1
|
Yu L, Liu J, Jia J, Yang J, Tong R, Zhang X, Zhang Y, Yin S, Li J, Sun D. Fusion Genes Landscape of Lung Cancer Patients From Inner Mongolia, China. Genes Chromosomes Cancer 2024; 63:e23258. [PMID: 39011998 DOI: 10.1002/gcc.23258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 07/17/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths globally. Gene fusion, a key driver of tumorigenesis, has led to the identification of numerous driver gene fusions for lung cancer diagnosis and treatment. However, previous studies focused on Western populations, leaving the possibility of unrecognized lung cancer-associated gene fusions specific to Inner Mongolia due to its unique genetic background and dietary habits. To address this, we conducted DNA sequencing analysis on tumor and adjacent nontumor tissues from 1200 individuals with lung cancer in Inner Mongolia. Our analysis established a comprehensive fusion gene landscape specific to lung cancer in Inner Mongolia, shedding light on potential region-specific molecular mechanisms underlying the disease. Compared to Western cohorts, we observed a higher occurrence of ALK and RET fusions in Inner Mongolian patients. Additionally, we discovered eight novel fusion genes in three patients: SLC34A2-EPHB1, CCT6P3-GSTP1, BARHL2-APC, HRAS-MELK, FAM134B-ERBB2, ABCB1-GIPC1, GPR98-ALK, and FAM134B-SALL1. These previously unreported fusion genes suggest potential regional specificity. Furthermore, we characterized the fusion genes' structures based on breakpoints and described their impact on major functional gene domains. Importantly, the identified novel fusion genes exhibited significant clinical and pathological relevance. Notably, patients with SLC34A2-EPHB1, CCT6P3-GSTP1, and BARHL2-APC fusions showed sensitivity to the combination of chemotherapy and immunotherapy. Patients with HRAS-MELK, FAM134B-ERBB2, and ABCB1-GIPC1 fusions showed sensitivity to chemotherapy. In summary, our study provides novel insights into the frequency, distribution, and characteristics of specific fusion genes, offering valuable guidance for the development of effective clinical treatments, particularly in Inner Mongolia.
Collapse
Affiliation(s)
- Lan Yu
- Clinical Medical Research Center, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| | - Jinyang Liu
- Department of Sciences, Geneis Beijing Co. Ltd., Beijing, China
- Department of Data Mining, Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Jianchao Jia
- Clinical Medical Research Center, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| | - Jie Yang
- Clinical Medical Research Center, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| | - Ruiying Tong
- Clinical Medical Research Center, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| | - Xiao Zhang
- Clinical Medical Research Center, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of the Metabolic Disease, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| | - Yun Zhang
- Department of Sciences, Geneis Beijing Co. Ltd., Beijing, China
- Department of Data Mining, Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Songtao Yin
- Department of Medical Imaging, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| | - Junlin Li
- Department of Medical Imaging, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| | - Dejun Sun
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
- Pulmonary and Critical Care Medicine, Inner Mongolian People's Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Lu S, Zheng X, Sun Y, Huang D, Wu L, Ji Q, Zhou C, Zhou J, Guo Y, Ge M, Ding D, Shao J, Zhang W, Gao M, Cheng Y. Patient-reported outcomes following selpercatinib treatment in Chinese patients with advanced RET fusion-positive non-small-cell lung cancer and thyroid cancer, and RET-mutant medullary thyroid cancer in the phase II LIBRETTO-321 trial. Ther Adv Med Oncol 2023; 15:17588359231189429. [PMID: 37655205 PMCID: PMC10467255 DOI: 10.1177/17588359231189429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/05/2023] [Indexed: 09/02/2023] Open
Abstract
Background Patient-reported outcomes (PROs) are increasingly becoming an important part of clinical trials as they are helpful in analyzing the safety and efficacy of treatment in chronic diseases like cancer. Objectives We report PROs and health-related quality of life (HRQoL) with selpercatinib treatment among Chinese patients with rearranged in transfection (RET) fusion-positive non-small-cell lung cancer (NSCLC), RET fusion-positive thyroid cancer (TC), and RET-mutant medullary TC (MTC) as an exploratory analysis of the LIBRETTO-321 trial. Design A total of 77 patients (47 RET fusion-positive NSCLC, 1 RET fusion-positive TC, and 29 RET-mutant MTC) were enrolled. Compliance for European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire-Core 30 (QLQ-C30) was 100% at baseline and >90% at each time point. Methods PROs were assessed using the EORTC QLQ-C30, and a bowel diary assessment for MTC patients with baseline diarrhea using the Systemic Therapy-Induced Diarrhea Assessment Tool. Data were collected at pre-dose; every 8 weeks from cycle 3; and every 12 weeks after cycle 13. A >10-point change from baseline was considered clinically meaningful. PRO changes were summarized through cycle 13. Results Most patients with NSCLC or MTC showed improvement or remained stable on the global health status and functional subscales. For global health status, 47.4% of NSCLC and MTC patients showed definite improvement with only 19.7% showing definite worsening. For functional subscales, less than 30% of the patients showed definite worsening. For symptom subscales, more than 64% of the patients either improved or remained stable for the symptoms. For MTC patients with bowel diary assessment (n = 5), there was no severity or worsening from baseline in the diarrheal episodes observed during treatment with selpercatinib. Conclusion The study demonstrated favorable PROs in Chinese patients with RET fusion-positive NSCLC, TC, and RET-mutant MTC treated with selpercatinib. HRQoL was improved or stable as assessed by EORTC QLQ-30. Trail registration This study was registered at ClinicalTrials.gov (https://clinicaltrials.gov/ct2/show/NCT04280081) ClinicalTrials.gov Identifier: NCT04280081.
Collapse
Affiliation(s)
- Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, 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
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Dingzhi Huang
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lin Wu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chengzhi Zhou
- Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianying Zhou
- Department of Respiratory Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ye Guo
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Minghua Ge
- Department of Head, Neck and Thyroid Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ding Ding
- Eli Lilly and Company, Shanghai, China
| | | | | | - Ming Gao
- Department of Thyroid and Neck Tumor, 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 300040, China
- Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, Tianjin 300121, China
- Tianjin Key Laboratory of General Surgery Inconstruction, Tianjin Union Medical Center, Tianjin 300121, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun 130012, Jilin, China
| |
Collapse
|
3
|
Nacchio M, Pisapia P, Pepe F, Russo G, Vigliar E, Porcelli T, Luongo C, Iaccarino A, Pagni F, Salvatore D, Troncone G, Malapelle U, Bellevicine C. Predictive molecular pathology in metastatic thyroid cancer: the role of RET fusions. