagement. Results: 80 patients were enrolled, 61 in dose-escalation/food-effect cohorts and 19 with pre-defined tumour types in the expansion cohort. The most common ARQ 087-related adverse events were fatigue (49%), nausea (46%), aspartate aminotransferase (AST) increase (30%), and diarrhoea (23%). Four patients (5%) experienced grade 1 treatment-related hyperphosphataemia. Dose-limiting toxicity was reversible grade 3 AST increase. The RP2D was 300 mg QD. Pharmacokinetics were linear and dose-proportional from 25 to 325 mg QD, and were unaffected by food. Statistically significant changes (P-value<0.05) suggest phosphate and FGF19 levels as markers of target engagement. In 18 evaluable patients with FGFR genetic alterations, 3 confirmed partial responses (two intrahepatic cholangiocarcinomas (iCCA) with FGFR2 fusions and one urothelial cancer with FGFR2 and FGF19 amplification) and two durable stable disease at ⩾16 weeks with tumour reduction (FGFR2 fusion-positive iCCA and adrenocortical carcinoma with FGFR1 amplification) were observed. Conclusions: ARQ 087 had manageable toxicity at the RP2D of 300 mg QD, showed pharmacodynamics effects, and achieved objective responses, notably in patients with FGFR2 genetic alterations.
Collapse
Affiliation(s)
- K P Papadopoulos
- South Texas Accelerated Research Therapeutics, 4383 Medical Drive, San Antonio, TX 78229, USA
| | - B F El-Rayes
- Winship Cancer Institute of Emory University, 1365-C Clifton Road NE, Atlanta, GA 30322, USA
| | - A W Tolcher
- South Texas Accelerated Research Therapeutics, 4383 Medical Drive, San Antonio, TX 78229, USA
| | - A Patnaik
- South Texas Accelerated Research Therapeutics, 4383 Medical Drive, San Antonio, TX 78229, USA
| | - D W Rasco
- South Texas Accelerated Research Therapeutics, 4383 Medical Drive, San Antonio, TX 78229, USA
| | - R D Harvey
- Winship Cancer Institute of Emory University, 1365-C Clifton Road NE, Atlanta, GA 30322, USA
| | - P M LoRusso
- Karmanos Cancer Institute, 4206-4th Floor HWCRC, 4100 John R, Detroit, MI 48201, USA
| | - J C Sachdev
- Virginia G. Piper Cancer Center, Scottsdale Healthcare, 10460N 92nd Street, Scottsdale, AZ 85258, USA
| | - G Abbadessa
- ArQule, Inc. One Wall Street, Burlington, MA 01803, USA
| | - R E Savage
- ArQule, Inc. One Wall Street, Burlington, MA 01803, USA
| | - T Hall
- ArQule, Inc. One Wall Street, Burlington, MA 01803, USA
| | - B Schwartz
- ArQule, Inc. One Wall Street, Burlington, MA 01803, USA
| | - Y Wang
- ArQule, Inc. One Wall Street, Burlington, MA 01803, USA
| | - J Kazakin
- ArQule, Inc. One Wall Street, Burlington, MA 01803, USA
| | - W L Shaib
- Winship Cancer Institute of Emory University, 1365-C Clifton Road NE, Atlanta, GA 30322, USA
| |
Collapse
|
277
|
Bavadi M, Niknam K, Shahraki O. Novel pyrrole derivatives bearing sulfonamide groups: Synthesis in vitro cytotoxicity evaluation, molecular docking and DFT study. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
278
|
The Evolving Role of Companion Diagnostics for Breast Cancer in an Era of Next-Generation Omics. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2185-2198. [DOI: 10.1016/j.ajpath.2017.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 02/06/2023]
|
279
|
Nishina T, Takahashi S, Iwasawa R, Noguchi H, Aoki M, Doi T. Safety, pharmacokinetic, and pharmacodynamics of erdafitinib, a pan-fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitor, in patients with advanced or refractory solid tumors. Invest New Drugs 2017; 36:424-434. [PMID: 28965185 DOI: 10.1007/s10637-017-0514-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/20/2017] [Indexed: 11/30/2022]
Abstract
Introduction This phase 1, open-label, multicenter, single-arm, dose-escalation study aimed to evaluate safety, pharmacokinetics (PK), and pharmacodynamics of erdafitinib (JNJ-42756493), an oral selective pan-fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitor, and to determine the recommended phase 2 dose in Japanese patients with advanced or refractory solid tumors. Methods Three to 6 patients were enrolled into sequentially escalating dose cohorts (erdafitinib 2, 4, or 6 mg) with a daily dosing schedule of 21-day cycles or a 7 days-on/7 days-off intermittent schedule (erdafitinib 10 mg or 12 mg) of 28-day cycles. Results Nineteen patients received escalating doses of erdafitinib with a daily or intermittent schedule. The most common treatment-emergent adverse events (TEAEs) were hyperphosphatemia (73.7%), nausea (36.8%), stomatitis (26.3%), dysgeusia (26.3%) and dry mouth (21.1%). The maximum tolerated dose was not reached in this study. No Grade 3 or higher TEAEs, or serious TEAEs were noted and no clinically significant changes in vital signs, laboratory parameters, and electrocardiogram readings were observed. However, one case of dose-limiting toxicity in the 12 mg intermittent dosing group was observed: Grade 2 detachment of retinal pigment epithelium (bilateral) with treatment discontinuation. The maximum plasma concentrations of erdafitinib exhibited a dose-dependent increase. The median tmax ranged from 2 to 3 h after the initial dose to 2-6 h following multiple daily dosing. Based on the safety and PK data, the 10 mg 7 days-on/7 days-off regimen was determined as the recommended phase 2 dose in this study. Conclusions Erdafitinib was well tolerated in Japanese patients with advanced or refractory solid tumors. TRIAL REGISTRATION NCT01962532.
