1
|
Giacomini A, Taranto S, Gazzaroli G, Faletti J, Capoferri D, Marcheselli R, Sciumè M, Presta M, Sacco A, Roccaro AM. The FGF/FGFR/c-Myc axis as a promising therapeutic target in multiple myeloma. J Exp Clin Cancer Res 2024; 43:294. [PMID: 39482742 PMCID: PMC11529022 DOI: 10.1186/s13046-024-03217-2] [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: 08/19/2024] [Accepted: 10/26/2024] [Indexed: 11/03/2024] Open
Abstract
Among blood cancers, multiple myeloma (MM) represents the second most common neoplasm and is characterized by the accumulation and proliferation of monoclonal plasma cells within the bone marrow. Despite the last few decades being characterized by the development of different therapeutic strategies against MM, at present such disease is still considered incurable. Although MM is highly heterogeneous in terms of genetic and molecular subtypes, about 67% of MM cases are associated with abnormal activity of the transcription factor c-Myc, which has so far revealed a protein extremely difficult to target. We have recently demonstrated that activation of fibroblast growth factor (FGF) signaling protects MM cells from oxidative stress-induced apoptosis by stabilizing the oncoprotein c-Myc. Accordingly, secretion of FGF ligands and autocrine activation of FGF receptors (FGFR) is observed in MM cells and FGFR3 genomic alterations represent some 15-20% MM cases and are associated with poor outcome. Thus, FGF/FGFR blockade may represent a promising strategy to indirectly target c-Myc in MM. On this basis, the present review aims at providing an overview of recently explored connections between the FGF/FGFR system and c-Myc oncoprotein, sustaining the therapeutic potential of targeting the FGF/FGFR/c-Myc axis in MM by using inhibitors targeting FGF ligands or FGF receptors. Importantly, the provided findings may represent the rationale for using FDA approved FGFR TK inhibitors (i.e. Pemigatinib, Futibatinib, Erdafitinib) for the treatment of MM patients presenting with an aberrant activation of this axis.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Sara Taranto
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Giorgia Gazzaroli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jessica Faletti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Davide Capoferri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Marcheselli
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Margherita Sciumè
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Sacco
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Aldo M Roccaro
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy.
| |
Collapse
|
2
|
Guo YJ, Ma MX, Tian T, Zhang JN, Guo XN, Qiao S. Myeloid/lymphoid neoplasms with eosinophilia and FGFR1 rearrangement t(8;13)(p11;q12): A case report and literature review. Oncol Lett 2024; 28:468. [PMID: 39119236 PMCID: PMC11306990 DOI: 10.3892/ol.2024.14601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
8p11 myeloproliferative syndrome (EMS) is a rare and aggressive hematological malignancy, characterized by myeloproliferative neoplasms, and associated with eosinophilia and T- or B-cell lineage lymphoblastic lymphoma. The pathogenesis is defined by the presence of chromosomal translocations associated with the fibroblast growth factor-1 (FGFR1) gene, located in the 8p11-12.1 chromosomal locus. At present, only ~100 cases have been reported globally. At least 15 partner genes have been identified, including the most common, the zinc finger MYM-type containing 2 (ZNF198)-FGFR1 fusion gene formed by t(8;13)(p11;q12). Different fusion genes determine the clinical manifestations and prognosis of the disease. Patients with EMS with t(8;13)(p11;q12) commonly present with lymphadenopathy and T-lymphoblastic lymphoma, which usually converts to acute myeloid leukemia (AML) with the progression of the disease. The present study describes the case of an elderly female patient with EMS with t(8;13)(p11;q12), presenting with myeloid/lymphoid syndrome (myeloproliferative neoplasms and T lymphoblastic lymphoma). The patient received the CHOPE regimen combined with tyrosine kinase inhibitor (dasatin) treatment and obtained short-term complete remission. However, 6 months later, the disease progressed from EMS to AML and the patient died due to ineffective induction therapy. The present study also reviews the relevant literature about this unusual entity to enhance the understanding of EMS.
Collapse
Affiliation(s)
- Yu-Jie Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Meng-Xue Ma
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Tian Tian
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jing-Nan Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiao-Nan Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shukai Qiao
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
3
|
Wu CH, Weng TF, Li JP, Wu KH. Biology and Therapeutic Properties of Mesenchymal Stem Cells in Leukemia. Int J Mol Sci 2024; 25:2527. [PMID: 38473775 DOI: 10.3390/ijms25052527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
This comprehensive review delves into the multifaceted roles of mesenchymal stem cells (MSCs) in leukemia, focusing on their interactions within the bone marrow microenvironment and their impact on leukemia pathogenesis, progression, and treatment resistance. MSCs, characterized by their ability to differentiate into various cell types and modulate the immune system, are integral to the BM niche, influencing hematopoietic stem cell maintenance and functionality. This review extensively explores the intricate relationship between MSCs and leukemic cells in acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, and chronic lymphocytic leukemia. This review also addresses the potential clinical applications of MSCs in leukemia treatment. MSCs' role in hematopoietic stem cell transplantation, their antitumor effects, and strategies to disrupt chemo-resistance are discussed. Despite their therapeutic potential, the dual nature of MSCs in promoting and inhibiting tumor growth poses significant challenges. Further research is needed to understand MSCs' biological mechanisms in hematologic malignancies and develop targeted therapeutic strategies. This in-depth exploration of MSCs in leukemia provides crucial insights for advancing treatment modalities and improving patient outcomes in hematologic malignancies.
