1
|
Mehra A, Sangwan R. A Promising Paradigm Shift in Cancer Treatment with FGFR Inhibitors. Anticancer Agents Med Chem 2025; 25:2-23. [PMID: 39192641 DOI: 10.2174/0118715206318833240819031953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024]
Abstract
FGFR have been demonstrated to perform a crucial role in biological processes but their overexpression has been perceived as the operator component in the occurrence and progression of different types of carcinoma. Out of all the interest around cancer, FGFR inhibitors have assembled pace over the past few years. Therefore, FGFR inhibitors are one of the main fundamental tools to reverse drug resistance, tumor growth, and angiogenesis. Currently, many FGFR inhibitors are under the development stage or have been developed. Due to great demand and hotspots, different pharmacophores were approached to access structurally diverse FGFR inhibitors. Here, we have selected to present several representative examples such as Naphthyl, Pyrimidine, Pyridazine, Indole, and Quinoline derivatives that illustrate the diversity and advances of FGFR inhibitors in medicinal chemistry. This review focuses on the SAR study of FGFR inhibitors last five years which will be a great future scope that influences the medicinal chemist to work towards more achievements in this area.
Collapse
Affiliation(s)
- Anuradha Mehra
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| | - Rekha Sangwan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| |
Collapse
|
2
|
Siripoon T, O'Donnell C, Jin Z, Mahipal A. Fibroblast growth factor therapies in biliary tract cancers: current and future state. Expert Opin Investig Drugs 2024; 33:1245-1255. [PMID: 39629832 DOI: 10.1080/13543784.2024.2430201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Cholangiocarcinoma is the rare and aggressive tumor with poor prognosis and limited therapeutic options. Recently, there have been promising developments in molecular targeted therapies for patients following the progression of first-line chemotherapy and immunotherapy combinations. Dysregulation of fibroblast Growth Factor Receptor (FGFR) signaling is significantly associated with tumorigenesis of intrahepatic cholangiocarcinoma and has been identified as a targetable alteration. This was possible through the discovery of crucial insights into the biochemical mechanisms and pathophysiology of the FGFR pathway. AREAS COVERED This review summarizes the current state of FGFR targeted therapies, mechanisms of resistance, and future directions for FGFR-targeted therapies in patients with cholangiocarcinoma. EXPERT OPINION Currently, pemigatinib and futibatinib are FDA approved FGFR-targeted therapies that have demonstrated remarkable responses. However, there is still a significant proportion of patients whose disease remains intrinsically resistant to treatment and most patients eventually develop secondary resistance after an initial response. Additionally, unique side effects of FGFR inhibitors may limit their efficacy in clinical practice and can have detrimental effects on quality of life. Several novel FGFR inhibitors are currently being investigated to overcome resistance mechanisms and reduce toxicities.
Collapse
Affiliation(s)
| | | | - Zhaohui Jin
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Amit Mahipal
- Department of Oncology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
3
|
Popoiu TA, Pîrvu CA, Popoiu CM, Iacob ER, Talpai T, Voinea A, Albu RS, Tãban S, Bãlãnoiu LM, Pantea S. Gastrointestinal Stromal Tumors (GISTs) in Pediatric Patients: A Case Report and Literature Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1040. [PMID: 39334573 PMCID: PMC11429550 DOI: 10.3390/children11091040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
Gastrointestinal stromal tumors (GISTs) are rare mesenchymal neoplasms that primarily affect adults, with pediatric cases constituting only 0.5-2.7% of the total. Pediatric GISTs present unique clinical, genetic, and pathological features that distinguish them from adult cases. This literature review aims to elucidate these differences, emphasizing diagnostic and therapeutic challenges. We discuss the resistance of pediatric GISTs to conventional chemotherapy and highlight the importance of surgical intervention, especially in emergency situations involving intra-abdominal bleeding. The review also explores the molecular characteristics of pediatric GISTs, including rare mutations such as quadruple-negative wild-type GIST with an FGF3 gene gain mutation. To illustrate these points, we conclude with a case from our clinic involving a 15-year-old female with multiple CD117-positive gastric GISTs and a quadruple-negative wild-type genetic profile who required urgent surgical intervention following a failed tumor embolization. This case underscores the critical need for early diagnosis and individualized therapeutic strategies combining oncologic and surgical care to improve outcomes in pediatric GIST patients.
Collapse
Affiliation(s)
- Tudor-Alexandru Popoiu
- Department of General Surgery, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department III of Functional Sciences, Discipline of Medical Informatics and Biostatistics, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cãtãlin-Alexandru Pîrvu
- Department of General Surgery, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cãlin-Marius Popoiu
- Department of Pediatric Surgery, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Emil Radu Iacob
- Department of Pediatric Surgery, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Tamas Talpai
- Department of General Surgery, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Amalia Voinea
- Department of General Surgery, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Rãzvan-Sorin Albu
- Department of General Surgery, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Sorina Tãban
- Department of Pathology, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Larisa-Mihaela Bãlãnoiu
- Department of General Surgery, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Stelian Pantea
- Department of General Surgery, "Victor Babeş" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
4
|
Shang S, Zhang L, Liu K, Lv M, Zhang J, Ju D, Wei D, Sun Z, Wang P, Yuan J, Zhu Z. Landscape of targeted therapies for advanced urothelial carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:641-677. [PMID: 38966172 PMCID: PMC11220318 DOI: 10.37349/etat.2024.00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/06/2024] [Indexed: 07/06/2024] Open
Abstract
Bladder cancer (BC) is the tenth most common malignancy globally. Urothelial carcinoma (UC) is a major type of BC, and advanced UC (aUC) is associated with poor clinical outcomes and limited survival rates. Current options for aUC treatment mainly include chemotherapy and immunotherapy. These options have moderate efficacy and modest impact on overall survival and thus highlight the need for novel therapeutic approaches. aUC patients harbor a high tumor mutation burden and abundant molecular alterations, which are the basis for targeted therapies. Erdafitinib is currently the only Food and Drug Administration (FDA)-approved targeted therapy for aUC. Many potential targeted therapeutics aiming at other molecular alterations are under investigation. This review summarizes the current understanding of molecular alterations associated with aUC targeted therapy. It also comprehensively discusses the related interventions for treatment in clinical research and the potential of using novel targeted drugs in combination therapy.
Collapse
Affiliation(s)
- Shihao Shang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Lei Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Kepu Liu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Maoxin Lv
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming 65000, Yunnan, China
| | - Jie Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
- College of Life Sciences, Northwest University, Xi’an 710068, Shaanxi, China
| | - Dongen Ju
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Di Wei
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Zelong Sun
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Pinxiao Wang
- School of Clinical Medicine, Xi’an Medical University, Xi’an 710021, Shaanxi, China
| | - Jianlin Yuan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| |
Collapse
|
5
|
Oh H, Kim J, Jung SH, Ha TH, Ahn YG, Nam G, Moon K, Singh P, Kim IS. Discovery of 2,6-Naphthyridine Analogues as Selective FGFR4 Inhibitors for Hepatocellular Carcinoma. J Med Chem 2024; 67:8445-8459. [PMID: 38706130 DOI: 10.1021/acs.jmedchem.4c00758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer and is responsible for 90% of cases. Approximately 30% of patients diagnosed with HCC are identified as displaying an aberrant expression of fibroblast growth factor 19 (FGF19)-fibroblast growth factor receptor 4 (FGFR4) as an oncogenic-driver pathway. Therefore, the control of the FGF19-FGFR4 signaling pathway with selective FGFR4 inhibitors can be a promising therapy for the treatment of HCC. We herein disclose the design and synthesis of novel FGFR4 inhibitors containing a 2,6-naphthyridine scaffold. Compound 11 displayed a nanomolar potency against Huh7 cell lines and high selectivity over FGFR1-3 that were comparable to that of fisogatinib (8) as a reference standard. Additionally, compound 11 demonstrated remarkable antitumor efficacy in the Huh7 and Hep3B HCC xenograft mouse model. Moreover, bioluminescence imaging experiments with the orthotopic mouse model support that compound 11 can be considered a promising candidate for treating HCC.
Collapse
MESH Headings
- Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/therapeutic use
- Mice
- Naphthyridines/pharmacology
- Naphthyridines/chemical synthesis
- Naphthyridines/chemistry
- Naphthyridines/therapeutic use
- Cell Line, Tumor
- Structure-Activity Relationship
- Xenograft Model Antitumor Assays
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/chemical synthesis
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/therapeutic use
- Cell Proliferation/drug effects
- Drug Discovery
- Mice, Nude
- Drug Screening Assays, Antitumor
Collapse
Affiliation(s)
- Heesook Oh
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong 18469, Republic of Korea
| | - Jisook Kim
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong 18469, Republic of Korea
| | - Seung Hyun Jung
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong 18469, Republic of Korea
| | - Tae Hee Ha
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong 18469, Republic of Korea
| | - Young Gil Ahn
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong 18469, Republic of Korea
| | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kyeongwon Moon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Pargat Singh
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
6
|
Peng Y, Zhang P, Mei W, Zeng C. Exploring FGFR signaling inhibition as a promising approach in breast cancer treatment. Int J Biol Macromol 2024; 267:131524. [PMID: 38608977 DOI: 10.1016/j.ijbiomac.2024.131524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/18/2023] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
As our grasp of cancer genomics deepens, we are steadily progressing towards the domain of precision medicine, where targeted therapy stands out as a revolutionary breakthrough in the landscape of cancer therapeutics. The fibroblast growth factor receptors (FGFR) pathway has been unveiled as a fundamental instigator in the pathophysiological mechanisms underlying breast carcinoma, paving the way for the exhilarating development of precision-targeted therapeutics. In the pursuit of exploring inhibitors that specifically target the FGFR signaling pathways, a multitude of kinase inhibitors targeting FGFR has been assiduously engineered to address the heterogeneous landscape of human malignancies. This review offers an exhaustive exploration of aberrations within the FGFR pathway and their functional implications in breast cancer. Additionally, we delve into cutting-edge therapeutic approaches for the treatment of breast cancer patients bearing FGFR alterations and the management of toxicity associated with FGFR inhibitors. Furthermore, our contemplation of the evolution of cutting-edge FGFR inhibitors foresees their potential to spearhead innovative therapeutic approaches in the ongoing combat against cancer.