Collapse
Affiliation(s)
- Tomohiro Nishina
- National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Shunji Takahashi
- The Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | | | | | | | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
280
|
Jafari B, Hamzeh-Mivehroud M, Alizadeh AA, Sharifi M, Dastmalchi S. An Alignment-Independent 3D-QSAR Study of FGFR2 Tyrosine Kinase Inhibitors. Adv Pharm Bull 2017; 7:409-418. [PMID: 29071223 PMCID: PMC5651062 DOI: 10.15171/apb.2017.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/08/2017] [Accepted: 08/12/2017] [Indexed: 01/09/2023] Open
Abstract
Purpose: Receptor tyrosine kinase (RTK) inhibitors are widely used pharmaceuticals in cancer therapy. Fibroblast growth factor receptors (FGFRs) are members of RTK superfamily which are highly expressed on the surface of carcinoma associate fibroblasts (CAFs). The involvement of FGFRs in different types of cancer makes them promising target in cancer therapy and hence, the identification of novel FGFR inhibitors is of great interest. In the current study we aimed to develop an alignment independent three dimensional quantitative structure-activity relationship (3D-QSAR) model for a set of 26 FGFR2 kinase inhibitors allowing the prediction of activity and identification of important structural features for these inhibitors. Methods: Pentacle software was used to calculate grid independent descriptors (GRIND) for the active conformers generated by docking followed by the selection of significant variables using fractional factorial design (FFD). The partial least squares (PLS) model generated based on the remaining descriptors was assessed by internal and external validation methods. Results: Six variables were identified as the most important probes-interacting descriptors with high impact on the biological activity of the compounds. Internal and external validations were lead to good statistical parameters (r2 values of 0.93 and 0.665, respectively). Conclusion: The results showed that the model has good predictive power and may be used for designing novel FGFR2 inhibitors.
Collapse
Affiliation(s)
- Behzad Jafari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Alizadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sharifi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
281
|
Katoh M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int J Oncol 2017; 51:1357-1369. [PMID: 29048660 PMCID: PMC5642388 DOI: 10.3892/ijo.2017.4129] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 09/15/2017] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs), which have the potential for self-renewal, differentiation and de-differentiation, undergo epigenetic, epithelial-mesenchymal, immunological and metabolic reprogramming to adapt to the tumor microenvironment and survive host defense or therapeutic insults. Intra-tumor heterogeneity and cancer-cell plasticity give rise to therapeutic resistance and recurrence through clonal replacement and reactivation of dormant CSCs, respectively. WNT signaling cascades cross-talk with the FGF, Notch, Hedgehog and TGFβ/BMP signaling cascades and regulate expression of functional CSC markers, such as CD44, CD133 (PROM1), EPCAM and LGR5 (GPR49). Aberrant canonical and non-canonical WNT signaling in human malignancies, including breast, colorectal, gastric, lung, ovary, pancreatic, prostate and uterine cancers, leukemia and melanoma, are involved in CSC survival, bulk-tumor expansion and invasion/metastasis. WNT signaling-targeted therapeutics, such as anti-FZD1/2/5/7/8 monoclonal antibody (mAb) (vantictumab), anti-LGR5 antibody-drug conjugate (ADC) (mAb-mc-vc-PAB-MMAE), anti-PTK7 ADC (PF-06647020), anti-ROR1 mAb (cirmtuzumab), anti-RSPO3 mAb (rosmantuzumab), small-molecule porcupine inhibitors (ETC-159, WNT-C59 and WNT974), tankyrase inhibitors (AZ1366, G007-LK, NVP-TNKS656 and XAV939) and β-catenin inhibitors (BC2059, CWP232228, ICG-001 and PRI-724), are in clinical trials or preclinical studies for the treatment of patients with WNT-driven cancers. WNT signaling-targeted therapeutics are applicable for combination therapy with BCR-ABL, EGFR, FLT3, KIT or RET inhibitors to treat a subset of tyrosine kinase-driven cancers because WNT and tyrosine kinase signaling cascades converge to β-catenin for the maintenance and expansion of CSCs. WNT signaling-targeted therapeutics might also be applicable for combination therapy with immune checkpoint blockers, such as atezolizumab, avelumab, durvalumab, ipilimumab, nivolumab and pembrolizumab, to treat cancers with immune evasion, although the context-dependent effects of WNT signaling on immunity should be carefully assessed. Omics monitoring, such as genome sequencing and transcriptome tests, immunohistochemical analyses on PD-L1 (CD274), PD-1 (PDCD1), ROR1 and nuclear β-catenin and organoid-based drug screening, is necessary to determine the appropriate WNT signaling-targeted therapeutics for cancer patients.