Collapse
Affiliation(s)
- Cheng-Hsien Wu
- School of Medicine, National Defense Medical Center, Taipei 114, Taiwan
| | - Te-Fu Weng
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Ju-Pi Li
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| |
Collapse
|
4
|
Mann JE, Smith JD, Kulkarni A, Foltin SK, Scheftz EB, Murray IR, Gensterblum-Miller E, Brummel CV, Bhangale A, Hoesli RC, Brenner JC. Genome-wide open reading frame profiling identifies fibroblast growth factor signaling as a driver of PD-L1 expression in head and neck squamous cell carcinoma. Oral Oncol 2023; 146:106562. [PMID: 37666053 PMCID: PMC11308298 DOI: 10.1016/j.oraloncology.2023.106562] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCC) are associated with significant treatment-related morbidity and poor disease-free and disease-specific survival, especially in the recurrent and metastatic (R/M HNSCC) setting. Inhibition of the programmed death-1/ligand-1 (PD-1/PD-L1) immune checkpoint is accepted as a first-line treatment strategy for R/M HNSCC and has expanded into the neoadjuvant, definitive, and adjuvant settings. To understand cellular signals modulating the PD-L1 in HNSCC, we profiled a HNSCC cell-line with a genome-wide open reading frame (ORF) library of 17,000 individual constructs (14,000 unique genes). We identified 335 ORFs enriched in PD-L1high cells and independently validated five of these ORFs (FGF6, IL17A, CD300C, KLR1C and NFKBIA) as drivers of PD-L1 upregulation. We showed that exogenous FGF ligand is sufficient to induce PD-L1 expression in multiple HNSCC cell lines and human immature dendritic cells. Accordingly, overexpression of FGFR1, FGFR3 or the FGFR3 S249C and D786N mutants common to HNSCC tumors also induced PD-L1 overexpression on tumor cells. Small molecule inhibition of FGF signaling abrogated PD-L1 upregulation in these models and also blocked "classical" IFNγ-regulated PD-L1 expression in a STAT1-independent manner. Finally, we found that FGF specifically upregulated a glycosylated form of PD-L1 in our study, and exogenous FGF led to concomitant upregulation of glycosyltransferases that may stabilize PD-L1 on the surface of HNSCC cells. Taken together, our study supports a potential role for FGF/FGFR pathway signaling as a mechanism driving immune escape and rationalizes further exploration of novel combination therapies to improve clinical responses to PD-1/PD-L1 axis inhibition in HNSCC.
Collapse
Affiliation(s)
- Jacqueline E Mann
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pathology, University of Michigan, Ann Arbor, MI 41809, USA
| | - Joshua D Smith
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aditi Kulkarni
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Susan K Foltin
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erin B Scheftz
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Isabel R Murray
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth Gensterblum-Miller
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA; Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 41809, USA
| | - Collin V Brummel
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Apurva Bhangale
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rebecca C Hoesli
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - J Chad Brenner
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA; Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 41809, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Freyer CW, Hughes ME, Carulli A, Bagg A, Hexner E. Pemigatinib for the treatment of myeloid/lymphoid neoplasms with FGFR1 rearrangement. Expert Rev Anticancer Ther 2023; 23:351-359. [PMID: 36927350 DOI: 10.1080/14737140.2023.2192930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
INTRODUCTION Myeloid/lymphoid neoplasms with fibroblast growth factor receptor-1 (FGFR1) rearrangements (MLNFGFR1) are rare entities with aggressive features and poor prognosis. Presentation is heterogeneous, ranging from myeloproliferative neoplasms (with or without eosinophilia) to T-cell lymphoma and acute leukemia. Historical treatments have been guided by the presenting phenotype with induction chemotherapy frequently used. Pemigatinib is a FGFR1-3 tyrosine kinase inhibitor that has demonstrated high complete hematologic and cytogenetic response rates in MLNFGFR1. AREAS COVERED We discuss the pathogenesis, presentation, and historical treatments for MLNFGFR1, in addition to clinical data using pemigatinib and other targeted therapies. Discussion of the mechanism of action and adverse events are also included. EXPERT OPINION Pemigatinib represents a significant advance in the management of MLNFGFR1. High rates of complete hematologic and cytogenetic response have been observed. While direct comparative data are unavailable, outcomes appear favorable compared to conventional approaches. Long term efficacy and tolerability are not yet known, and allogeneic hematopoietic stem cell transplant (alloHSCT) has been and continues to be the treatment with the highest chance of long term disease free survival in responding patients. Combinations of pemigatinib and chemotherapy, particularly for more aggressive phenotypes, warrant future investigation as does the use of pemigatinib maintenance following alloHSCT.
Collapse
Affiliation(s)
- Craig W Freyer
- Department of Pharmacy Services, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.,Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell E Hughes
- Department of Pharmacy Services, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.,Lymphoma Program, Hematology/Oncology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alison Carulli
- Department of Pharmacy Services, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Hexner
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
6
|
Tian C, Li Y, Wang L, Si J, Zheng Y, Kang J, Wang Y, You MJ, Zheng G. Blockade of FGF2/FGFR2 partially overcomes bone marrow mesenchymal stromal cells mediated progression of T-cell acute lymphoblastic leukaemia. Cell Death Dis 2022; 13:922. [PMID: 36333298 PMCID: PMC9636388 DOI: 10.1038/s41419-022-05377-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
The development of acute lymphoblastic leuakemia (ALL) is partly attributed to the effects of bone marrow (BM) microenvironment, especially mesenchymal stromal cells (MSCs), which interact bilaterally with leukaemia cells, leading to ALL progression. In order to find MSCs-based microenvironment targeted therapeutic strategies, Notch1-induced T-cell ALL (T-ALL) mice models were used and dynamic alterations of BM-MSCs with increased cell viability during T-ALL development was observed. In T-ALL mice derived stroma-based condition, leukaemia cells showed significantly elevated growth capacity indicating that MSCs participated in leukaemic niche formation. RNA sequence results revealed that T-ALL derived MSCs secreted fibroblast growth factor 2 (FGF2), which combined with fibroblast growth factor receptor 2 (FGFR2) on leukaemia cells, resulting in activation of PI3K/AKT/mTOR signalling pathway in leukaemia cells. In vitro blocking the interaction between FGF2 and FGFR2 with BGJ398 (infigratinib), a FGFR1-3 kinase inhibitor, or knockdown FGF2 in MSCs by interference caused deactivation of PI3K/AKT/mTOR pathway and dysregulations of genes associated with cell cycle and apoptosis in ALL cells, leading to decrease of leukaemia cells. In mouse model received BGJ398, overall survival was extended and dissemination of leukaemia cells in BM, spleen, liver and peripheral blood was decreased. After subcutaneous injection of primary human T-ALL cells with MSCs, tumour growth was suppressed when FGF2/FGFR2 was interrupted. Thus, inhibition of FGF2/FGFR2 interaction appears to be a valid strategy to overcome BM-MSCs mediated progression of T-ALL, and BGJ398 could indeed improve outcomes in T-ALL, which provide theoretical basis of BGJ398 as a BM microenvironment based therapeutic strategy to control disease progression.