Collapse
Affiliation(s)
- Yan Peng
- Department of Obstetrics, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Pengfei Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Wuxuan Mei
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China; Department of General Medicine, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China.
| |
Collapse
|
7
|
Gupta S, Kashyap M, Bansal Y, Bansal G. In silico insights into design of novel VEGFR-2 inhibitors: SMILES-based QSAR modelling, and docking studies on substituted benzo-fused heteronuclear derivatives. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:265-284. [PMID: 38591137 DOI: 10.1080/1062936x.2024.2332203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/14/2024] [Indexed: 04/10/2024]
Abstract
Eight QSAR models (M1-M8) were developed from a dataset of 118 benzo-fused heteronuclear derivatives targeting VEGFR-2 by Monte Carlo optimization method of CORALSEA 2023 software. Models were generated with hybrid optimal descriptors using both SMILES and Graphs with zero- and first-order Morgan extended connectivity index from a training set of 103 derivatives. All statistical parameters for model validation were within the prescribed limits, establishing the models to be robust and of excellent quality. Among all models, split-2 of M5 was the best-fit as reflected by r v a lidation 2 , Q v a lidation 2 and MAE . Mechanistic interpretation of this model assisted the identification of structural descriptors as promoters and hinderers for VEGFR-2 inhibition. These descriptors were utilized to design novel VEGFR-2 inhibitors (YS01-YS07) by bringing modifications in compound MS90 in the dataset. Docking of all designed compounds, MS90 and sorafenib with VEGFR-2 binding site revealed favourable binding interactions. Docking score of YS07 was higher than that of MS90 and sorafenib. Molecular dynamics simulation study revealed sustained interactions of YS07 with key amino acids of VEGFR-2 at a run time of 100 ns. This study concludes the development of a best fit QSAR model which can assist the design of new anticancer agents targeting VEGFR-2.
Collapse
Affiliation(s)
- S Gupta
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - M Kashyap
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Y Bansal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - G Bansal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
8
|
Katoh M, Loriot Y, Brandi G, Tavolari S, Wainberg ZA, Katoh M. FGFR-targeted therapeutics: clinical activity, mechanisms of resistance and new directions. Nat Rev Clin Oncol 2024; 21:312-329. [PMID: 38424198 DOI: 10.1038/s41571-024-00869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Fibroblast growth factor (FGF) signalling via FGF receptors (FGFR1-4) orchestrates fetal development and contributes to tissue and whole-body homeostasis, but can also promote tumorigenesis. Various agents, including pan-FGFR inhibitors (erdafitinib and futibatinib), FGFR1/2/3 inhibitors (infigratinib and pemigatinib), as well as a range of more-specific agents, have been developed and several have entered clinical use. Erdafitinib is approved for patients with urothelial carcinoma harbouring FGFR2/3 alterations, and futibatinib and pemigatinib are approved for patients with cholangiocarcinoma harbouring FGFR2 fusions and/or rearrangements. Clinical benefit from these agents is in part limited by hyperphosphataemia owing to off-target inhibition of FGFR1 as well as the emergence of resistance mutations in FGFR genes, activation of bypass signalling pathways, concurrent TP53 alterations and possibly epithelial-mesenchymal transition-related isoform switching. The next generation of small-molecule inhibitors, such as lirafugratinib and LOXO-435, and the FGFR2-specific antibody bemarituzumab are expected to have a reduced risk of hyperphosphataemia and the ability to overcome certain resistance mutations. In this Review, we describe the development and current clinical role of FGFR inhibitors and provide perspective on future research directions including expansion of the therapeutic indications for use of FGFR inhibitors, combination of these agents with immune-checkpoint inhibitors and the application of novel technologies, such as artificial intelligence.
Collapse
Affiliation(s)
| | - Yohann Loriot
- Drug Development Department (DITEP), Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
- INSERM U981, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simona Tavolari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Zev A Wainberg
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Masaru Katoh
- M & M Precision Medicine, Tokyo, Japan.
- Department of Omics Network, National Cancer Center, Tokyo, Japan.
| |
Collapse
|
9
|
Abdel-Mohsen HT, Ibrahim MA, Nageeb AM, El Kerdawy AM. Receptor-based pharmacophore modeling, molecular docking, synthesis and biological evaluation of novel VEGFR-2, FGFR-1, and BRAF multi-kinase inhibitors. BMC Chem 2024; 18:42. [PMID: 38395926 PMCID: PMC10893631 DOI: 10.1186/s13065-024-01135-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
A receptor-based pharmacophore model describing the binding features required for the multi-kinase inhibition of the target kinases (VEGFR-2, FGFR-1, and BRAF) were constructed and validated. It showed a good overall quality in discriminating between the active and the inactive in a compiled test set compounds with F1 score of 0.502 and Mathew's correlation coefficient of 0.513. It described the ligand binding to the hinge region Cys or Ala, the glutamate residue of the Glu-Lys αC helix conserved pair, the DFG motif Asp at the activation loop, and the allosteric back pocket next to the ATP binding site. Moreover, excluded volumes were used to define the steric extent of the binding sites. The application of the developed pharmacophore model in virtual screening of an in-house scaffold dataset resulted in the identification of a benzimidazole-based scaffold as a promising hit within the dataset. Compounds 8a-u were designed through structural optimization of the hit benzimidazole-based scaffold through (un)substituted aryl substitution on 2 and 5 positions of the benzimidazole ring. Molecular docking simulations and ADME properties predictions confirmed the promising characteristics of the designed compounds in terms of binding affinity and pharmacokinetic properties, respectively. The designed compounds 8a-u were synthesized, and they demonstrated moderate to potent VEGFR-2 inhibitory activity at 10 µM. Compound 8u exhibited a potent inhibitory activity against the target kinases (VEGFR-2, FGFR-1, and BRAF) with IC50 values of 0.93, 3.74, 0.25 µM, respectively. The benzimidazole derivatives 8a-u were all selected by the NCI (USA) to conduct their anti-proliferation screening. Compounds 8a and 8d resulted in a potent mean growth inhibition % (GI%) of 97.73% and 92.51%, respectively. Whereas compounds 8h, 8j, 8k, 8o, 8q, 8r, and 8u showed a mean GI% > 100% (lethal effect). The most potent compounds on the NCI panel of 60 different cancer cell lines were progressed further to NCI five-dose testing. The benzimidazole derivatives 8a, 8d, 8h, 8j, 8k, 8o, 8q, 8r and 8u exhibited potent anticancer activity on the tested cell lines reaching sub-micromolar range. Moreover, 8u was found to induce cell cycle arrest of MCF-7 cell line at the G2/M phase and accumulating cells at the sub-G1 phase as a result of cell apoptosis.
Collapse
Affiliation(s)
- Heba T Abdel-Mohsen
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, P.O. 12622, Cairo, Egypt.
| | - Marwa A Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, P.O. 11562, Cairo, Egypt
| | - Amira M Nageeb
- High Throughput Molecular and Genetic Technology Lab, Center of Excellence for Advanced Sciences, Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki, P.O. 12622, Cairo, Egypt
| | - Ahmed M El Kerdawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, P.O. 11562, Cairo, Egypt
- School of Pharmacy, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, Lincolnshire, UK
| |
Collapse
|
10
|
Brinch CM, Aggerholm-Pedersen N, Hogdall E, Krarup-Hansen A. Medical Oncological Treatment for Patients with Gastrointestinal Stromal Tumour (GIST) - a Systematic Review. Crit Rev Oncol Hematol 2022; 172:103650. [PMID: 35283299 DOI: 10.1016/j.critrevonc.2022.103650] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/11/2022] [Accepted: 03/07/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Chemotherapy is ineffective in treating patients with Gastrointestinal Stromal Tumour (GIST). However, several types of tyrosine kinase inhibitors have been investigated since the approval of imatinib in 2001. The purpose of this report was to systematically review studies on the efficacy of neoadjuvant, adjuvant, and lifelong medical oncological treatment of GIST. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed throughout the review process. The protocol was submitted to the International prospective register of systematic reviews database (ID 251724). A systematic literature search was performed, including phase II- and III studies of biological treatment, reporting on treatment effect in patients with GIST. RESULTS Of 308 identified publications, 42 studies were included in this review. CONCLUSION This review gives an overview of the existing evidence for approved lines of oncological treatments and potential alternatives for patients with GIST in the neoadjuvant-, adjuvant- and life-long setting.
Collapse
Affiliation(s)
- Charlotte Margareta Brinch
- Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 1, DK-2730, Herlev.
| | - Ninna Aggerholm-Pedersen
- Department of Oncology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus, Denmark.
| | - Estrid Hogdall
- Department of Pathology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 73, DK-2730, Herlev, Denmark.
| | - Anders Krarup-Hansen
- Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Borgmester Ib Juuls Vej 1, DK-2730, Herlev.
| |
Collapse
|
11
|
Pérez Piñero C, Giulianelli S, Lamb CA, Lanari C. New Insights in the Interaction of FGF/FGFR and Steroid Receptor Signaling in Breast Cancer. Endocrinology 2022; 163:6491899. [PMID: 34977930 DOI: 10.1210/endocr/bqab265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Indexed: 11/19/2022]
Abstract
Luminal breast cancer (BrCa) has a favorable prognosis compared with other tumor subtypes. However, with time, tumors may evolve and lead to disease progression; thus, there is a great interest in unraveling the mechanisms that drive tumor metastasis and endocrine resistance. In this review, we focus on one of the many pathways that have been involved in tumor progression, the fibroblast growth factor/fibroblast growth factor receptor (FGFR) axis. We emphasize in data obtained from in vivo experimental models that we believe that in luminal BrCa, tumor growth relies in a crosstalk with the stromal tissue. We revisited the studies that illustrate the interaction between hormone receptors and FGFR. We also highlight the most frequent alterations found in BrCa cell lines and provide a short review on the trials that use FGFR inhibitors in combination with endocrine therapies. Analysis of these data suggests there are many players involved in this pathway that might be also targeted to decrease FGF signaling, in addition to specific FGFR inhibitors that may be exploited to increase their efficacy.
Collapse
Affiliation(s)
- Cecilia Pérez Piñero
- Instituto de Biología y Medicina Experimental, IBYME CONICET, C1428ADN Ciudad de Buenos Aires, Argentina
| | - Sebastián Giulianelli
- Instituto de Biología y Medicina Experimental, IBYME CONICET, C1428ADN Ciudad de Buenos Aires, Argentina
- Instituto de Biología de Organismos Marinos, IBIOMAR-CCT CENPAT-CONICET, U9120ACD Puerto Madryn, Argentina
| | - Caroline A Lamb
- Instituto de Biología y Medicina Experimental, IBYME CONICET, C1428ADN Ciudad de Buenos Aires, Argentina
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental, IBYME CONICET, C1428ADN Ciudad de Buenos Aires, Argentina
| |
Collapse
|
12
|
Soltan OM, Shoman ME, Abdel-Aziz SA, Narumi A, Konno H, Abdel-Aziz M. Molecular hybrids: A five-year survey on structures of multiple targeted hybrids of protein kinase inhibitors for cancer therapy. Eur J Med Chem 2021; 225:113768. [PMID: 34450497 DOI: 10.1016/j.ejmech.2021.113768] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/23/2021] [Accepted: 08/08/2021] [Indexed: 02/07/2023]
Abstract
Protein kinases have grown over the past few years as a crucial target for different cancer types. With the multifactorial nature of cancer, and the fast development of drug resistance for conventional chemotherapeutics, a strategy for designing multi-target agents was suggested to potentially increase drug efficacy, minimize side effects and retain the proper pharmacokinetic properties. Kinase inhibitors were used extensively in such strategy. Different kinase inhibitor agents which target EGFR, VEGFR, c-Met, CDK, PDK and other targets were merged into hybrids with conventional chemotherapeutics such as tubulin polymerization and topoisomerase inhibitors. Other hybrids were designed gathering kinase inhibitors with targeted cancer therapy such as HDAC, PARP, HSP 90 inhibitors. Nitric oxide donor molecules were also merged with kinase inhibitors for cancer therapy. The current review presents the hybrids designed in the past five years discussing their design principles, results and highlights their future perspectives.