Collapse
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo 104-0045, Japan
| |
Collapse
|
282
|
Yan WF, Nie CF, Wu G, Zhang JC, Zhu YZ, Zhang W, Sun PC. Soluble interleukin‑2 receptor as a factor associated with angiogenesis in gastric cancer. Mol Med Rep 2017; 16:6916-6919. [PMID: 28901504 DOI: 10.3892/mmr.2017.7434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 05/30/2017] [Indexed: 11/05/2022] Open
Abstract
Angiogenesis serves a role in the growth, metastasis and prognosis of tumors. The aim of the present study was to evaluate the angiogenic ability and clinical significance of the immune biomarker soluble interleukin‑2 receptor (sIL‑2R) in gastric cancer (GC) patients. Serum levels of sIL‑2R were measured in 35 GC patients with different stages of disease and 32 healthy individuals, and it was investigated whether the levels were associated with angiogenesis factors, including vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)‑β1. Human umbilical vein endothelial cells (HUVECs) were pretreated with or without recombinant human (rh)sIL‑2R, VEGF and TGF‑β1 for 24 h, and then the HUVECSs were harvested to determine the degree of angiogenesis. The supernatants were also collected for VEGF and TGF‑β1 testing. Serum levels of sIL‑2R were higher in GC patients than in healthy individuals, as were the levels of VEGF and TGF‑β1. In addition, serum levels of sIL‑2R were positively associated with the levels of VEGF and TGF‑β1. Angiogenesis of HUVECs was also increased by rhsIL‑2R pretreatment. VEGF and TGF‑β1 secretion were also incre-ased in supernatants that were pretreated with rhsIL‑2R. The results of the present study suggested that serum levels of sIL‑2R contributes to the pathophysiology of GC progression and may be used as a prognostic biomarker for GC.
Collapse
Affiliation(s)
- Wen-Feng Yan
- Department of General Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Chang-Fu Nie
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Zhengzhou University, Division of Hepatobiliary and Pancreatic Surgery, Henan Tumor Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Gang Wu
- Department of General Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Jian-Cheng Zhang
- Department of General Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Yuan-Zeng Zhu
- Department of General Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Wei Zhang
- Department of General Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Pei-Chun Sun
- Department of General Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
283
|
Englinger B, Kallus S, Senkiv J, Heilos D, Gabler L, van Schoonhoven S, Terenzi A, Moser P, Pirker C, Timelthaler G, Jäger W, Kowol CR, Heffeter P, Grusch M, Berger W. Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer. J Exp Clin Cancer Res 2017; 36:122. [PMID: 28882160 PMCID: PMC5590147 DOI: 10.1186/s13046-017-0592-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Studying the intracellular distribution of pharmacological agents, including anticancer compounds, is of central importance in biomedical research. It constitutes a prerequisite for a better understanding of the molecular mechanisms underlying drug action and resistance development. Hyperactivated fibroblast growth factor receptors (FGFRs) constitute a promising therapy target in several types of malignancies including lung cancer. The clinically approved small-molecule FGFR inhibitor nintedanib exerts strong cytotoxicity in FGFR-driven lung cancer cells. However, subcellular pharmacokinetics of this compound and its impact on therapeutic efficacy remain obscure. METHODS 3-dimensional fluorescence spectroscopy was conducted to asses cell-free nintedanib fluorescence properties. MTT assay was used to determine the impact of the lysosome-targeting agents bafilomycin A1 and chloroquine combined with nintedanib on lung cancer cell viability. Flow cytometry and live cell as well as confocal microscopy were performed to analyze uptake kinetics as well as subcellular distribution of nintedanib. Western blot was conducted to investigate protein expression. Cryosections of subcutaneous tumor allografts were generated to detect intratumoral nintedanib in mice after oral drug administration. RESULTS Here, we report for the first time drug-intrinsic fluorescence properties of nintedanib in living and fixed cancer cells as well as in cryosections derived from allograft tumors of orally treated mice. Using this feature in conjunction with flow cytometry and confocal microscopy allowed to determine cellular drug accumulation levels, impact of the ABCB1 efflux pump and to uncover nintedanib trapping into lysosomes. Lysosomal sequestration - resulting in an organelle-specific and pH-dependent nintedanib fluorescence - was identified as an intrinsic resistance mechanism in FGFR-driven lung cancer cells. Accordingly, combination of nintedanib with agents compromising lysosomal acidification (bafilomycin A1, chloroquine) exerted distinctly synergistic growth inhibitory effects. CONCLUSION Our findings provide a powerful tool to dissect molecular factors impacting organismal and intracellular pharmacokinetics of nintedanib. Regarding clinical application, prevention of lysosomal trapping via lysosome-alkalization might represent a promising strategy to circumvent cancer cell-intrinsic nintedanib resistance.