Collapse
Affiliation(s)
- Chen Tian
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Yueyang Li
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| | - Lina Wang
- grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| | - Junqi Si
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Yaxin Zheng
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Junnan Kang
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| | - Yafei Wang
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - M. James You
- grid.240145.60000 0001 2291 4776Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77479 USA
| | - Guoguang Zheng
- grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| |
Collapse
|
7
|
Cai B, Liu Y, Chong Y, Mori SF, Matsunaga A, Zhang H, Fang X, Chang CS, Cowell JK, Hu T. A truncated derivative of FGFR1 kinase cooperates with FLT3 and KIT to transform hematopoietic stem cells in syndromic and de novo AML. Mol Cancer 2022; 21:156. [PMID: 35906694 PMCID: PMC9336057 DOI: 10.1186/s12943-022-01628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background Myeloid and lymphoid malignancies associated with chimeric FGFR1 kinases are the hallmark of stem cell leukemia and lymphoma syndrome (SCLL). In all cases, FGFR1 kinase is constitutively phosphoactivated as a result of chromosome translocations, which lead to acquisition of dimerization motifs in the chimeric proteins. Recently, we demonstrated that these chimeric kinases could be cleaved by granzyme B to generate a truncated derivative, tnFGFR1, which localized exclusively into the nucleus and was not phosphorylated. Methods Stem cell transduction and transplantation in syngeneic mice was used to assess the transforming ability of tnFGFR1 in bone marrow stem cells, and RPPA and RNA-Seq was used to examine the related signaling pathways and regulated target genes. Results For the first time, we show that this non-classical truncated form of FGFR1 can independently lead to oncogenic transformation of hematopoietic stem cells in an animal model in vivo. These leukemia cells show a mixed immunophenotype with a B-cell B220 + Igm- profile in the majority of cells and Kit+ in virtually all cells, suggesting a stem cell disease. tnFGFR1, however, does not activate classic FGFR1 downstream signaling pathways but induces a distinct profile of altered gene expression with significant upregulation of transmembrane signaling receptors including FLT3 and KIT. We further show that de novo human AML also express tnFGFR1 which correlates with upregulation of FLT3 and KIT as in mouse leukemia cells. ChIP analysis demonstrates tnFGFR1 occupancy at the Flt3 and Kit promoters, suggesting a direct transcriptional regulation. Cells transformed with tnFGFR1 are insensitive to FGFR1 inhibitors but treatment of these cells with the Quizartinib (AC220) FLT3 inhibitor, suppresses in vitro growth and development of leukemia in vivo. Combined treatment with FGFR1 and FLT3 inhibitors provides increased survival compared to FGFR1 inhibition alone. Conclusions This study demonstrates a novel model for transformation of hematopoietic stem cells by chimeric FGFR1 kinases with the combined effects of direct protein activation by the full-length kinases and transcriptional regulation by the truncated nuclear tnFGFR1 derivative, which is associated with GZMB expression levels. Genes significantly upregulated by tnFGFR1 include Flt3 and Kit which promote a leukemia stem cell phenotype. In human AML, tnFGFR1 activation leads to increased FLT3 and KIT expression, and higher FLT3 and GZMB expression levels are associated with an inferior prognosis. These observations provide insights into the relative therapeutic value of targeting FGFR1 and FLT3 in treating AML with this characteristic gene expression profile. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-022-01628-3.
Collapse
Affiliation(s)
- Baohuan Cai
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA.,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Liu
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA.,Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yating Chong
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Stephanie Fay Mori
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Atsuko Matsunaga
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Hualei Zhang
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA.,Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xuexiu Fang
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Chang-Sheng Chang
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - John K Cowell
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Tianxiang Hu
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
8
|
Zhang H, Cai B, Liu Y, Chong Y, Matsunaga A, Mori SF, Fang X, Kitamura E, Chang CS, Wang P, Cowell JK, Hu T. RHOA-regulated IGFBP2 promotes invasion and drives progression of BCR-ABL1 chronic myeloid leukemia. Haematologica 2022; 108:122-134. [PMID: 35833297 PMCID: PMC9827165 DOI: 10.3324/haematol.2022.280757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Indexed: 02/05/2023] Open
Abstract
The Philadelphia 9;22 chromosome translocation has two common isoforms that are preferentially associated with distinct subtypes of leukemia. The p210 variant is the hallmark of chronic myeloid leukemia (CML) whereas p190 is frequently associated with B-cell acute lymphoblastic leukemia. The only sequence difference between the two isoforms is the guanidine exchange factor domain. This guanidine exchange factor is reported to activate RHO family GTPases in response to diverse extracellular stimuli. It is not clear whether and, if so, how RHOA contributes to progression of p210 CML. Here we show that knockout of RHOA in the K562 and KU812, p210-expressing cell lines leads to suppression of leukemogenesis in animal models in vivo. RNA-sequencing analysis of the mock control and null cells demonstrated a distinct change in the gene expression profile as a result of RHOA deletion, with significant downregulation of genes involved in cell activation and cell adhesion. Cellular analysis revealed that RHOA knockout leads to impaired cell adhesion and migration and, most importantly, the homing ability of leukemia cells to the bone marrow, which may be responsible for the attenuated leukemia progression. We also identified IGFBP2 as an important downstream target of RHOA. Further mechanistic investigation showed that RHOA activation leads to relocation of the serum response factor (SRF) into the nucleus, where it directly activates IGFBP2. Knockout of IGFBP2 in CML cells suppressed cell adhesion/invasion, as well as leukemogenesis in vivo. This elevated IGFBP2 expression was confirmed in primary CML samples. Thus, we demonstrate one mechanism whereby the RHOA-SRF-IGFBP2 signaling axis contributes to the development of leukemia in cells expressing the p210 BCR-ABL1 fusion kinase.