Collapse
Affiliation(s)
- Osama M Soltan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Mai E Shoman
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt.
| | - Salah A Abdel-Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, 61111, Minia, Egypt
| | - Atsushi Narumi
- Department of Organic Materials Science, Graduate School of Organic Materials Science, Yamagata University, Jonan 4-3-16, Yonezawa, 992-8510, Japan
| | - Hiroyuki Konno
- Department of Biological Engineering, Graduate School of Science and Engineering, Yamagata University, Jonan 4-3-16, Yonezawa, 992-8510, Japan
| | - Mohamed Abdel-Aziz
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt.
| |
Collapse
|
13
|
García-Quiroz J, Cárdenas-Ochoa N, García-Becerra R, Morales-Guadarrama G, Méndez-Pérez EA, Santos-Cuevas C, Ramírez-Nava GJ, Segovia-Mendoza M, Prado-García H, Avila E, Larrea F, Díaz L. Antitumoral effects of dovitinib in triple-negative breast cancer are synergized by calcitriol in vivo and in vitro. J Steroid Biochem Mol Biol 2021; 214:105979. [PMID: 34438041 DOI: 10.1016/j.jsbmb.2021.105979] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/25/2021] [Accepted: 08/18/2021] [Indexed: 12/21/2022]
Abstract
Chemotherapy is a standard therapeutic option for triple-negative breast cancer (TNBC); however, its effectiveness is often compromised by drug-related toxicity and resistance development. Herein, we aimed to evaluate whether an improved antineoplastic effect could be achieved in vitro and in vivo in TNBC by combining dovitinib, a multi-kinase inhibitor, with calcitriol, a natural anticancer hormone. In vitro, cell proliferation and cell-cycle distribution were studied by sulforhodamine B-assays and flow cytometry. In vivo, dovitinib/calcitriol effects on tumor growth, angiogenesis, and endothelium activation were evaluated in xenografted mice by caliper measures, Itgb3/VEGFR2-immunohistochemistry and 99mTc-Ethylenediamine-N,N-diacetic acid/hydrazinonicotinamyl-Glu[cyclo(Arg-Gly-Asp-D-Phe-Lys)]2 (99mTc-RGD2)-tumor uptake. The drug combination elicited a synergistically improved antiproliferative effect in TNBC-derived cells, which allowed a 7-fold and a 3.3-fold dovitinib dose-reduction in MBCDF-Tum and HCC-1806 cells, respectively. Mechanistically, the co-treatment induced a cell cycle profile suggestive of cell death and DNA damage (accumulation of cells in SubG1, S, and G2/M phases), increased the number of multinucleated cells and inhibited tumor growth to a greater extent than each compound alone. Tumor uptake of 99mTc-RGD2 was reduced by dovitinib, suggesting angiogenesis inhibition, which was corroborated by decreased endothelial cell growth, tumor-vessel density and VEGFR2 expression. In summary, calcitriol synergized dovitinib anticancer effects in vitro and in vivo, allowing for a significant dose-reduction of dovitinib while maintaining its antiproliferative potency. Our results suggest the beneficial convergence of independent antitumor mechanisms of dovitinib and calcitriol to inhibit TNBC-tumor growth.
Collapse
Affiliation(s)
- Janice García-Quiroz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Nohemí Cárdenas-Ochoa
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, 04510, Ciudad de México, Mexico.
| | - Gabriela Morales-Guadarrama
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Edgar A Méndez-Pérez
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Clara Santos-Cuevas
- Departamento de Materiales Radioactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Estado de México, Mexico.
| | - Gerardo J Ramírez-Nava
- Departamento de Materiales Radioactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, 52750, Estado de México, Mexico.
| | - Mariana Segovia-Mendoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, 04510, Ciudad de México, Mexico.
| | - Heriberto Prado-García
- Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Belisario Domínguez Sección XVI, C.P. 14080, Tlalpan, Ciudad de México, Mexico.
| | - Euclides Avila
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Fernando Larrea
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| |
Collapse
|
14
|
Napolitano A, Ostler AE, Jones RL, Huang PH. Fibroblast Growth Factor Receptor (FGFR) Signaling in GIST and Soft Tissue Sarcomas. Cells 2021; 10:cells10061533. [PMID: 34204560 PMCID: PMC8235236 DOI: 10.3390/cells10061533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Sarcomas are a heterogeneous group of rare malignancies originating from mesenchymal tissues with limited therapeutic options. Recently, alterations in components of the fibroblast growth factor receptor (FGFR) signaling pathway have been identified in a range of different sarcoma subtypes, most notably gastrointestinal stromal tumors, rhabdomyosarcomas, and liposarcomas. These alterations include genetic events such as translocations, mutations, and amplifications as well as transcriptional overexpression. Targeting FGFR has therefore been proposed as a novel potential therapeutic approach, also in light of the clinical activity shown by multi-target tyrosine kinase inhibitors in specific subtypes of sarcomas. Despite promising preclinical evidence, thus far, clinical trials have enrolled very few sarcoma patients and the efficacy of selective FGFR inhibitors appears relatively low. Here, we review the known alterations of the FGFR pathway in sarcoma patients as well as the preclinical and clinical evidence for the use of FGFR inhibitors in these diseases. Finally, we discuss the possible reasons behind the current clinical data and highlight the need for biomarker stratification to select patients more likely to benefit from FGFR targeted therapies.
Collapse
Affiliation(s)
- Andrea Napolitano
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (A.N.); (A.E.O.); (R.L.J.)
- Department of Medical Oncology, University Campus Bio-Medico, 00128 Rome, Italy
| | - Alexandra E. Ostler
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (A.N.); (A.E.O.); (R.L.J.)
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, 203 Fulham Road, London SW3 6JJ, UK; (A.N.); (A.E.O.); (R.L.J.)
- The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Paul H. Huang
- The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Correspondence: ; Tel.: +44-207-153-5554
| |
Collapse
|
15
|
Margiotta A. All Good Things Must End: Termination of Receptor Tyrosine Kinase Signal. Int J Mol Sci 2021; 22:ijms22126342. [PMID: 34198477 PMCID: PMC8231876 DOI: 10.3390/ijms22126342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/28/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are membrane receptors that regulate many fundamental cellular processes. A tight regulation of RTK signaling is fundamental for development and survival, and an altered signaling by RTKs can cause cancer. RTKs are localized at the plasma membrane (PM) and the major regulatory mechanism of signaling of RTKs is their endocytosis and degradation. In fact, RTKs at the cell surface bind ligands with their extracellular domain, become active, and are rapidly internalized where the temporal extent of signaling, attenuation, and downregulation are modulated. However, other mechanisms of signal attenuation and termination are known. Indeed, inhibition of RTKs’ activity may occur through the modulation of the phosphorylation state of RTKs and the interaction with specific proteins, whereas antagonist ligands can inhibit the biological responses mediated by the receptor. Another mechanism concerns the expression of endogenous inactive receptor variants that are deficient in RTK activity and take part to inactive heterodimers or hetero-oligomers. The downregulation of RTK signals is fundamental for several cellular functions and the homeostasis of the cell. Here, we will review the mechanisms of signal attenuation and termination of RTKs, focusing on FGFRs.
Collapse
Affiliation(s)
- Azzurra Margiotta
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| |
Collapse
|
16
|
Halting the FGF/FGFR axis leads to antitumor activity in Waldenström macroglobulinemia by silencing MYD88. Blood 2021; 137:2495-2508. [PMID: 33197938 DOI: 10.1182/blood.2020008414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/30/2020] [Indexed: 01/12/2023] Open
Abstract
The human fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) axis deregulation is largely involved in supporting the pathogenesis of hematologic malignancies, including Waldenström macroglobulinemia (WM). WM is still an incurable disease, and patients succumb because of disease progression. Therefore, novel therapeutics designed to specifically target deregulated signaling pathways in WM are required. We aimed to investigate the role of FGF/FGFR system blockade in WM by using a pan-FGF trap molecule (NSC12). Wide-transcriptome profiling confirmed inhibition of FGFR signaling in NSC12-treated WM cells; unveiling a significant inhibition of MYD88 was also confirmed at the protein level. Importantly, the NSC12-dependent silencing of MYD88 was functionally active, as it led to inhibition of MYD88-driven pathways, such as BTK and SYK, as well as the MYD88-downstream target HCK. Of note, both canonical and noncanonical NF-κB cascades were downregulated in WM cells upon NSC12 treatment. Functional sequelae exerted by NSC12 in WM cells were studied, demonstrating significant inhibition of WM cell growth, induction of WM cell apoptosis, halting MAPK, JAK/STAT3, and PI3K-Akt pathways. Importantly, NSC12 exerted an anti-WM effect even in the presence of bone marrow microenvironment, both in vitro and in vivo. Our studies provide the evidence for using NSC12 as a specific FGF/FGFR system inhibitor, thus representing a novel therapeutic strategy in WM.
Collapse
|
17
|
Liu G, Chen T, Ding Z, Wang Y, Wei Y, Wei X. Inhibition of FGF-FGFR and VEGF-VEGFR signalling in cancer treatment. Cell Prolif 2021; 54:e13009. [PMID: 33655556 PMCID: PMC8016646 DOI: 10.1111/cpr.13009] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/18/2021] [Accepted: 01/29/2021] [Indexed: 02/05/2023] Open
Abstract
The sites of targeted therapy are limited and need to be expanded. The FGF‐FGFR signalling plays pivotal roles in the oncogenic process, and FGF/FGFR inhibitors are a promising method to treat FGFR‐altered tumours. The VEGF‐VEGFR signalling is the most crucial pathway to induce angiogenesis, and inhibiting this cascade has already got success in treating tumours. While both their efficacy and antitumour spectrum are limited, combining FGF/FGFR inhibitors with VEGF/VEGFR inhibitors are an excellent way to optimize the curative effect and expand the antitumour range because their combination can target both tumour cells and the tumour microenvironment. In addition, biomarkers need to be developed to predict the efficacy, and combination with immune checkpoint inhibitors is a promising direction in the future. The article will discuss the FGF‐FGFR signalling pathway, the VEGF‐VEGFR signalling pathway, the rationale of combining these two signalling pathways and recent small‐molecule FGFR/VEGFR inhibitors based on clinical trials.
Collapse
Affiliation(s)
- Guihong Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Chen
- Cardiology Department, Chengdu NO.7 People's Hospital, Chengdu Tumor Hospital, Chengdu, China
| | - Zhenyu Ding
- Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Recent advances of dual FGFR inhibitors as a novel therapy for cancer. Eur J Med Chem 2021; 214:113205. [PMID: 33556787 DOI: 10.1016/j.ejmech.2021.113205] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/06/2021] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
Fibroblast growth factor receptor (FGFR) includes four highly conserved transmembrane receptor tyrosine kinases (FGFR1-4). FGF and FGFR regulate many biological processes, such as angiogenesis, wound healing and tissue regeneration. The abnormal expression of FGFR is related to the tumorigenesis, tumor progression and drug resistance of anti-tumor treatments in many types of tumors. Nowadays there are many anti-cancer drugs targeting FGFR. However, traditional single-target anti-tumor drugs are easy to acquire drug resistance. The therapeutic effect can be enhanced by simultaneously inhibiting FGFR and another target (such as VEGFR, EGFR, PI3K, CSF-1R, etc.). We know drug combination can bring problems such as drug interactions. Therefore, the development of FGFR dual target inhibitors is an important direction. In this paper, we reviewed the research on dual FGFR inhibitors in recent years and made brief comments on them.