Collapse
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Sebastian Kallus
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Julia Senkiv
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Institute of Cell Biology NAS of Ukraine, Drahomanova str 14/16, 79005 Lviv, Ukraine
| | - Daniela Heilos
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Department of Pharmacology and Toxicology, University of Vienna, Althanstr. 14, 1090 Vienna, Austria
| | - Lisa Gabler
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Sushilla van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Alessio Terenzi
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090 Vienna, Austria
| | - Patrick Moser
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Gerald Timelthaler
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Walter Jäger
- Department of Pharmaceutical Chemistry, Division of Clinical Pharmacy and Diagnostics, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| |
Collapse
|
284
|
Katoh M, Katoh M. Molecular genetics and targeted therapy of WNT-related human diseases (Review). Int J Mol Med 2017; 40:587-606. [PMID: 28731148 PMCID: PMC5547940 DOI: 10.3892/ijmm.2017.3071] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/12/2017] [Indexed: 12/15/2022] Open
Abstract
Canonical WNT signaling through Frizzled and LRP5/6 receptors is transduced to the WNT/β-catenin and WNT/stabilization of proteins (STOP) signaling cascades to regulate cell fate and proliferation, whereas non-canonical WNT signaling through Frizzled or ROR receptors is transduced to the WNT/planar cell polarity (PCP), WNT/G protein-coupled receptor (GPCR) and WNT/receptor tyrosine kinase (RTK) signaling cascades to regulate cytoskeletal dynamics and directional cell movement. WNT/β-catenin signaling cascade crosstalks with RTK/SRK and GPCR-cAMP-PKA signaling cascades to regulate β-catenin phosphorylation and β-catenin-dependent transcription. Germline mutations in WNT signaling molecules cause hereditary colorectal cancer, bone diseases, exudative vitreoretinopathy, intellectual disability syndrome and PCP-related diseases. APC or CTNNB1 mutations in colorectal, endometrial and prostate cancers activate the WNT/β-catenin signaling cascade. RNF43, ZNRF3, RSPO2 or RSPO3 alterations in breast, colorectal, gastric, pancreatic and other cancers activate the WNT/β-catenin, WNT/STOP and other WNT signaling cascades. ROR1 upregulation in B-cell leukemia and solid tumors and ROR2 upregulation in melanoma induce invasion, metastasis and therapeutic resistance through Rho-ROCK, Rac-JNK, PI3K-AKT and YAP signaling activation. WNT signaling in cancer, stromal and immune cells dynamically orchestrate immune evasion and antitumor immunity in a cell context-dependent manner. Porcupine (PORCN), RSPO3, WNT2B, FZD5, FZD10, ROR1, tankyrase and β-catenin are targets of anti-WNT signaling therapy, and ETC-159, LGK974, OMP-18R5 (vantictumab), OMP-54F28 (ipafricept), OMP-131R10 (rosmantuzumab), PRI-724 and UC-961 (cirmtuzumab) are in clinical trials for cancer patients. Different classes of anti-WNT signaling therapeutics are necessary for the treatment of APC/CTNNB1-, RNF43/ZNRF3/RSPO2/RSPO3- and ROR1-types of human cancers. By contrast, Dickkopf-related protein 1 (DKK1), SOST and glycogen synthase kinase 3β (GSK3β) are targets of pro-WNT signaling therapy, and anti-DKK1 (BHQ880 and DKN-01) and anti-SOST (blosozumab, BPS804 and romosozumab) monoclonal antibodies are being tested in clinical trials for cancer patients and osteoporotic post-menopausal women. WNT-targeting therapeutics have also been applied as reagents for in vitro stem-cell processing in the field of regenerative medicine.