Collapse
Affiliation(s)
- Hualei Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Baohuan Cai
- Georgia Cancer Center, Augusta University, Augusta, GA, USA,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Liu
- Georgia Cancer Center, Augusta University, Augusta, GA, USA,Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yating Chong
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | | | | | - Xuexiu Fang
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Eiko Kitamura
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | | | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - John K. Cowell
- Georgia Cancer Center, Augusta University, Augusta, GA, USA,J. K. Cowell
| | - Tianxiang Hu
- Georgia Cancer Center, Augusta University, Augusta, GA, USA,T. Hu
| |
Collapse
|
9
|
Proteomic analysis reveals dual requirement for Grb2 and PLCγ1 interactions for BCR-FGFR1-Driven 8p11 cell proliferation. Oncotarget 2022; 13:659-676. [PMID: 35574218 PMCID: PMC9093983 DOI: 10.18632/oncotarget.28228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/19/2022] [Indexed: 12/03/2022] Open
Abstract
Translocation of Fibroblast Growth Factor Receptors (FGFRs) often leads to aberrant cell proliferation and cancer. The BCR-FGFR1 fusion protein, created by chromosomal translocation t(8;22)(p11;q11), contains Breakpoint Cluster Region (BCR) joined to Fibroblast Growth Factor Receptor 1 (FGFR1). BCR-FGFR1 represents a significant driver of 8p11 myeloproliferative syndrome, or stem cell leukemia/lymphoma, which progresses to acute myeloid leukemia or T-cell lymphoblastic leukemia/lymphoma. Mutations were introduced at Y177F, the binding site for adapter protein Grb2 within BCR; and at Y766F, the binding site for the membrane associated enzyme PLCγ1 within FGFR1. We examined anchorage-independent cell growth, overall cell proliferation using hematopoietic cells, and activation of downstream signaling pathways. BCR-FGFR1-induced changes in protein phosphorylation, binding partners, and signaling pathways were dissected using quantitative proteomics to interrogate the protein interactome, the phosphoproteome, and the interactome of BCR-FGFR1. The effects on BCR-FGFR1-stimulated cell proliferation were examined using the PLCγ1 inhibitor U73122, and the irreversible FGFR inhibitor futibatinib (TAS-120), both of which demonstrated efficacy. An absolute requirement is demonstrated for the dual binding partners Grb2 and PLCγ1 in BCR-FGFR1-driven cell proliferation, and new proteins such as ECSIT, USP15, GPR89, GAB1, and PTPN11 are identified as key effectors for hematopoietic transformation by BCR-FGFR1.
Collapse
|
10
|
Zhang X, Wang F, Yan F, Huang D, Wang H, Gao B, Gao Y, Hou Z, Lou J, Li W, Yan J. Identification of a novel HOOK3-FGFR1 fusion gene involved in activation of the NF-kappaB pathway. Cancer Cell Int 2022; 22:40. [PMID: 35081975 PMCID: PMC8793161 DOI: 10.1186/s12935-022-02451-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/31/2021] [Indexed: 12/15/2022] Open
Abstract
Background Rearrangements involving the fibroblast growth factor receptor 1 (FGFR1) gene result in 8p11 myeloproliferative syndrome (EMS), which is a rare and aggressive hematological malignancy that is often initially diagnosed as myelodysplastic syndrome (MDS). Clinical outcomes are typically poor due to relative resistance to tyrosine kinase inhibitors (TKIs) and rapid transformation to acute leukemia. Deciphering the transcriptomic signature of FGFR1 fusions may open new treatment strategies for FGFR1 rearrangement patients. Methods DNA sequencing (DNA-seq) was performed for 20 MDS patients and whole exome sequencing (WES) was performed for one HOOK3-FGFR1 fusion positive patient. RNA sequencing (RNA-seq) was performed for 20 MDS patients and 8 healthy donors. Fusion genes were detected using the STAR-Fusion tool. Fluorescence in situ hybridization (FISH), quantitative real-time PCR (qRT-PCR), and Sanger sequencing were used to confirm the HOOK3-FGFR1 fusion gene. The phosphorylation antibody array was performed to validate the activation of nuclear factor-kappaB (NF-kappaB) signaling. Results We identified frequently recurrent mutations of ASXL1 and U2AF1 in the MDS cohort, which is consistent with previous reports. We also identified a novel in-frame HOOK3-FGFR1 fusion gene in one MDS case with abnormal monoclonal B-cell lymphocytosis and ring chromosome 8. FISH analysis detected the FGFR1 break-apart signal in myeloid blasts only. qRT-PCR and Sanger sequencing confirmed the HOOK3-FGFR1 fusion transcript with breakpoints located at the 11th exon of HOOK3 and 10th exon of FGFR1, and Western blot detected the chimeric HOOK3-FGFR1 fusion protein that is presumed to retain the entire tyrosine kinase domain of FGFR1. The transcriptional feature of HOOK3-FGFR1 fusion was characterized by the significant enrichment of the NF-kappaB pathway by comparing the expression profiling of FGFR1 fusion positive MDS with 8 healthy donors and FGFR1 fusion negative MDS patients. Further validation by phosphorylation antibody array also showed NF-kappaB activation, as evidenced by increased phosphorylation of p65 (Ser 536) and of IKBalpha (Ser 32). Conclusions The HOOK3-FGFR1 fusion gene may contribute to the pathogenesis of MDS and activate the NF-kappaB pathway. These findings highlight a potential novel approach for combination therapy for FGFR1 rearrangement patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02451-y.