Collapse
|
19
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 454] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
20
|
Praphanwittaya P, Jansook P, Loftsson T. Aqueous solubility of kinase inhibitors: III the effect of acidic counter ion on the dovitinib/γ-cyclodextrin complexation. J INCL PHENOM MACRO 2020. [DOI: 10.1007/s10847-020-01009-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
21
|
Dai H, Liu C, Li P, Mai Z, Tan X, Chen S, Zhou Z, Tang Z, Miao J, Liu L, Fang Y. Risk of Dyslipidemia Associated with VEGF/VEGFR Inhibitors: A Meta-Analysis. Transl Oncol 2020; 13:100779. [PMID: 32375082 PMCID: PMC7205761 DOI: 10.1016/j.tranon.2020.100779] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/02/2020] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE This meta-analysis was performed to investigate hyperlipidemia in patients with carcinoma treated with vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor (VEGFR) inhibitors. METHODS We searched for eligible phase II and III studies using PubMed and Embase databases. We then summarized reported occurrences of hyperlipidemia in patients with different cancers. Relative risk ratios (RRs) and 95% confidence intervals (CIs) were calculated by Revman 5 software in meta-analysis. RESULTS Eleven trials (4760 subjects) were included in this meta-analysis. Overall, VEGF/VEGFR inhibitors had similar incidence of hypertriglyceridemia (RR = 0.56, 95% CI = 0.24%-1.32%, P = .19), hypercholesterolemia (RR = 1.15, 95% CI = 0.42%-3.16%, P = .78), and LDL elevation (RR = 4.58, 95% CI = 0.80%-26.25%, P = .09) than control drugs, under high heterogeneity. Moreover, subgroup analyses found VEGF/VEGFR inhibitors had higher incidence of hypertriglyceridemia (RR = 1.86, 95% CI = 1.37%-2.52%, P < .001) and hypercholesterolemia (RR = 2.95, 95% CI = 2.02%-4.30%, P = .006) than blank control or placebo control drugs (placebo-controlled-group), although with lower incidence of hypertriglyceridemia (RR = 0.29, 95% CI = 0.12%-0.69%, P < .001) and hypercholesterolemia (RR = 0.39, 95% CI = 0.28%-0.56%, P < .001) than positive control drugs (positive-controlled-groups). CONCLUSION The use of VEGF/VEGFR inhibitors, especially the multitargeted VEGFR tyrosine kinase inhibitors (VEGFR-TKIs), was associated with higher risk of hyperlipidemia than the use of placebo, but this risk was less than that associated with mTOR or FGFR inhibitors. It indicated that clinicians need to pay close attention to the occurrence of hyperlipidemia in VEGFR-TKIs therapies.
Collapse
Affiliation(s)
- Huihui Dai
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Chang Liu
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Peijuan Li
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Zhangfeng Mai
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Xiaoming Tan
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Sijing Chen
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Ziling Zhou
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Zhiben Tang
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Jingwei Miao
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518
| | - Lizhong Liu
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518.
| | - Yi Fang
- Phase I Clinical Research Unit, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, No. B24, Yinquan road, Qingcheng district, Qingyuan city, Guangdong Province, China 511518; Department of Pharmacy, Peking University People's Hospital, Beijing, China. 100034.
| |
Collapse
|
22
|
Solubilization and in vitro permeation of dovitinib/cyclodextrin complexes and their aggregates. J INCL PHENOM MACRO 2020. [DOI: 10.1007/s10847-020-00995-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
23
|
Silveira IC, Carneiro ACDM, Hiss LS, Crema VO. Anti-Proliferative Role of the Tyrosine Kinase Inhibitors TKI-258 on Oral Squamous Cell Carcinoma In Vitro. Anticancer Agents Med Chem 2020; 20:751-755. [PMID: 32053087 DOI: 10.2174/1871520620666200213110944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Identification of the antitumor role of tyrosine kinase inhibitors, such as TKI-258, may lead to novel therapeutics for Oral Squamous Cell Carcinoma (OSCC), which has high mortality rates. TKI-258 blocks Fibroblast Growth Factor Receptors (FGFRs), Platelet-Derived Growth Factor Receptors (PDGFRs), and Endothelial Growth Factor Receptor (VEGFRs). OBJECTIVE This study aimed to evaluate the effect of TKI-258 treatment on cell proliferation in SCC-4 cells of OSCC. METHODS BrdU and KI-67 assays were performed by using SCC-4 cells. Control was compared to 1, 5 and 10μM TKI-258 treatment. Control vehicle was compared to: 60μM LY294002 (LY), 2μM Wortmannin (WTN) and LY+WNT. Moreover, TKI 5μM treatment was compared to: TKI 5μM+LY; TKI 5 μM+WTN; TKI 5μM+LY+WTN. After 6h of treatments, immunofluorescence stained BrdU and KI-67 positive cells. Morphometry of proliferative cells was analyzed considering significance of p<0.05. RESULTS BrdU and KI-67 assays results were similar for all experiments. TKI-258 treatment leads to an important reduction in proliferation rate in SCC-4 cells in a concentration dependent manner. As expected, there was a significant reduction in the percentage of proliferative cells that had PI3K inhibited. When compared with TKI 5 treatment, proliferating cells were significantly lower with simultaneous PI3K inhibition. CONCLUSION This study demonstrated that TKI-258 plays an anti-proliferative role on SCC-4 cells of OSCC. It could be interesting to block multiples pathways such as FGFRs, PDGFRs and VEGFRs. Therefore, TKI-258 is a promising option for novel therapeutics for OSCC, especially if associated with PI3K inhibition.
Collapse
Affiliation(s)
- Isadora C Silveira
- Structural Biology Department, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Anna Cecília D M Carneiro
- Structural Biology Department, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Lorraine S Hiss
- Structural Biology Department, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Virgínia O Crema
- Structural Biology Department, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| |
Collapse
|
24
|
Casadei C, Dizman N, Schepisi G, Cursano MC, Basso U, Santini D, Pal SK, De Giorgi U. Targeted therapies for advanced bladder cancer: new strategies with FGFR inhibitors. Ther Adv Med Oncol 2019; 11:1758835919890285. [PMID: 31803255 PMCID: PMC6878604 DOI: 10.1177/1758835919890285] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022] Open
Abstract
Inhibitors of fibroblast growth factor receptor (FGFR) represent an outstanding treatment approach for selected patients with urothelial cancer (UC). These agents are changing the clinical approach to a subgroup of UC, the luminal-papillary subtype, characterized by FGFR mutations, fusions, or amplification. In this review, we provide an overview of the results of recent clinical trials on FGFR tyrosine kinase inhibitors (TKIs) currently in clinical development for the treatment of UC: erdafitinib, rogaratinib, infigratinib, and the monoclonal antibody vofatamab. The Food and Drug Administration recently granted accelerated approval to erdafitinib for patients with advanced UC with alterations of FGFR2 or FGFR3 after progression on platinum-based chemotherapy. We also look at future therapeutic options of combination regimens with immune-checkpoint inhibitors as strategies for improving the antitumor effects of this class of drug, and for preventing or delaying the development of resistance.
Collapse
Affiliation(s)
- Chiara Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Nazli Dizman
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Giuseppe Schepisi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Umberto Basso
- Medical Oncology Unit, Istituto Oncologico Veneto, IOV-IRCCS, Padova, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Rome, Italy
| | - Sumanta K. Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, Meldola, 47014, Italy
| |
Collapse
|
25
|
Wang D, Yang L, Yu W, Zhang Y. Investigational fibroblast growth factor receptor 2 antagonists in early phase clinical trials to treat solid tumors. Expert Opin Investig Drugs 2019; 28:903-916. [PMID: 31560229 DOI: 10.1080/13543784.2019.1672655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Fibroblast growth factor receptor 2 (FGFR2) is a highly conserved transmembrane tyrosine kinase receptor. FGFR2 dysregulation occurs in numerous human solid tumors and overexpression is closely associated with tumor progression. FGFR2 has recently been reported as a therapeutic target for cancer. Several targeted therapies are being investigated to disrupt FGFR2 activity; these include multi-target tyrosine kinase inhibitors (TKIs), pan-FGFR targeted TKIs and FGFR2 monoclonal antibodies. Areas: This review examines FGFR2 regulation and function in cancer and its potential as a target for cancer treatment. Expert opinion: Highly specific FGFR2 blockers have not yet been developed and moreover, resistance to FGFR2-targeted therapies is a challenge. More sophisticated patient selection strategies would help improve FGFR2-targeted therapies and combination therapy is considered the most promising approach for cancer patients with FGFR2 alterations.
Collapse
Affiliation(s)
- Dan Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Henan Key Laboratory for Tumor Immunology and Biotherapy , Zhengzhou , Henan , P.R. China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Henan Key Laboratory for Tumor Immunology and Biotherapy , Zhengzhou , Henan , P.R. China
| | - Weina Yu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Henan Key Laboratory for Tumor Immunology and Biotherapy , Zhengzhou , Henan , P.R. China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , P.R. China.,Henan Key Laboratory for Tumor Immunology and Biotherapy , Zhengzhou , Henan , P.R. China.,School of Life Sciences, Zhengzhou University , Zhengzhou , Henan , P.R. China
| |
Collapse
|
26
|
Comparison of the Effects of Dovitinib and Bevacizumab on Reducing Neovascularization in an Experimental Rat Corneal Neovascularization Model. Cornea 2019; 38:1161-1168. [PMID: 31180924 DOI: 10.1097/ico.0000000000002012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To compare the inhibitory effects of dovitinib and bevacizumab for treatment of corneal neovascularization (CNV). METHODS Thirty-nine adult female Sprague Dawley rats weighing 180 to 250 g were used. CNV was induced by silver nitrate in the right eye of each rat. After the chemical burn, the animals were randomized into 5 groups. Group 1 did not receive any chemical substance. Group 2 received dimethyl sulfoxide, group 3 received bevacizumab 5 mg/mL, group 4 received dovitinib 5 mg/mL, and group 5 received bevacizumab 5 mg/mL + dovitinib 5 mg/mL topically administered twice daily for 14 days. On the 14th day, slit-lamp examination was performed, and anterior segment photographs were taken. The corneal neovascular area was measured on photographs as the percentage of the cornea's total area using computer imaging analysis. The corneal sections were stained with hematoxylin and eosin for histopathological examination. RESULTS A statistically significant decrease in the percentage of CNV was found in all treatment groups (group 3, group 4, and group 5) compared with the control group (group 1) (P < 0.01). A statistically significant difference in the percentage of CNV was found among group 3, group 4, and group 5 (P = 0.003). The percentage of CNV in group 4 was significantly higher than that in group 3 and group 5 (P1 = 0.004; P2 = 0.006). There was no statistically significant difference in the percentage of CNV between group 3 and group 5 (P = 0.228). CONCLUSIONS Dovitinib is a newly developed multitargeted tyrosine kinase inhibitor. Topical administration of dovitinib effectively inhibited CNV, but this effect of dovitinib was found less than topical bevacizumab.