Collapse
Affiliation(s)
| | - Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo 104-0045, Japan
| |
Collapse
|
285
|
FGFR2-Driven Signaling Counteracts Tamoxifen Effect on ERα-Positive Breast Cancer Cells. Neoplasia 2017; 19:791-804. [PMID: 28869838 PMCID: PMC5964976 DOI: 10.1016/j.neo.2017.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/02/2017] [Accepted: 07/12/2017] [Indexed: 02/07/2023] Open
Abstract
Signaling mediated by growth factors receptors has long been suggested as one of the key factors responsible for failure of endocrine treatment in breast cancer (BCa). Herein we present that in the presence of tamoxifen, FGFs (Fibroblast Growth Factors) promote BCa cell growth with the strongest effect being produced by FGF7. FGFR2 was identified as a mediator of FGF7 action and the FGFR2-induced signaling was found to underlie cancer-associated fibroblasts-dependent resistance to tamoxifen. FGF7/FGFR2-triggered pathway was shown to induce ER phosphorylation, ubiquitination and subsequent ER proteasomal degradation which counteracted tamoxifen-promoted ER stabilization. We also identified activation of PI3K/AKT signaling targeting ER-Ser167 and regulation of Bcl-2 expression as a mediator of FGFR2-promoted resistance to tamoxifen. Analysis of tissue samples from patients with invasive ductal carcinoma revealed an inversed correlation between expression of FGFR2 and ER, thus supporting our in vitro data. These results unveil the complexity of ER regulation by FGFR2-mediated signaling likely to be associated with BCa resistance to endocrine therapy.
Collapse
|
286
|
Todorović-Raković N, Radulovic M, Vujasinović T, Rabi ZA, Milovanović J, Nikolić-Vukosavljević D. bFGF in tumor tissue independently prognosticates disease outcome of a natural course of invasive breast cancer. Cancer Biomark 2017; 20:151-158. [DOI: 10.3233/cbm-170022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
287
|
Cui J, Peng X, Gao D, Dai Y, Ai J, Li Y. Optimization of 1H-indazol-3-amine derivatives as potent fibroblast growth factor receptor inhibitors. Bioorg Med Chem Lett 2017; 27:3782-3786. [DOI: 10.1016/j.bmcl.2017.06.068] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/31/2017] [Accepted: 06/26/2017] [Indexed: 11/29/2022]
|
288
|
A Phase 1 Study of LY2874455, an Oral Selective pan-FGFR Inhibitor, in Patients with Advanced Cancer. Target Oncol 2017; 12:463-474. [DOI: 10.1007/s11523-017-0502-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
289
|
Larsen K, Momeni J, Farajzadeh L, Callesen H. Splice variants of porcine PPHLN1 encoding periphilin-1. GENE REPORTS 2017. [DOI: 10.1016/j.genrep.2017.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
290
|
Toledo RA. Genetics of Pheochromocytomas and Paragangliomas: An Overview on the Recently Implicated Genes MERTK, MET, Fibroblast Growth Factor Receptor 1, and H3F3A. Endocrinol Metab Clin North Am 2017; 46:459-489. [PMID: 28476232 DOI: 10.1016/j.ecl.2017.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genomic studies conducted by different centers have uncovered various new genes mutated in pheochromocytomas and paragangliomas (PPGLs) at germline, mosaic, and/or somatic levels, greatly expanding our knowledge of the genetic events occurring in these tumors. The current review focuses on very new findings and discusses the previously not recognized role of MERTK, MET, fibroblast growth factor receptor 1, and H3F3A genes in syndromic and nonsyndromic PPGLs. These 4 new genes were selected because although their association with PPGLs is very recent, mounting evidence was generated that rapidly consolidated the prominence of these genes in the molecular pathogenesis of PPGLs.