Collapse
Affiliation(s)
- Xuehong Zhang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China
| | - Furong Wang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China
| | - Fanzhi Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China
| | - Dan Huang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China
| | - Haina Wang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China
| | - Beibei Gao
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China
| | - Yuan Gao
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China
| | - Zhijie Hou
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China
| | - Jiacheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, 116044, Dalian, China
| | - Weiling Li
- Department of Biotechnology College of Basic Medical Science, Dalian Medical University, 116044, Dalian, China.
| | - Jinsong Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem-Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem-Cell Transplantation and Translational Medicine, Dalian Key Laboratory of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China. .,Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, 116027, Dalian, China.
| |
Collapse
|
11
|
Cowell JK, Hu T. Mechanisms of resistance to FGFR1 inhibitors in FGFR1-driven leukemias and lymphomas: implications for optimized treatment. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:607-619. [PMID: 34734169 PMCID: PMC8562765 DOI: 10.20517/cdr.2021.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myeloid and lymphoid neoplasms with eosinophilia and FGFR1 rearrangements (MLN-eo FGFR1) disease is derived from a pluripotent hematopoietic stem cell and has a complex presentation with a myeloproliferative disorder with or without eosinophilia and frequently presents with mixed lineage T- or B-lymphomas. The myeloproliferative disease frequently progresses to AML and lymphoid neoplasms can develop into acute lymphomas. No matter the cell type involved, or clinical presentation, chromosome translocations involving the FGFR1 kinase and various partner genes, which leads to constitutive activation of downstream oncogenic signaling cascades. These patients are not responsive to treatment regimens developed for other acute leukemias and survival is poor. Recent development of specific FGFR1 inhibitors has suggested an alternative therapeutic approach but resistance is likely to evolve over time. Mouse models of this disease syndrome have been developed and are being used for preclinical evaluation of FGFR1 inhibitors. Cell lines from these models have now been developed and have been used to investigate the mechanisms of resistance that might be expected in clinical cases. So far, a V561M mutation in the kinases domain and deletion of PTEN have been recognized as leading to resistance and both operate through the PI3K/AKT signaling axis. One of the important consequences is the suppression of PUMA, a potent enforcer of apoptosis, which operates through BCL2. Targeting BCL2 in the resistant cells leads to suppression of leukemia development in mouse models, which potentially provides an opportunity to treat patients that become resistant to FGFR1 inhibitors. In addition, elucidation of molecular mechanisms underlying FGFR1-driven leukemias and lymphomas also provides new targets for combined treatment as another option to bypass the FGFR1 inhibitor resistance and improve patient outcome.
Collapse
Affiliation(s)
- John K Cowell
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd, Augusta, GA 30912, USA
| |
Collapse
|
12
|
Lee J, Ju KD, Kim HJ, Tsogbadrakh B, Ryu H, Kang E, Kang M, Yang J, Kang HG, Ahn C, Oh KH. Soluble α-klotho anchors TRPV5 to the distal tubular cell membrane independent of FGFR1 by binding TRPV5 and galectin-1 simultaneously. Am J Physiol Renal Physiol 2021; 320:F559-F568. [PMID: 33615893 DOI: 10.1152/ajprenal.00044.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hypercalciuria is one of the early manifestations of diabetic nephropathy (DN). This is partially due to a decrease in the expression of renal transient receptor potential vanilloid type 5 (TRPV5), which is responsible for renal Ca2+ reabsorption. Soluble klotho has been previously determined to increase TRPV5 by cleaving sialic acid, causing TRPV5 to bind to membrane protein galectin-1. However, a recent study showed that soluble klotho binds to α2-3-sialyllactose, where sialic acid is located, on TRPV5, rather than cleave it. Here, we report that soluble klotho tethers TRPV5 on the membrane by binding both TRPV5 and galectin-1, thereby protecting membrane TRPV5 from diabetes-induced endocytosis. In the present study, we injected recombinant soluble α-klotho protein (rKL) into db/db and db/m mice for 8 wk and collected urine and kidneys. We administered rKL, AZD4547 [fibroblast growth factor (FGF) receptor type 1 inhibitor], and OTX008 (galectin-1 inhibitor) to cultured mouse distal tubular cells with or without 30 mM high-glucose (HG) exposure. db/db mice showed increased renal Ca2+ excretion and decreased renal TRPV5 expression. rKL treatment reversed this change. In vitro, TRPV5 expression in distal tubular cells decreased under HG conditions, and rKL successfully upregulated TRPV5 with or without FGF23. Also, immunofluorescence showed colocalization of klotho, TRPV5, and galectin-1 in distal tubule cells, suggesting that klotho binds to both TRPV5 and galectin-1. Moreover, when both FGF receptor type 1 and galectin-1 were inhibited, rKL failed to increase TRPV5 under HG conditions. Our results indicate that soluble klotho prevents TRPV5 from degradation and subsequent diabetes-induced endocytosis by anchoring TRPV5 through binding with both TRPV5 and galectin-1.NEW & NOTEWORTHY Soluble α-klotho anchors transient receptor potential vanilloid type 5 (TRPV5) on the apical membrane of the distal tubule by binding both TRPV5 and a membrane-abundant protein, galectin-1. This newly discovered mechanism works even when fibroblast growth factor (FGF)23 signaling is inhibited by treatment with FGF receptor type 1 inhibitor. Therefore, we identified how soluble α-klotho increases TRPV5 without FGF23. We confirmed this mechanism by observing that soluble α-klotho fails to enhance TRPV5 when both FGF receptor type 1 and galectin-1 are inhibited.