Collapse
|
27
|
Dovitinib Triggers Apoptosis and Autophagic Cell Death by Targeting SHP-1/ p-STAT3 Signaling in Human Breast Cancers. JOURNAL OF ONCOLOGY 2019; 2019:2024648. [PMID: 31485222 PMCID: PMC6710795 DOI: 10.1155/2019/2024648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 07/29/2019] [Indexed: 01/13/2023]
Abstract
Breast cancer is the most common cancer and the leading cause of cancer deaths in women worldwide. The rising incidence rate and female mortality make it a significant public health concern in recent years. Dovitinib is a novel multitarget receptor tyrosine kinase inhibitor, which has been enrolled in several clinical trials in different cancers. However, its antitumor efficacy has not been well determined in breast cancers. Our results demonstrated that dovitinib showed significant antitumor activity in human breast cancer cell lines with dose- and time-dependent manners. Downregulation of phosphor-(p)-STAT3 and its subsequent effectors Mcl-1 and cyclin D1 was responsible for this drug effect. Ectopic expression of STAT3 rescued the breast cancer cells from cell apoptosis induced by dovitinib. Moreover, SHP-1 inhibitor reversed the downregulation of p-STAT3 induced by dovitinib, indicating that SHP-1 mediated the STAT3 inhibition effect of dovitinib. In addition to apoptosis, we found for the first time that dovitinib also activated autophagy to promote cell death in breast cancer cells. In conclusion, dovitinib induced both apoptosis and autophagy to block the growth of breast cancer cells by regulating the SHP-1-dependent STAT3 inhibition.
Collapse
|
28
|
Exploitation of phage display for the development of anti-cancer agents targeting fibroblast growth factor signaling pathways: New strategies to tackle an old challenge. Cytokine Growth Factor Rev 2019; 46:54-65. [DOI: 10.1016/j.cytogfr.2019.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/05/2019] [Accepted: 03/11/2019] [Indexed: 01/20/2023]
|
29
|
Okuno T, Yashiro M, Masuda G, Togano S, Kuroda K, Miki Y, Hirakawa K, Ohsawa M, Wanibuchi H, Ohira M. Establishment of a New Scirrhous Gastric Cancer Cell Line with FGFR2 Overexpression, OCUM-14. Ann Surg Oncol 2019; 26:1093-1102. [PMID: 30652228 DOI: 10.1245/s10434-018-07145-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND The prognosis of scirrhous gastric carcinoma (SGC), which is characterized by rapid infiltration and proliferation of cancer cells accompanied by extensive stromal fibrosis, is extremely poor. In this study, we report the establishment of a unique SGC cell line from a gastric cancer patient in whom an autopsy was performed. METHODS A new SGC cell line, OCUM-14, was established from malignant ascites of a male patient with SGC. A postmortem autopsy was performed on the patient. Characterization of OCUM-14 cells was analyzed by microscopic examination, reverse transcription polymerase chain reaction, fluorescence in situ hybridization analysis, immunohistochemical examination, CCK-8 assay, and in vivo assay. RESULTS OCUM-14 cells grew singly or in clusters, and were floating and round-shaped. Most OCUM-14 cells had many microvilli on their surfaces. The doubling time was 43.1 h, and the subcutaneous inoculation of 1.0 × 107 OCUM-14 cells into mice resulted in 50% tumor formation. mRNA expressions of fibroblast growth factor receptor 2 (FGFR2) and human epidermal growth factor receptor 2 (HER2) were observed in OCUM-14 cells. FGFR2, but not HER2, overexpression was found in OCUM-14 cells. The heterogeneous overexpression of FGFR2 was also found in both the primary tumor and metastatic lesions of the peritoneum, lymph node, bone marrow, and lung of the patient. The FGFR2 inhibitors AZD4547 and BGJ398 significantly decreased the growth of OCUM-14 cells, while paclitaxel and 5-fluorouracil significantly decreased the proliferation of OCUM-14 cells, but cisplatin did not. CONCLUSION A new gastric cancer cell line, OCUM-14, was established from SGC and showed FGFR2 overexpression. OCUM-14 might be useful for elucidating the characteristic mechanisms of SGC and clarifying the effect of FGFR2 inhibitors on SGC.
Collapse
Affiliation(s)
- Tomohisa Okuno
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan.,Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan. .,Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan. .,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan.
| | - Go Masuda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan
| | - Shingo Togano
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan.,Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Kenji Kuroda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan.,Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Yuichiro Miki
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan.,Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan
| | - Masahiko Ohsawa
- Department of Diagnostic Pathology, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Hideki Wanibuchi
- Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Masaichi Ohira
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka City, Osaka, Japan
| |
Collapse
|
30
|
Haq F, Sung YN, Park I, Kayani MA, Yousuf F, Hong SM, Ahn SM. FGFR1 expression defines clinically distinct subtypes in pancreatic cancer. J Transl Med 2018; 16:374. [PMID: 30593273 PMCID: PMC6311038 DOI: 10.1186/s12967-018-1743-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/12/2018] [Indexed: 12/31/2022] Open
Abstract
Background The clinical significance of fibroblast growth factor receptor 1 (FGFR1) protein expression in pancreatic cancer is largely unknown. In this study, we aimed investigate the clinical significance of FGFR1 expression in pancreatic cancer. Methods First, we investigated the relationship between FGFR pathway gene expression and clinicopathological data in three pancreatic cancer cohorts containing 313 cases. Subsequently, to confirm the findings from the discovery cohorts, we performed immunohistochemistry (IHC) of FGFR1 protein in a validation cohort of 205 pancreatic cancer cases. Results In discovery cohort 1, FGFR1 and Klotho beta (KLB) overexpression was associated with low tumor stage (P < 0.05), low tumor grade (P < 0.05), and better overall survival. Multivariate analysis predicted FGFR1 (P < 0.05) as a prognostic factor for better overall survival. In discovery cohorts 2 and 3, only FGFR1 overexpression was associated with better overall survival (P < 0.05). In the validation cohort, there were 15.7% and 61% strong and weak/moderate FGFR1-positive cases, respectively. FGFR1-positive cases showed better overall survival than FGFR1-negative cases (P < 0.05). Furthermore, multivariate analysis revealed FGFR1 positivity as an independent prognostic factor for better overall survival in pancreatic cancer patients (hazard ratio 0.677, 95% confidence interval 0.471–0.972, P = 0.035). Conclusions FGFR1 expression, as estimated by IHC, may be used to define clinically distinct subtypes in pancreatic cancer. Moreover, FGFR1-based subclassification of pancreatic cancer may lead to new therapeutic approaches for the FGFR1-positive subtype. Electronic supplementary material The online version of this article (10.1186/s12967-018-1743-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Farhan Haq
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - You-Na Sung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Olympic-Ro 43Gil 88, Songpa-Gu, Seoul, Republic of Korea
| | - Inkeun Park
- Division of Oncology, Department of Internal Medicine, Gachon University Gil Hospital, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | | | - Faizah Yousuf
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Olympic-Ro 43Gil 88, Songpa-Gu, Seoul, Republic of Korea.
| | - Sung-Min Ahn
- Division of Oncology, Department of Internal Medicine, Gachon University Gil Hospital, Gachon University Gil Medical Center, Incheon, Republic of Korea. .,Department of Genome Medicine and Science, College of Medicine, Gachon Institute of Genome Medicine and Science, Gachon University, Seongnam, Republic of Korea.
| |
Collapse
|
31
|
Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol 2018; 16:105-122. [DOI: 10.1038/s41571-018-0115-y] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Świderska KW, Szlachcic A, Opaliński Ł, Zakrzewska M, Otlewski J. FGF2 Dual Warhead Conjugate with Monomethyl Auristatin E and α-Amanitin Displays a Cytotoxic Effect towards Cancer Cells Overproducing FGF Receptor 1. Int J Mol Sci 2018; 19:ijms19072098. [PMID: 30029518 PMCID: PMC6073801 DOI: 10.3390/ijms19072098] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 02/03/2023] Open
Abstract
In the rapidly developing field of targeted cancer therapy there is growing interest towards therapeutics combining two or more compounds to achieve synergistic action and minimize the chance of cancer resistance to treatment. We developed a fibroblast growth factor 2 (FGF2)-conjugate bearing two cytotoxic drugs with independent mode of action: α-amanitin and monomethyl auristatin E. Drugs are covalently attached to the targeting protein in a site-specific manner via maleimide-thiol conjugation and Cu(I)-catalyzed alkyne-azide cycloaddition. The dual warhead conjugate binds to FGF receptor 1 (FGFR1) and utilizes receptor-mediated endocytosis for selective internalization into cancer cells with FGFR1. The developed conjugate displays high cytotoxicity towards all tested FGFR1-positive cell lines. Most importantly, the improved cytotoxic effect of both drugs is observed for lung cancer cell line NCI-H446. The single drug-FGF2 conjugates have no impact on the viability of NCI-H446 cells, whereas the dual warhead-FGF2 conjugate selectively and efficiently kills these FGFR1 positive cancer cells. Due to the diversified mode of action the dual warhead-FGF2 conjugate may overcome the potential acquired resistance of FGFR1-overproducing cancer cells towards single cytotoxic drugs.
Collapse
Affiliation(s)
- Karolina Weronika Świderska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Anna Szlachcic
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
33
|
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: 74] [Impact Index Per Article: 10.6] [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
|
34
|
Lee Y, Jung JI, Park KY, Kim SA, Kim J. Synergistic inhibition effect of TNIK inhibitor KY-05009 and receptor tyrosine kinase inhibitor dovitinib on IL-6-induced proliferation and Wnt signaling pathway in human multiple myeloma cells. Oncotarget 2018; 8:41091-41101. [PMID: 28467797 PMCID: PMC5522218 DOI: 10.18632/oncotarget.17056] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/21/2017] [Indexed: 12/01/2022] Open
Abstract
Multiple myeloma is a fetal form of plasma cell malignancy characterized by abnormal clonal proliferation of plasma cells. Especially, the canonical Wnt signaling pathway mediated by β-catenin is activated in multiple myeloma cells, stimulating their proliferation. Here, we investigated the relationship between interleukin-6-induced proliferation of multiple myeloma cells and Traf2- and Nck-interacting kinase (TNIK) expression in Wnt signaling. Interleukin-6 increased the proliferation of multiple myeloma cells and TNIK mRNA and protein expression. In addition, we examined the effect on TNIK of TNIK inhibitor KY-05009 and receptor tyrosine kinase inhibitor dovitinib and whether inhibition of TNIK suppresses the interleukin-6-induced proliferation of multiple myeloma cells. KY-05009 and dovitinib synergistically inhibited interleukin-6-stimulated proliferation and induced apoptosis through the inhibition of Wnt signaling in MM cells. Our results provide crucial information that TNIK is involved in the interleukin-6-dependent proliferation of multiple myeloma cells and inhibition of Wnt signaling involving TNIK could be a therapeutic strategy for the treatment of interleukin-6-dependent multiple myeloma.