Collapse
Affiliation(s)
- Rodrigo Almeida Toledo
- Division of Hematology and Medical Oncology, Department of Medicine, Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Dr, San Antonio, TX 78229, USA; Clinical Research Program, Spanish National Cancer Research Centre, CNIO, Calle de Melchor Fernández Almagro, 3, Madrid 28029, Spain.
| |
Collapse
|
291
|
Deregulation of kinase signaling and lymphoid development in EBF1-PDGFRB ALL leukemogenesis. Leukemia 2017; 32:38-48. [PMID: 28555080 PMCID: PMC5709252 DOI: 10.1038/leu.2017.166] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/10/2017] [Accepted: 05/17/2017] [Indexed: 01/06/2023]
Abstract
The chimeric fusion oncogene early B-cell factor 1-platelet-derived growth factor receptor-β (EBF1-PDGFRB) is a recurrent lesion observed in Philadelphia-like B-acute lymphoblastic leukemia (B-ALL) and is associated with particularly poor prognosis. While it is understood that this fusion activates tyrosine kinase signaling, the mechanisms of transformation and importance of perturbation of EBF1 activity remain unknown. EBF1 is a nuclear transcription factor required for normal B-lineage specification, commitment and development. Conversely, PDGFRB is a receptor tyrosine kinase that is normally repressed in lymphocytes, yet PDGFRB remains a common fusion partner in leukemias. Here, we demonstrate that the EBF1-PDGFRB fusion results in loss of EBF1 function, multimerization and autophosphorylation of the fusion protein, activation of signal transducer and activator of transcription 5 (STAT5) signaling and gain of interleukin-7 (IL-7)-independent cell proliferation. Deregulation and loss of EBF1 function is critically dependent on the nuclear export activity of the transmembrane (TM) domain of PDGFRB. Deletion of the TM domain partially rescues EBF1 function and restores IL-7 dependence, without requiring kinase inhibition. Moreover, we demonstrate that EBF1-PDGFRB synergizes with loss of IKAROS function in a fully penetrant B-ALL in vivo. Thus, we establish that EBF1-PDGFRB is sufficient to drive leukemogenesis through TM-dependent loss of transcription factor function, increased proliferation and synergy with additional genetic insults including loss of IKAROS function.
Collapse
|
292
|
Discovery and optimization of selective FGFR4 inhibitors via scaffold hopping. Bioorg Med Chem Lett 2017; 27:2420-2423. [PMID: 28433531 DOI: 10.1016/j.bmcl.2017.04.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/16/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022]
Abstract
Introduction of a Michael acceptor on a flexible scaffold derived from pan-FGFR inhibitors has successfully yielded a novel series of highly potent FGFR4 inhibitors with selectivity over FGFR1. Due to reduced lipophilicity and aromatic ring count, this series demonstrated improved solubility and permeability. However, plasma instability and fast metabolism limited its potential for in vivo studies. Efforts have been made to address these problems, which led to the discovery of compound (-)-11 with improved stability, CYP inhibition, and good activity/selectivity for further optimization.
Collapse
|
293
|
Perera TP, Jovcheva E, Mevellec L, Vialard J, De Lange D, Verhulst T, Paulussen C, Van De Ven K, King P, Freyne E, Rees DC, Squires M, Saxty G, Page M, Murray CW, Gilissen R, Ward G, Thompson NT, Newell DR, Cheng N, Xie L, Yang J, Platero SJ, Karkera JD, Moy C, Angibaud P, Laquerre S, Lorenzi MV. Discovery and Pharmacological Characterization of JNJ-42756493 (Erdafitinib), a Functionally Selective Small-Molecule FGFR Family Inhibitor. Mol Cancer Ther 2017; 16:1010-1020. [DOI: 10.1158/1535-7163.mct-16-0589] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/28/2016] [Accepted: 03/15/2017] [Indexed: 11/16/2022]
|
294
|
Analyses of publicly available genomics resources define FGF-2-expressing bladder carcinomas as EMT-prone, proliferative tumors with low mutation rates and high expression of CTLA-4, PD-1 and PD-L1. Signal Transduct Target Ther 2017; 2. [PMID: 28515962 PMCID: PMC5431749 DOI: 10.1038/sigtrans.2016.45] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factor 2 (FGF-2) is overexpressed in a subset of invasive bladder carcinomas and its overexpression correlates with poor prognosis. Analyses of publicly available databases addressing the molecular mechanisms that may be responsible for the poor prognosis of these tumors, revealed that FGF-2 expression correlates positively with the expression of epithelial to mesenchymal transition (EMT)-promoting transcription factors and with changes in gene expression that are characteristic of EMT. The same analyses also revealed that FGF-2 correlates negatively with the expression, mutation and copy number variations of FGFR-3, all of which are associated with noninvasive bladder carcinomas. Finally, they showed that FGF-2 expression correlates with the expression of FGFR-1, the expression of the IIIc variant of FGFR-2 and with the expression of Akt3. The latter observation is significant because our earlier studies had shown that Akt3 regulates FGFR-2 alternative splicing, shifting the balance toward the IIIc relative to the IIIb FGFR-2 splice variant. As the IIIc variant is recognized by FGF-2, while the IIIb variant is not, we conclude that Akt3 may facilitate the FGF-2 response. FGF-2 is known to promote the expression of KDM2B, which functions in concert with EZH2 to repress the EZH2-targeting microRNA miR-101, activating a switch, which stably upregulates EZH2. The cancer genome atlas (TCGA) data showing a correlation between KDM2B and EZH2 expression and Oncomine data, showing a correlation between KDM2B and tumor progression, strongly support the role of the FGF-2/KDM2B/miR-101/EZH2 pathway in bladder cancer. These observations combined, suggest a model according to which FGF-2 induces EMT, cell proliferation and cancer stem cell self-renewal by coupling the Akt3 and KDM2B-controlled pathways outlined above, in bladder carcinomas. Further analyses of publicly available databases, revealed that FGF-2-expressing bladder carcinomas carry fewer genetic alterations and they tend to express high levels of CTLA-4, PD-1 and PD-L1, which suggests immune blockade by checkpoint activation. EMT, enhanced proliferation and immune checkpoint activation combined, may be responsible for the poor prognosis of FGF-2-expressing bladder carcinomas.