Collapse
Affiliation(s)
- Jinho Lee
- Center of Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Kyung Don Ju
- Center of Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Hyo Jin Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | | | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eunjeong Kang
- Department of Internal Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Minjung Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jaeseok Yang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hee Gyung Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.,Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea.,Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Liu Y, Cai B, Chong Y, Zhang H, Kemp CA, Lu S, Chang CS, Ren M, Cowell JK, Hu T. Downregulation of PUMA underlies resistance to FGFR1 inhibitors in the stem cell leukemia/lymphoma syndrome. Cell Death Dis 2020; 11:884. [PMID: 33082322 PMCID: PMC7576156 DOI: 10.1038/s41419-020-03098-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/18/2022]
Abstract
Resistance to molecular therapies frequently occur due to genetic changes affecting the targeted pathway. In myeloid and lymphoid leukemias/lymphomas resulting from constitutive activation of FGFR1 kinases, resistance has been shown to be due either to mutations in FGFR1 or deletions of PTEN. RNA-Seq analysis of the resistant clones demonstrates expression changes in cell death pathways centering on the p53 upregulated modulator of apoptosis (Puma) protein. Treatment with different tyrosine kinase inhibitors (TKIs) revealed that, in both FGFR1 mutation and Pten deletion-mediated resistance, sustained Akt activation in resistant cells leads to compromised Puma activation, resulting in suppression of TKI-induced apoptosis. This suppression of Puma is achieved as a result of sequestration of inactivated p-Foxo3a in the cytoplasm. CRISPR/Cas9 mediated knockout of Puma in leukemic cells led to an increased drug resistance in the knockout cells demonstrating a direct role in TKI resistance. Since Puma promotes cell death by targeting Bcl2, TKI-resistant cells showed high Bcl2 levels and targeting Bcl2 with Venetoclax (ABT199) led to increased apoptosis in these cells. In vivo treatment of mice xenografted with resistant cells using ABT199 suppressed leukemogenesis and led to prolonged survival. This in-depth survey of the underlying genetic mechanisms of resistance has identified a potential means of treating FGFR1-driven malignancies that are resistant to FGFR1 inhibitors.
Collapse
Affiliation(s)
- Yun Liu
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA.,Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baohuan Cai
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA.,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yating Chong
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Hualei Zhang
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA.,Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chesley-Anne Kemp
- College of Allied Health Sciences, Augusta University, Augusta, GA, 30912, USA
| | - Sumin Lu
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | | | - Mingqiang Ren
- Consortium for Health and Military Performance (CHAMP), Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - John K Cowell
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Tianxiang Hu
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
14
|
Qin J, Xie F, Wang F, Lu H. mRNA Expression of FGFR1 as Potential Marker for Predicting Prognosis of Surgical Resection of Small Cell Lung Cancer may be better than Protein Expression and Gene Amplification. J Cancer 2020; 11:4691-4699. [PMID: 32626515 PMCID: PMC7330682 DOI: 10.7150/jca.44476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/12/2020] [Indexed: 11/10/2022] Open
Abstract
Purpose: Fibroblast growth factor receptor 1 (FGFR1) alterations have been described in many cancers, including lung cancer, but the role has not been elucidated specifically in small cell lung cancer (SCLC). The present study aimed to identify the frequency of FGFR1 alterations among Chinese patients with surgically resected SCLC and the association with the clinicopathological characteristics and the survival were also investigated. Methods: FGFR1 protein expression, FGFR1 amplification, FGFR1 mutations, and messenger RNA (mRNA) levels, were determined by immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), polymerase chain reaction (PCR) and reverse transcription-polymerase chain reaction (RT-PCR), respectively in primary tumors from 33 patients with resected SCLC. Results: 7/33(21.2%) of the specimens were positive for FGFR1 protein expression. FGFR1 amplification was identified in 4/28 cases (14.3%). If the cut-off value was determined to be 3.5, FGFR1 mRNA positivity was considered in 7/33 cases (21.2%). However, no mutation was detected in the 33 SCLC postoperative tissue specimens. No significant association was observed between FGFR1 protein expression or amplification and clinicalcharacteristics or prognosis. There was a distinct trend for mRNA level and poor prognosis, including recurrence-free survival (RFS) (p = 0.07) and overall survival (OS) (p= 0.08), but they did not reach statistical significance. Conclusions: As novel FGFR1-targeted therapies are developed, FISH, IHC, especially mRNA were detected, which should be considered as biomarkers of FGFR1 pathway dysregulation in SCLC.
Collapse
Affiliation(s)
- Jing Qin
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (lung and esophagus), Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, 310022, P.R. China.,Department of Thoracic Medical Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, 310022, P.R. China
| | - Fajun Xie
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (lung and esophagus), Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, 310022, P.R. China.,Department of Thoracic Medical Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, 310022, P.R. China
| | - Fenfang Wang
- Graduate School, WenZhou Medical University, Wenzhou, 325035, P.R. China
| | - Hongyang Lu
- Graduate School, WenZhou Medical University, Wenzhou, 325035, P.R. China.,Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (lung and esophagus), Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, 310022, P.R. China.,Department of Thoracic Medical Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, 310022, P.R. China
| |
Collapse
|
15
|
Ronca R, Ghedini GC, Maccarinelli F, Sacco A, Locatelli SL, Foglio E, Taranto S, Grillo E, Matarazzo S, Castelli R, Paganini G, Desantis V, Cattane N, Cattaneo A, Mor M, Carlo-Stella C, Belotti A, Roccaro AM, Presta M, Giacomini A. FGF Trapping Inhibits Multiple Myeloma Growth through c-Myc Degradation-Induced Mitochondrial Oxidative Stress. Cancer Res 2020; 80:2340-2354. [PMID: 32094301 DOI: 10.1158/0008-5472.can-19-2714] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/10/2019] [Accepted: 02/14/2020] [Indexed: 11/16/2022]
Abstract
Multiple myeloma, the second most common hematologic malignancy, frequently relapses because of chemotherapeutic resistance. Fibroblast growth factors (FGF) act as proangiogenic and mitogenic cytokines in multiple myeloma. Here, we demonstrate that the autocrine FGF/FGFR axis is essential for multiple myeloma cell survival and progression by protecting multiple myeloma cells from oxidative stress-induced apoptosis. In keeping with the hypothesis that the intracellular redox status can be a target for cancer therapy, FGF/FGFR blockade by FGF trapping or tyrosine kinase inhibitor impaired the growth and dissemination of multiple myeloma cells by inducing mitochondrial oxidative stress, DNA damage, and apoptotic cell death that were prevented by the antioxidant vitamin E or mitochondrial catalase overexpression. In addition, mitochondrial oxidative stress occurred as a consequence of proteasomal degradation of the c-Myc oncoprotein that led to glutathione depletion. Accordingly, expression of a proteasome-nondegradable c-Myc protein mutant was sufficient to avoid glutathione depletion and rescue the proapoptotic effects due to FGF blockade. These findings were confirmed on bortezomib-resistant multiple myeloma cells as well as on bone marrow-derived primary multiple myeloma cells from newly diagnosed and relapsed/refractory patients, including plasma cells bearing the t(4;14) translocation obtained from patients with high-risk multiple myeloma. Altogether, these findings dissect the mechanism by which the FGF/FGFR system plays a nonredundant role in multiple myeloma cell survival and disease progression, and indicate that FGF targeting may represent a therapeutic approach for patients with multiple myeloma with poor prognosis and advanced disease stage. SIGNIFICANCE: This study provides new insights into the mechanisms by which FGF antagonists promote multiple myeloma cell death. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/11/2340/F1.large.jpg.