Collapse
Affiliation(s)
- Yura Lee
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824, Korea.,Present address: Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul 03722, Korea
| | - Jung-Il Jung
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824, Korea
| | | | - Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824, Korea
| | - Jiyeon Kim
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824, Korea
| |
Collapse
|
35
|
Richard NP, Pippa R, Cleary MM, Puri A, Tibbitts D, Mahmood S, Christensen DJ, Jeng S, McWeeney S, Look AT, Chang BH, Tyner JW, Vitek MP, Odero MD, Sears R, Agarwal A. Combined targeting of SET and tyrosine kinases provides an effective therapeutic approach in human T-cell acute lymphoblastic leukemia. Oncotarget 2018; 7:84214-84227. [PMID: 27705940 PMCID: PMC5356656 DOI: 10.18632/oncotarget.12394] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/24/2016] [Indexed: 12/21/2022] Open
Abstract
Recent evidence suggests that inhibition of protein phosphatase 2A (PP2A) tumor suppressor activity via the SET oncoprotein contributes to the pathogenesis of various cancers. Here we demonstrate that both SET and c-MYC expression are frequently elevated in T-ALL cell lines and primary samples compared to healthy T cells. Treatment of T-ALL cells with the SET antagonist OP449 restored the activity of PP2A and reduced SET interaction with the PP2A catalytic subunit, resulting in a decrease in cell viability and c-MYC expression in a dose-dependent manner. Since a tight balance between phosphatases and kinases is required for the growth of both normal and malignant cells, we sought to identify a kinase inhibitor that would synergize with SET antagonism. We tested various T-ALL cell lines against a small-molecule inhibitor screen of 66 compounds targeting two-thirds of the tyrosine kinome and found that combined treatment of T-ALL cells with dovitinib, an orally active multi-targeted small-molecule receptor tyrosine kinase inhibitor, and OP449 synergistically reduced the viability of all tested T-ALL cell lines. Mechanistically, combined treatment with OP449 and dovitinib decreased total and phospho c-MYC levels and reduced ERK1/2, AKT, and p70S6 kinase activity in both NOTCH-dependent and independent T-ALL cell lines. Overall, these results suggest that combined targeting of tyrosine kinases and activation of serine/threonine phosphatases may offer novel therapeutic strategies for the treatment of T-ALL.
Collapse
Affiliation(s)
- Nameeta P Richard
- Randall Children's Hospital at Legacy Emanuel, Children's Cancer and Blood Disorders Program, Portland, OR 97227, USA.,Division of Pediatric Hematology Oncology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Raffaella Pippa
- Division of Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Megan M Cleary
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Alka Puri
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Deanne Tibbitts
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Shawn Mahmood
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Dale J Christensen
- Research and Development, Oncotide Pharmaceuticals, Research Triangle Park, NC 27710, USA .,Spyryx Biosciences, Durham, NC 27713, USA
| | - Sophia Jeng
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Shannon McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - A Thomas Look
- Dana-Farber Cancer Institute, Harvard Cancer Center, Boston, MA 02215, USA
| | - Bill H Chang
- Division of Pediatric Hematology Oncology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jeffrey W Tyner
- Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Michael P Vitek
- Research and Development, Oncotide Pharmaceuticals, Research Triangle Park, NC 27710, USA
| | - María D Odero
- Division of Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Rosalie Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR USA-97239.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA-97239
| | - Anupriya Agarwal
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, USA-97239.,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR USA-97239.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA-97239
| |
Collapse
|
36
|
Joensuu H, Blay JY, Comandone A, Martin-Broto J, Fumagalli E, Grignani G, Del Muro XG, Adenis A, Valverde C, Pousa AL, Bouché O, Italiano A, Bauer S, Barone C, Weiss C, Crippa S, Camozzi M, Castellana R, Le Cesne A. Dovitinib in patients with gastrointestinal stromal tumour refractory and/or intolerant to imatinib. Br J Cancer 2017; 117:1278-1285. [PMID: 28850565 PMCID: PMC5672922 DOI: 10.1038/bjc.2017.290] [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: 04/07/2017] [Revised: 07/04/2017] [Accepted: 07/31/2017] [Indexed: 12/20/2022] Open
Abstract
Background: This multicentre phase II trial (DOVIGIST) evaluated the antitumour activity of dovitinib as second-line treatment of patients with gastrointestinal stromal tumour (GIST) refractory to imatinib or who do not tolerate imatinib. Methods: Patients received oral dovitinib 500 mg day−1, 5 days on/2 days off, until GIST progression or unacceptable toxicity, with an objective to evaluate efficacy, assessed as the disease control rate (DCR) at 12 weeks. Tumour assessment and response to dovitinib therapy were evaluated by Response Evaluation Criteria In Solid Tumours (RECIST v1.1) and the Choi criteria. Secondary objectives included assessment of progression-free survival (PFS), safety and tolerability, and DCR at the end of treatment. Results: Thirty-eight of the 39 patients enrolled had histologically confirmed GIST. The DCR at 12 weeks was 52.6% (90% confidence interval (CI), 38.2–66.7%) meeting the preset efficacy criterion for the primary end point. The objective response rate (complete response+partial response) was 2.6% (1 of 38; 90% CI, 0.1–11.9%), and 5.3% (n=2; 90% CI, 0.9–15.7%) at the end of the study. The median PFS was 4.6 months (90% CI, 2.8–7.4 months). Dose interruption was required in 26 patients (66.7%), of which 18 (69.2%) were due to adverse events. The most frequently observed grade 3 adverse events included hypertension (n=7), fatigue (n=5), vomiting (n=4), hypertriglyceridaemia (n=4), and γ-glutamyltransferase increase (n=4). Conclusions: Dovitinib is an active treatment for patients with GIST who are intolerant to imatinib or whose GIST progresses on imatinib.
Collapse
Affiliation(s)
- Heikki Joensuu
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Haartmaninkatu 4, Helsinki, Finland
| | - Jean-Yves Blay
- University Claude Bernard Lyon I, Centre Leon Berard, Lyon, France
| | | | | | - Elena Fumagalli
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | - Antoine Italiano
- Institut Bergonie, Comprehensive Cancer Centre, Bordeaux, France
| | - Sebastian Bauer
- Sarcoma Center, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
| | - Carlo Barone
- University Hospital A. Gemelli, Universitá Cattolica, Rome, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Distribution of Vascular Patterns in Different Subtypes of Renal Cell Carcinoma. A Morphometric Study in Two Distinct Types of Blood Vessels. Pathol Oncol Res 2017; 24:515-524. [DOI: 10.1007/s12253-017-0262-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/21/2017] [Indexed: 10/19/2022]
|
38
|
Levin PA, Dowell JE. Spotlight on bevacizumab and its potential in the treatment of malignant pleural mesothelioma: the evidence to date. Onco Targets Ther 2017; 10:2057-2066. [PMID: 28435296 PMCID: PMC5391166 DOI: 10.2147/ott.s113598] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare, but aggressive cancer. Surgery and radiation offer limited benefit, and systemic chemotherapy remains the primary treatment modality for the majority of patients. Vascular endothelial growth factor (VEGF) and its receptor have been recognized as important players in the biology of this disease. Bevacizumab is a monoclonal antibody that binds VEGF and blocks its interaction with the VEGF receptor. Recent studies have shown benefit with the addition of bevacizumab to the combination of cisplatin and pemetrexed in MPM. This combination is now included in the National Comprehensive Cancer Network guidelines (with a category 2A recommendation) as a possible first-line treatment for unresectable MPM in appropriately selected patients. This review discusses the rationale behind the use of bevacizumab in MPM, as well as summarizes the pharmacology, efficacy, safety, and toxicity of bevacizumab across multiple trials. The use of small-molecule inhibitors of angiogenesis in the treatment of MPM is also discussed.
Collapse
Affiliation(s)
- Pavel A Levin
- Division of Hematology/Oncology, University of Texas Southwestern Medical Center
| | - Jonathan E Dowell
- Division of Hematology/Oncology, University of Texas Southwestern Medical Center.,Section of Hematology/Oncology, Veteran Affairs North Texas Health Care System, Dallas, TX, USA
| |
Collapse
|
39
|
Yadav SS, Li J, Stockert JA, Herzog B, O'Connor J, Garzon-Manco L, Parsons R, Tewari AK, Yadav KK. Induction of Neuroendocrine Differentiation in Prostate Cancer Cells by Dovitinib (TKI-258) and its Therapeutic Implications. Transl Oncol 2017; 10:357-366. [PMID: 28342996 PMCID: PMC5369368 DOI: 10.1016/j.tranon.2017.01.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer (PCa) remains the second-leading cause of cancer-related deaths in American men with an estimated mortality of more than 26,000 in 2016 alone. Aggressive and metastatic tumors are treated with androgen deprivation therapies (ADT); however, the tumors acquire resistance and develop into lethal castration resistant prostate cancer (CRPC). With the advent of better therapeutics, the incidences of a more aggressive neuroendocrine prostate cancer (NEPC) variant continue to emerge. Although de novo occurrences of NEPC are rare, more than 25% of the therapy-resistant patients on highly potent new-generation anti-androgen therapies end up with NEPC. This, along with previous observations of an increase in the number of such NE cells in aggressive tumors, has been suggested as a mechanism of resistance development during prostate cancer progression. Dovitinib (TKI-258/CHIR-258) is a pan receptor tyrosine kinase (RTK) inhibitor that targets VEGFR, FGFR, PDGFR, and KIT. It has shown efficacy in mouse-model of PCa bone metastasis, and is presently in clinical trials for several cancers. We observed that both androgen receptor (AR) positive and AR-negative PCa cells differentiate into a NE phenotype upon treatment with Dovitinib. The NE differentiation was also observed when mice harboring PC3-xenografted tumors were systemically treated with Dovitinib. The mechanistic underpinnings of this differentiation are unclear, but seem to be supported through MAPK-, PI3K-, and Wnt-signaling pathways. Further elucidation of the differentiation process will enable the identification of alternative salvage or combination therapies to overcome the potential resistance development.