Collapse
|
295
|
Cha HJ, Choi JH, Park IC, Kim CH, An SK, Kim TJ, Lee JH. Selective FGFR inhibitor BGJ398 inhibits phosphorylation of AKT and STAT3 and induces cytotoxicity in sphere-cultured ovarian cancer cells. Int J Oncol 2017; 50:1279-1288. [DOI: 10.3892/ijo.2017.3913] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 03/03/2017] [Indexed: 11/06/2022] Open
|
296
|
Glaser AP, Fantini D, Shilatifard A, Schaeffer EM, Meeks JJ. The evolving genomic landscape of urothelial carcinoma. Nat Rev Urol 2017; 14:215-229. [PMID: 28169993 DOI: 10.1038/nrurol.2017.11] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Survival of patients with urothelial carcinoma (including bladder cancer and upper tract urothelial carcinoma) is limited by our current approaches to staging, surgery, and chemotherapy. Large-scale, next-generation sequencing collaborations, such as The Cancer Genome Atlas, have already identified drivers and vulnerabilities of urothelial carcinoma. This disease has a high degree of mutational heterogeneity and a high frequency of somatic mutations compared with other solid tumours, potentially resulting in an increased neoantigen burden. Mutational heterogeneity is mediated by multiple factors including the apolipoprotein B mRNA editing enzyme catalytic polypeptide family of enzymes, smoking exposure, viral integrations, and intragene and intergene fusion proteins. The mutational landscape of urothelial carcinoma, including specific mutations in pathways and driver genes, such as FGFR3, ERBB2, PIK3CA, TP53, and STAG2, affects tumour aggressiveness and response to therapy. The next generation of therapies for urothelial carcinoma will be based on patient-specific targetable mutations found in individual tumours. This personalized-medicine approach to urothelial carcinoma has already resulted in unique clinical trial design and has the potential to improve patient outcomes and survival.
Collapse
Affiliation(s)
- Alexander P Glaser
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Damiano Fantini
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Ali Shilatifard
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Edward M Schaeffer
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Joshua J Meeks
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| |
Collapse
|
297
|
Therapeutics Targeting FGF Signaling Network in Human Diseases. Trends Pharmacol Sci 2016; 37:1081-1096. [DOI: 10.1016/j.tips.2016.10.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 12/14/2022]
|
298
|
Cho SH, Hong CS, Kim HN, Shin MH, Kim KR, Shim HJ, Hwang JE, Bae WK, Chung IJ. FGFR4 Arg388 Is Correlated with Poor Survival in Resected Colon Cancer Promoting Epithelial to Mesenchymal Transition. Cancer Res Treat 2016; 49:766-777. [PMID: 27857023 PMCID: PMC5512371 DOI: 10.4143/crt.2016.457] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 10/19/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Fibroblast growth factor receptor 4 (FGFR4) plays an important role in cancer progression during tumor proliferation, invasion, and metastasis. This study evaluated the prognostic role of FGFR4 polymorphism in patients with resected colon cancer, including the underlying mechanism. MATERIALS AND METHODS FGFR4 polymorphism was characterized in patientswho received curative resection for stage III colon cancer. FGFR4-dependent signal pathways involving cell proliferation, invasion, and migration according to genotypes were also evaluated in transfected colon cancer cell lines. RESULTS Among a total of 273 patients, the GG of FGFR4 showed significantly better overall survival than the AG or AA, regardless of adjuvant treatment. In the group of AG or AA, combination of folinic acid, fluorouracil, and oxaliplatin (FOLFOX) resulted in better survival than fluorouracil/leucovorin or no adjuvant chemotherapy. However, in GG, there was no difference among treatment regimens. Using multivariate analyses, the Arg388 carriers, together with age, N stage, poor differentiation, absence of a lymphocyte response, and no adjuvant chemotherapy, had a significantly worse OS than patients with the Gly388 allele. In transfected colon cancer cells, overexpression of Arg388 significantly increased cell proliferation and changes in epithelial to mesenchymal transition markers compared with cells overexpressing the Gly388 allele. CONCLUSION The Arg388 allele of FGFR4 may be a biomarker and a candidate target for adjuvant treatment of patients with resected colon cancer.