Collapse
Affiliation(s)
- Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia C Ghedini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Sacco
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Silvia L Locatelli
- Department of Oncology and Hematology, Humanitas Clinical and Research Center - IRCCS and Humanitas University, Milan, Italy
| | - Eleonora Foglio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Taranto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Matarazzo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Giuseppe Paganini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Azienda Ospedaliera Consorziale Universitaria Policlinico di Bari, Bari, Italy
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Marco Mor
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Carmelo Carlo-Stella
- Department of Oncology and Hematology, Humanitas Clinical and Research Center - IRCCS and Humanitas University, Milan, Italy
| | - Angelo Belotti
- Department of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Aldo M Roccaro
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
16
|
Hu T, Chong Y, Lu S, McGuinness M, Williams DA, Cowell JK. Rac1/2 activation promotes FGFR1 driven leukemogenesis in stem cell leukemia/lymphoma syndrome. Haematologica 2020; 105:e68-e71. [PMID: 31221776 PMCID: PMC7012474 DOI: 10.3324/haematol.2018.208058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Tianxiang Hu
- Georgia Cancer Center, 1410 Laney Walker Blvd., Augusta GA
| | - Yating Chong
- Georgia Cancer Center, 1410 Laney Walker Blvd., Augusta GA
| | - Sumin Lu
- Georgia Cancer Center, 1410 Laney Walker Blvd., Augusta GA
| | - Meaghan McGuinness
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David A Williams
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - John K Cowell
- Georgia Cancer Center, 1410 Laney Walker Blvd., Augusta GA
| |
Collapse
|
17
|
Wang T, Wang Z, Zhang L, Wen L, Cai W, Yang X, Pan J, Ruan C, Wu D, Sun A, Chen S. Identification of a novel
TFG–FGFR1
fusion gene in an acute myeloid leukaemia patient with t(3;8)(q12;p11). Br J Haematol 2019; 188:177-181. [PMID: 31802477 DOI: 10.1111/bjh.16294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Tingjing Wang
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Zheng Wang
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Ling Zhang
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Lijun Wen
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Wenzhi Cai
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Xiaofei Yang
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Jinlan Pan
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Changgeng Ruan
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Depei Wu
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
- Collaborative Innovation Center of Hematology Institute of Blood and Marrow Transplantation Soochow University Suzhou P.R. China
| | - Aining Sun
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
| | - Suning Chen
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health Jiangsu Institute of Hematology the First Affiliated Hospital of Soochow University Soochow University Suzhou P.R. China
- Collaborative Innovation Center of Hematology Institute of Blood and Marrow Transplantation Soochow University Suzhou P.R. China
| |
Collapse
|
18
|
Hu T, Chong Y, Cai B, Liu Y, Lu S, Cowell JK. DNA methyltransferase 1-mediated CpG methylation of the miR-150-5p promoter contributes to fibroblast growth factor receptor 1-driven leukemogenesis. J Biol Chem 2019; 294:18122-18130. [PMID: 31628193 DOI: 10.1074/jbc.ra119.010144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/09/2019] [Indexed: 01/10/2023] Open
Abstract
MicroRNA-150-5p (miR-150-5p) plays a complex role in normal early hematopoietic development and is also implicated in the development of various different leukemias. We have reported previously that, in myeloid and lymphoid malignancies associated with dysregulated fibroblast growth factor receptor 1 (FGFR1) activities, miR-150-5p is down-regulated compared with healthy cells. Here, using murine cells, we found that this down-regulation is accompanied by CpG methylation of the miR-150-5p promoter region. Of note, analysis of human acute lymphoblastic leukemia (ALL) cohorts also revealed an inverse relationship between miR-150-5p expression and disease progression. We also found that the DNA methyltransferase 1 (DNMT1) enzyme is highly up-regulated in FGFR1-driven leukemias and lymphomas and that FGFR1 inhibition reduces DNMT1 expression. DNMT1 knockdown in stem cell leukemia/lymphoma (SCLL) cells increased miR-150-5p levels and reduced levels of the MYB proto-oncogene transcription factor, a key regulator of leukemogenesis. FGFR1 directly activates the MYC proto-oncogene basic helix-loop-helix transcription factor, which, as we show here, binds and activates the DNMT1 promoter. MYC knockdown decreased DNMT1 expression, which, in turn, increased miR-150-5p expression. One of the known targets of miR-150-5p is MYB, and treatment of leukemic cells with the MYB inhibitor mebendazole dose-dependently increased apoptosis and reduced cell viability. Moreover, mebendazole treatment of murine xenografts models of FGFR1-driven leukemias enhanced survival. These findings provide evidence that MYC activates MYB by up-regulating DNMT1, which silences miR-150-5p and promotes SCLL progression. We propose that inclusion of mebendazole in a combination therapy with FGFR1 inhibitors may be a valuable option to manage SCLL.