Collapse
Affiliation(s)
- Shalini S Yadav
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - Jinyi Li
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - Jennifer A Stockert
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - Bryan Herzog
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - James O'Connor
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - Luis Garzon-Manco
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - Ramon Parsons
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574
| | - Kamlesh K Yadav
- Department of Urology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029-6574.
| |
Collapse
|
40
|
Porta R, Borea R, Coelho A, Khan S, Araújo A, Reclusa P, Franchina T, Van Der Steen N, Van Dam P, Ferri J, Sirera R, Naing A, Hong D, Rolfo C. FGFR a promising druggable target in cancer: Molecular biology and new drugs. Crit Rev Oncol Hematol 2017; 113:256-267. [PMID: 28427515 DOI: 10.1016/j.critrevonc.2017.02.018] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 02/08/2017] [Accepted: 02/15/2017] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION The Fibroblast Growth Factor Receptor (FGFR) family consists of Tyrosine Kinase Receptors (TKR) involved in several biological functions. Recently, alterations of FGFR have been reported to be important for progression and development of several cancers. In this setting, different studies are trying to evaluate the efficacy of different therapies targeting FGFR. AREAS COVERED This review summarizes the current status of treatments targeting FGFR, focusing on the trials that are evaluating the FGFR profile as inclusion criteria: Multi-Target, Pan-FGFR Inhibitors and anti-FGF (Fibroblast Growth Factor)/FGFR Monoclonal Antibodies. EXPERT OPINION Most of the TKR share intracellular signaling pathways; therefore, cancer cells tend to overcome the inhibition of one tyrosine kinase receptor by activating another. The future of TKI (Tyrosine Kinase Inhibitor) therapy will potentially come from multi-targeted TKIs that target different TKR simultaneously. It is crucial to understand the interaction of the FGF-FGFR axis with other known driver TKRs. Based on this, it is possible to develop therapeutic strategies targeting multiple connected TKRs at once. One correct step in this direction is the reassessment of multi target inhibitors considering the FGFR status of the tumor. Another opportunity arises from assessing the use of FGFR TKI on patients harboring FGFR alterations.
Collapse
Affiliation(s)
- Rut Porta
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Girona, Spain; Girona Biomedical Research Institute (IDIBGi), Girona, Spain; Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| | - Roberto Borea
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital (UZA) and Center for Oncological Research (CORE) Antwerp University, Edegem, Antwerp, Belgium(2)
| | - Andreia Coelho
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital (UZA) and Center for Oncological Research (CORE) Antwerp University, Edegem, Antwerp, Belgium(2)
| | - Shahanavaj Khan
- Nanomedicine and Biotechnology Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - António Araújo
- Department of Medical Oncology, Centro Hospitalar do Porto, Porto, Portugal
| | - Pablo Reclusa
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital (UZA) and Center for Oncological Research (CORE) Antwerp University, Edegem, Antwerp, Belgium(2)
| | - Tindara Franchina
- Medical Oncology Unit A.O. Papardo & Department of Human Pathology, University of Messina, Messina, Italy
| | - Nele Van Der Steen
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Edegem, Antwerp, Belgium
| | - Peter Van Dam
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital (UZA) and Center for Oncological Research (CORE) Antwerp University, Edegem, Antwerp, Belgium(2)
| | - Jose Ferri
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital (UZA) and Center for Oncological Research (CORE) Antwerp University, Edegem, Antwerp, Belgium(2)
| | - Rafael Sirera
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital (UZA) and Center for Oncological Research (CORE) Antwerp University, Edegem, Antwerp, Belgium(2)
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - David Hong
- Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Christian Rolfo
- Phase I-Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital (UZA) and Center for Oncological Research (CORE) Antwerp University, Edegem, Antwerp, Belgium(2).
| |
Collapse
|
41
|
Semrad TJ, Kim EJ, Tanaka MS, Sands J, Roberts C, Burich RA, Li Y, Gandara DR, Lara P, Mack PC. Phase II Study of Dovitinib in Patients Progressing on Anti-Vascular Endothelial Growth Factor Therapy. Cancer Treat Res Commun 2017; 10:21-26. [PMID: 28736761 DOI: 10.1016/j.ctarc.2016.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Prior work identified the fibroblast growth factor (FGF) pathway as a mediator of resistance to anti-vascular endothelial growth factor (VEGF) therapy. We tested dovitinib, an inhibitor of both FGF and VEGF receptors, in patients progressing on anti-VEGF treatment. METHODS Patients with measurable advanced colorectal or non-small cell lung cancer with progression despite anti-VEGF treatment within 56 days, good performance status and adequate organ function were eligible. A research tumor biopsy was followed by treatment with dovitinib 500 mg on a 5-day on/2-day off schedule for 28-day cycles. The primary endpoint of tumor response was evaluated every 2 cycles. Secondary endpoints included toxicity and 8-week disease control rate. Intratumor mRNA expression of angiogenic mediators was analyzed using a next generation sequencing based expression array. RESULTS Ten patients treated previously with bevacizumab or ziv-aflibercept enrolled. The study closed with termination of dovitinib development. No responses were observed in 7 evaluable patients. The best response was stable disease in 1 patient. Common toxicities included gastrointestinal, metabolic, and biochemical derangements. All patients experienced at least one grade ≥ 3 treatment-related adverse event, most commonly fatigue, elevated GGT, and lymphopenia. Expression of multiple angiogenic mediators was common in tumors progressing on anti-VEGF therapy including high levels of FGFR1 and VEGFA. CONCLUSIONS We found no evidence for the activity of dovitinib in patients who had recently progressed on anti-VEGF therapy and toxicities were significant. In tumors progressing despite anti-VEGF therapy, a multitude of pro-angiogenic mediators are expressed, including members of the FGF pathway.
Collapse
Affiliation(s)
- Thomas J Semrad
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | - Edward J Kim
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | - Michael S Tanaka
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | | | | | - Rebekah A Burich
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | - Yu Li
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | - David R Gandara
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | - Primo Lara
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | - Philip C Mack
- Division of Hematology/Oncology, University of California, Davis Comprehensive Cancer Center, Sacramento, California
| |
Collapse
|
42
|
Hahn NM, Bivalacqua TJ, Ross AE, Netto GJ, Baras A, Park JC, Chapman C, Masterson TA, Koch MO, Bihrle R, Foster RS, Gardner TA, Cheng L, Jones DR, McElyea K, Sandusky GE, Breen T, Liu Z, Albany C, Moore ML, Loman RL, Reed A, Turner SA, De Abreu FB, Gallagher T, Tsongalis GJ, Plimack ER, Greenberg RE, Geynisman DM. A Phase II Trial of Dovitinib in BCG-Unresponsive Urothelial Carcinoma with FGFR3 Mutations or Overexpression: Hoosier Cancer Research Network Trial HCRN 12-157. Clin Cancer Res 2016; 23:3003-3011. [PMID: 27932416 DOI: 10.1158/1078-0432.ccr-16-2267] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 12/11/2022]
Abstract
Purpose: To assess the clinical and pharmacodynamic activity of dovitinib in a treatment-resistant, molecularly enriched non-muscle-invasive urothelial carcinoma of the bladder (NMIUC) population.Experimental Design: A multi-site pilot phase II trial was conducted. Key eligibility criteria included the following: Bacillus Calmette-Guerin (BCG)-unresponsive NMIUC (>2 prior intravesical regimens) with increased phosphorylated FGFR3 (pFGFR3) expression by centrally analyzed immunohistochemistry (IHC+) or FGFR3 mutations (Mut+) assessed in a CLIA-licensed laboratory. Patients received oral dovitinib 500 mg daily (5 days on/2 days off). The primary endpoint was 6-month TURBT-confirmed complete response (CR) rate.Results: Between 11/2013 and 10/2014, 13 patients enrolled (10 IHC+ Mut-, 3 IHC+ Mut+). Accrual ended prematurely due to cessation of dovitinib clinical development. Demographics included the following: median age 70 years; 85% male; carcinoma in situ (CIS; 3 patients), Ta/T1 (8 patients), and Ta/T1 + CIS (2 patients); median prior regimens 3. Toxicity was frequent with all patients experiencing at least one grade 3-4 event. Six-month CR rate was 8% (0% in IHC+ Mut-; 33% in IHC+ Mut+). The primary endpoint was not met. Pharmacodynamically active (94-5,812 nmol/L) dovitinib concentrations in urothelial tissue were observed in all evaluable patients. Reductions in pFGFR3 IHC staining were observed post-dovitinib treatment.Conclusions: Dovitinib consistently achieved biologically active concentrations within the urothelium and demonstrated pharmacodynamic pFGFR3 inhibition. These results support systemic administration as a viable approach to clinical trials in patients with NMIUC. Long-term dovitinib administration was not feasible due to frequent toxicity. Absent clinical activity suggests that patient selection by pFGFR3 IHC alone does not enrich for response to FGFR3 kinase inhibitors in urothelial carcinoma. Clin Cancer Res; 23(12); 3003-11. ©2016 AACR.
Collapse
Affiliation(s)
- Noah M Hahn
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland. .,The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, Maryland.,The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Trinity J Bivalacqua
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.,The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, Maryland.,The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Ashley E Ross
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.,The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, Maryland.,The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - George J Netto
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.,The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, Maryland.,The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Alex Baras
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland.,The Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, Maryland.,The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Jong Chul Park
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Carolyn Chapman
- Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Timothy A Masterson
- Department of Urology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Michael O Koch
- Department of Urology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Richard Bihrle
- Department of Urology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Richard S Foster
- Department of Urology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Thomas A Gardner
- Department of Urology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - David R Jones
- Department of Clinical Pharmacology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Kyle McElyea
- Department of Pathology and Laboratory Medicine, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - George E Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Timothy Breen
- Hoosier Cancer Research Network, Indianapolis, Indiana
| | - Ziyue Liu
- Indiana University Department of Biostatistics, Schools of Public Health and Medicine, Indianapolis, Indiana
| | - Costantine Albany
- Division of Hematology and Oncology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Marietta L Moore
- Division of Hematology and Oncology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Rhoda L Loman
- Division of Hematology and Oncology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Angela Reed
- Division of Hematology and Oncology, Indiana University Simon Cancer Center, Indianapolis, Indiana
| | - Scott A Turner
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Francine B De Abreu
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Torrey Gallagher
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Gregory J Tsongalis
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Elizabeth R Plimack
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | - Daniel M Geynisman
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
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]
|
44
|
Design, synthesis and biological evaluation of pyrazolylaminoquinazoline derivatives as highly potent pan-fibroblast growth factor receptor inhibitors. Bioorg Med Chem Lett 2016; 26:2594-9. [PMID: 27117427 DOI: 10.1016/j.bmcl.2016.04.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 11/20/2022]
Abstract
Fibroblast growth factor receptors (FGFRs) are important oncology targets due to the dysregulation of this signaling pathway in a wide variety of human cancers. We identified a series of pyrazolylaminoquinazoline derivatives as potent FGFR inhibitors with low nanomolar potency. The representative compound 29 strongly inhibited FGFR1-3 kinase activity and suppressed FGFR signaling transduction in FGFR-addicted cancer cells; FGFRs-driven cell proliferation was also strongly inhibited regardless of mechanistic complexity implicated in FGFR activation, which further confirmed that 29 was a potent pan-FGFR inhibitor. The flexibility of our structure offered the potential to preserve good affinity for mutant FGFR, which is important for developing TKIs with long-term efficacy.