Collapse
Affiliation(s)
- Sang Hee Cho
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Chang Soo Hong
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hee Nam Kim
- Department of Preventive Medicine, Chonnam National Medical School, Gwangju, Korea
| | - Min Ho Shin
- Department of Preventive Medicine, Chonnam National Medical School, Gwangju, Korea
| | - Ka Rham Kim
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyun Jeong Shim
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Jun Eul Hwang
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Woo Kyun Bae
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Ik Joo Chung
- Department of Hemato-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| |
Collapse
|
299
|
Fazilaty H, Mehdipour P. Genetics of breast cancer bone metastasis: a sequential multistep pattern. Clin Exp Metastasis 2014; 31:595-612. [PMID: 24493024 DOI: 10.1007/s10585-014-9642-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/26/2014] [Indexed: 02/05/2023]
Abstract
Bone metastasis accounts for the vast majority of breast cancer (BC) metastases, and is related to a high rate of morbidity and mortality. A number of seminal studies have uncovered gene expression signatures involved in BC development and bone metastasis; each of them points at a distinct step of the 'invasion-metastasis cascade'. In this review, we provide most recently discovered functions of sets of genes that are selected from widely accepted gene signatures that are implicate in BC progression and bone metastasis. We propose a possible sequential pattern of gene expression that may lead a benign primary breast tumor to get aggressiveness and progress toward bone metastasis. A panel of genes which primarily deal with features like DNA replication, survival, proliferation, then, angiogenesis, migration, and invasion has been identified. TGF-β, FGF, NFκB, WNT, PI3K, and JAK-STAT signaling pathways, as the key pathways involved in breast cancer development and metastasis, are evidently regulated by several genes in all three signatures. Epithelial to mesenchymal transition that is also an important mechanism in cancer stem cell generation and metastasis is evidently regulated by these genes. This review provides a comprehensive insight regarding breast cancer bone metastasis that may lead to a better understanding of the disease and take step toward better treatments.
Collapse
Affiliation(s)
- Hassan Fazilaty
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Pour Sina Street, P.O. Box: 14176-13151, Keshavarz Boulevard, Tehran, Iran
| | | |
Collapse
|
300
|
Frycz BA, Murawa D, Wysocki-Borejsza M, Marciniak R, Murawa P, Drews M, Jagodziński PP. Expression of 17β-hydroxysteroid dehydrogenase type 1 in gastric cancer. Biomed Pharmacother 2013; 67:651-7. [PMID: 23916544 DOI: 10.1016/j.biopha.2013.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 06/17/2013] [Indexed: 12/15/2022] Open
Abstract
There are several findings suggesting the protective role of estrogens in gastric carcinogenesis. Extragonadal 17β-estradiol (E2) may be formed during estrone (E1) reduction to E2 by 17-β-hydroxysteroid dehydrogenase type 1 (HSD17B1). Therefore, we studied the HSD17B1 transcript and protein levels in primary nontumoral and tumoral gastric tissue from the same 21 patients with gastric cancer (GC). We also assessed the effect of 5-Aza-2'-deoxycytidine (5-dAzaC), on the methylation status of HSD17B1 and its expression and conversion of E1 to E2 in HGC-27 and EPG 85-257 GC cells. We identified the presence of HSD17B1 transcript and protein in HGC-27 and EPG 85-257 GC cells as well as in primary nontumoral and tumoral tissues from patients with GC. Moreover, we found that 5-dAzaC significantly up-regulated the HSD17B1 transcript and protein levels, which is associated with increased conversion of E1 to E2 in HGC-27 and EPG 85-257 GC cells. The changes in HSD17B1 expression in both HGC-27 and EPG 85-257 cells were accompanied by 5-dAzaC induced DNA demethylation in the 5' flanking region. Our results demonstrated that HSD17B1 expression and its ability to convert the weak estrogen E1 to the more potent E2 can be associated with DNA methylation in the 5' flanking region in GC cells.
Collapse
Affiliation(s)
- Bartosz Adam Frycz
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 6 Święcickiego St., 60-781 Poznań, Poland.
| | | | | | | | | | | | | |
Collapse
|