Collapse
Affiliation(s)
- Tianxiang Hu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912
| | - Yating Chong
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912
| | - Baohuan Cai
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912; Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun Liu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912; Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sumin Lu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912
| | - John K Cowell
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912.
| |
Collapse
|
19
|
Chong Y, Liu Y, Lu S, Cai B, Qin H, Chang CS, Ren M, Cowell JK, Hu T. Critical individual roles of the BCR and FGFR1 kinase domains in BCR-FGFR1-driven stem cell leukemia/lymphoma syndrome. Int J Cancer 2019; 146:2243-2254. [PMID: 31525277 DOI: 10.1002/ijc.32665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/14/2019] [Accepted: 08/29/2019] [Indexed: 01/09/2023]
Abstract
Constitutive activation of FGFR1, as a result of diverse chromosome translocations, is the hallmark of stem cell leukemia/lymphoma syndrome. The BCR-FGFR1 variant is unique in that the BCR component contributes a serine-threonine kinase (STK) to the N-terminal end of the chimeric FGFR1 kinase. We have deleted the STK domain and mutated the critical Y177 residue and demonstrate that the transforming activity of these mutated genes is reduced compared to the BCR-FGFR1 parental kinase. In addition, we demonstrate that deletion of the FGFR1 tyrosine kinase domain abrogates transforming ability, which is not compensated for by BCR STK activity. Unbiased screening for proteins that are inactivated as a result of loss of the BCR STK identified activated S6 kinase and SHP2 kinase. Genetic and pharmacological inhibition of SHP2 function in SCLL cells expressing BCR-FGFR1 in vitro leads to reduced viability and increased apoptosis. In vivo treatment of SCLL in mice with SHP099 leads to suppression of leukemogenesis, supporting an important role for SHP2 in FGFR1-driven leukemogenesis. In combination with the BGJ398 FGFR1 inhibitor, cell viability in vitro is further suppressed and acts synergistically with SHP099 in vivo suggesting a potential combined targeted therapy option in this subtype of SCLL disease.
Collapse
Affiliation(s)
| | - Yun Liu
- Georgia Cancer Center, Augusta, GA.,Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Sumin Lu
- Georgia Cancer Center, Augusta, GA
| | - Baohuan Cai
- Georgia Cancer Center, Augusta, GA.,Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | | | | | | | | | | |
Collapse
|
20
|
Peiris MN, Meyer AN, Nelson KN, Bisom-Rapp EW, Donoghue DJ. Oncogenic fusion protein BCR-FGFR1 requires the breakpoint cluster region-mediated oligomerization and chaperonin Hsp90 for activation. Haematologica 2019; 105:1262-1273. [PMID: 31439673 PMCID: PMC7193502 DOI: 10.3324/haematol.2019.220871] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/14/2019] [Indexed: 01/07/2023] Open
Abstract
Mutation and translocation of fibroblast growth factor receptors often lead to aberrant signaling and cancer. This work focuses on the t(8;22)(p11;q11) chromosomal translocation which creates the breakpoint cluster region (BCR) fibroblast growth factor receptor1 (FGFR1) (BCR-FGFR1) fusion protein. This fusion occurs in stem cell leukemia/lymphoma, which can progress to atypical chronic myeloid leukemia, acute myeloid leukemia, or B-cell lymphoma. This work focuses on the biochemical characterization of BCR-FGFR1 and identification of novel therapeutic targets. The tyrosine kinase activity of FGFR1 is required for biological activity as shown using transformation assays, interleukin-3 independent cell proliferation, and liquid chromatography/mass spectroscopy analyses. Furthermore, BCR contributes a coiled-coil oligomerization domain, also essential for oncogenic transformation by BCR-FGFR1. The importance of salt bridge formation within the coiled-coil domain is demonstrated, as disruption of three salt bridges abrogates cellular transforming ability. Lastly, BCR-FGFR1 acts as a client of the chaperonin heat shock protein 90 (Hsp90), suggesting that BCR-FGFR1 relies on Hsp90 complex to evade proteasomal degradation. Transformed cells expressing BCR-FGFR1 are sensitive to the Hsp90 inhibitor Ganetespib, and also respond to combined treatment with Ganetespib plus the FGFR inhibitor BGJ398. Collectively, these data suggest novel therapeutic approaches for future stem cell leukemia/lymphoma treatment: inhibition of BCR oligomerization by disruption of required salt bridges; and inhibition of the chaperonin Hsp90 complex.
Collapse
Affiliation(s)
- Malalage N Peiris
- Department of Chemistry and Biochemistry, University of California San Diego
| | - April N Meyer
- Department of Chemistry and Biochemistry, University of California San Diego
| | - Katelyn N Nelson
- Department of Chemistry and Biochemistry, University of California San Diego
| | - Ezra W Bisom-Rapp
- Department of Chemistry and Biochemistry, University of California San Diego
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego .,Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
21
|
BCR: a promiscuous fusion partner in hematopoietic disorders. Oncotarget 2019; 10:2738-2754. [PMID: 31105873 PMCID: PMC6505627 DOI: 10.18632/oncotarget.26837] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/23/2019] [Indexed: 12/23/2022] Open
Abstract
Considerable advances have been made in our understanding of the molecular basis of hematopoietic cancers. The discovery of the BCR-ABL fusion protein over 50 years ago has brought about a new era of therapeutic progress and overall improvement in patient care, mainly due to the development and use of personalized medicine and tyrosine kinase inhibitors (TKIs). However, since the detection of BCR-ABL, BCR has been identified as a commonly occurring fusion partner in hematopoietic disorders. BCR has been discovered fused to additional tyrosine kinases, including: Fibroblast Growth Factor Receptor 1 (FGFR1), Platelet-derived Growth Factor Receptor Alpha (PDGFRA), Ret Proto-Oncogene (RET), and Janus Kinase 2 (JAK2). While BCR translocations are infrequent in hematopoietic malignancies, clinical evidence suggests that patients who harbor these mutations benefit from TKIs and additional personalized therapies. The improvement of further methodologies for characterization of these fusions is crucial to determine a patient’s treatment regimen, and optimal outcome. However, potential relapse and drug resistance among patients’ highlights the need for additional treatment options and further understanding of these oncogenic fusion proteins. This review explores the mechanisms behind cancer progression of these BCR oncogenic fusion proteins, comparing their similarities and differences, examining the significance of BCR as a partner gene, and discussing current treatment options for these translocation-induced hematopoietic malignancies.
Collapse
|