Collapse
|
45
|
Katoh M. FGFR inhibitors: Effects on cancer cells, tumor microenvironment and whole-body homeostasis (Review). Int J Mol Med 2016; 38:3-15. [PMID: 27245147 PMCID: PMC4899036 DOI: 10.3892/ijmm.2016.2620] [Citation(s) in RCA: 303] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/23/2016] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor (FGF)2, FGF4, FGF7 and FGF20 are representative paracrine FGFs binding to heparan-sulfate proteoglycan and fibroblast growth factor receptors (FGFRs), whereas FGF19, FGF21 and FGF23 are endocrine FGFs binding to Klotho and FGFRs. FGFR1 is relatively frequently amplified and overexpressed in breast and lung cancer, and FGFR2 in gastric cancer. BCR-FGFR1, CNTRL-FGFR1, CUX1-FGFR1, FGFR1OP-FGFR1, MYO18A-FGFR1 and ZMYM2-FGFR1 fusions in myeloproliferative neoplasms are non-receptor-type FGFR kinases, whereas FGFR1-TACC1, FGFR2-AFF3, FGFR2-BICC1, FGFR2-PPHLN1, FGFR3-BAIAP2L1 and FGFR3-TACC3 fusions in solid tumors are transmembrane-type FGFRs with C-terminal alterations. AZD4547, BGJ398 (infigratinib), Debio-1347 and dovitinib are FGFR1/2/3 inhibitors; BLU9931 is a selective FGFR4 inhibitor; FIIN-2, JNJ-42756493, LY2874455 and ponatinib are pan-FGFR inhibitors. AZD4547, dovitinib and ponatinib are multi-kinase inhibitors targeting FGFRs, colony stimulating factor 1 receptor (CSF1R), vascular endothelial growth factor (VEGF)R2, and others. The tumor microenvironment consists of cancer cells and stromal/immune cells, such as cancer-associated fibroblasts (CAFs), endothelial cells, M2-type tumor-associating macrophages (M2-TAMs), myeloid-derived suppressor cells (MDSCs) and regulatory T cells. FGFR inhibitors elicit antitumor effects directly on cancer cells, as well as indirectly through the blockade of paracrine signaling. The dual inhibition of FGF and CSF1 or VEGF signaling is expected to enhance the antitumor effects through the targeting of immune evasion and angiogenesis in the tumor microenvironment. Combination therapy using tyrosine kinase inhibitors (FGFR or CSF1R inhibitors) and immune checkpoint blockers (anti-PD-1 or anti-CTLA-4 monoclonal antibodies) may be a promising choice for cancer patients. The inhibition of FGF19-FGFR4 signaling is associated with a risk of liver toxicity, whereas the activation of FGF23-FGFR4 signaling is associated with a risk of heart toxicity. Endocrine FGF signaling affects the pathophysiology of cancer patients who are prescribed FGFR inhibitors. Whole-genome sequencing is necessary for the detection of promoter/enhancer alterations of FGFR genes and rare alterations of other genes causing FGFR overexpression. To sustain the health care system in an aging society, a benefit-cost analysis should be performed with a focus on disease-free survival and the total medical cost before implementing genome-based precision medicine for cancer patients.
Collapse
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo 104-0045, Japan
| |
Collapse
|
46
|
Lee Y, Bae KJ, Chon HJ, Kim SH, Kim SA, Kim J. A Receptor Tyrosine Kinase Inhibitor, Dovitinib (TKI-258), Enhances BMP-2-Induced Osteoblast Differentiation In Vitro. Mol Cells 2016; 39:389-94. [PMID: 27025387 PMCID: PMC4870186 DOI: 10.14348/molcells.2016.2300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 02/16/2016] [Accepted: 02/25/2016] [Indexed: 02/07/2023] Open
Abstract
Dovitinib (TKI258) is a small molecule multi-kinase inhibitor currently in clinical phase I/II/III development for the treatment of various types of cancers. This drug has a safe and effective pharmacokinetic/pharmacodynamic profile. Although dovitinib can bind several kinases at nanomolar concentrations, there are no reports relating to osteoporosis or osteoblast differentiation. Herein, we investigated the effect of dovitinib on human recombinant bone morphogenetic protein (BMP)-2-induced osteoblast differentiation in a cell culture model. Dovitinib enhanced the BMP-2-induced alkaline phosphatase (ALP) induction, which is a representative marker of osteoblast differentiation. Dovitinib also stimulated the translocation of phosphorylated Smad1/5/8 into the nucleus and phosphorylation of mitogen-activated protein kinases, including ERK1/2 and p38. In addition, the mRNA expression of BMP-4, BMP-7, ALP, and OCN increased with dovitinib treatment. Our results suggest that dovitinib has a potent stimulating effect on BMP-2-induced osteoblast differentiation and this existing drug has potential for repositioning in the treatment of bone-related disorders.
Collapse
Affiliation(s)
- Yura Lee
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824,
Korea
| | - Kyoung Jun Bae
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824,
Korea
| | - Hae Jung Chon
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824,
Korea
| | - Seong Hwan Kim
- Laboratory of Translational Therapeutics, Korea Research Institute of Chemical Technology, Eulji University, Daejeon 34824,
Korea
| | - Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon 34824,
Korea
| | - Jiyeon Kim
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824,
Korea
| |
Collapse
|
47
|
Traf2- and Nck-interacting kinase (TNIK) is involved in the anti-cancer mechanism of dovitinib in human multiple myeloma IM-9 cells. Amino Acids 2016; 48:1591-9. [PMID: 26995282 DOI: 10.1007/s00726-016-2214-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/09/2016] [Indexed: 01/02/2023]
Abstract
Traf2- and Nck-interacting kinase (TNIK) is a member of the germinal center kinase family. TNIK was first identified as a kinase that is involved in regulating cytoskeletal organization in many types of cells, and it was recently proposed as a novel therapeutic target in several types of human cancers. Although previous studies suggest that TNIK plays a pivotal role in cancer cell survival and prognosis, its function in hematological cancer cell survival has not been investigated. Here we investigated the relationship between TNIK function and cell viability in multiple myeloma IM-9 cells using TNIK small interfering RNA (siRNA) transfection and dovitinib treatment. Treatment of IM-9 cells with TNIK siRNA and dovitinib treatment reduced cell proliferation. The ATP competing kinase assay and western blot analysis showed that dovitinib strongly inhibited both the interaction of TNIK with ATP (K i, 13 nM) and the activation of Wnt signaling effectors such as β-catenin and TCF4. Dovitinib also induced caspase-dependent apoptosis in IM-9 cells without significant cytotoxicity in PBMCs. Our results provide new evidence that TNIK may be involved in the proliferation of multiple myeloma IM-9 cells and in the anti-cancer activity of dovitinib via inhibition of the endogenous Wnt signaling pathway.
Collapse
|
48
|
Porta C, Giglione P, Paglino C. Targeted therapy for renal cell carcinoma: focus on 2nd and 3rd line. Expert Opin Pharmacother 2016; 17:643-55. [PMID: 26630127 DOI: 10.1517/14656566.2016.1127353] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Second- and third-line treatments are more and more frequently administered to metastatic renal cell carcinoma (mRCC) patients. AREAS COVERED Here we discuss the various levels of evidence supporting presently available recommendations, trying to address a number of as yet unanswered issues, and also to take a glowing glance at the future. To do this, we interrogated the Medline database, as well as the proceedings of the main Oncological and Urological conferences for relevant studies. EXPERT OPINION Until recently, with regard to choosing the second line treatment after the failure of therapy with vascular endothelial growth factor receptors-tyrosine kinase inhibitors (VEGFR-TKIs), the continued inhibition of the VEGF/VEGR pathway, or else the switch to an mTOR inhibitor, is recommended. These two options are characterized by partly different targets, completely different toxicity profiles, but a comparable efficacy. This scenario will change soon, after the publication of two randomized, controlled, phase III trials in which cabozantinib and nivolumab proved to be superior as compared to everolimus. As regards third line treatment, where a sequence of two VEGFR-TKIs has been used beforehand, the choice is represented by the mTOR inhibitor everolimus, whilst if a VEGFR-TKI followed by everolimus has been chosen, a return to VEGF pathway inhibition is suggested.
Collapse
Affiliation(s)
- Camillo Porta
- a Medical Oncology , I.R.C.C.S. San Matteo University Hospital Foundation , Pavia , Italy.,b Italian Group of Nephro-Oncology/Gruppo Italiano di Oncologia Nefrologica (G.I.O.N.)
| | - Palma Giglione
- a Medical Oncology , I.R.C.C.S. San Matteo University Hospital Foundation , Pavia , Italy
| | - Chiara Paglino
- a Medical Oncology , I.R.C.C.S. San Matteo University Hospital Foundation , Pavia , Italy.,b Italian Group of Nephro-Oncology/Gruppo Italiano di Oncologia Nefrologica (G.I.O.N.)
| |
Collapse
|
49
|
Lim SM, Chung WY, Nam KH, Kang SW, Lim JY, Kim HG, Shin SH, Sun JM, Kim SG, Kim JH, Kang CW, Kim HR, Cho BC. An open label, multicenter, phase II study of dovitinib in advanced thyroid cancer. Eur J Cancer 2015; 51:1588-95. [DOI: 10.1016/j.ejca.2015.05.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 05/08/2015] [Indexed: 12/13/2022]
|
50
|
de Brot S, Ntekim A, Cardenas R, James V, Allegrucci C, Heery DM, Bates DO, Ødum N, Persson JL, Mongan NP. Regulation of vascular endothelial growth factor in prostate cancer. Endocr Relat Cancer 2015; 22:R107-23. [PMID: 25870249 DOI: 10.1530/erc-15-0123] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is the most common malignancy affecting men in the western world. Although radical prostatectomy and radiation therapy can successfully treat PCa in the majority of patients, up to ~30% will experience local recurrence or metastatic disease. Prostate carcinogenesis and progression is typically an androgen-dependent process. For this reason, therapies for recurrent PCa target androgen biosynthesis and androgen receptor function. Such androgen deprivation therapies (ADT) are effective initially, but the duration of response is typically ≤24 months. Although ADT and taxane-based chemotherapy have delivered survival benefits, metastatic PCa remains incurable. Therefore, it is essential to establish the cellular and molecular mechanisms that enable localized PCas to invade and disseminate. It has long been accepted that metastases require angiogenesis. In the present review, we examine the essential role for angiogenesis in PCa metastases, and we focus in particular on the current understanding of the regulation of vascular endothelial growth factor (VEGF) in localized and metastatic PCa. We highlight recent advances in understanding the role of VEGF in regulating the interaction of cancer cells with tumor-associated immune cells during the metastatic process of PCa. We summarize the established mechanisms of transcriptional and post-transcriptional regulation of VEGF in PCa cells and outline the molecular insights obtained from preclinical animal models of PCa. Finally, we summarize the current state of anti-angiogenesis therapies for PCa and consider how existing therapies impact VEGF signaling.
Collapse
Affiliation(s)
- Simone de Brot
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Atara Ntekim
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Ryan Cardenas
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Victoria James
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Cinzia Allegrucci
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - David M Heery
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - David O Bates
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Niels Ødum
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Jenny L Persson
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| | - Nigel P Mongan
- Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA Faculty of Medicine and Health SciencesSchool of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UKDepartment of PharmacologySchool of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UKCancer BiologyDivision of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UKDepartment of International HealthImmunology and Microbiology, University of Copenhagen, Copenhagen, DenmarkClinical Research CenterLund University, Malmö, SwedenDepartment of PharmacologyWeill Cornell Medical College, New York, New York 10065, USA
| |
Collapse
|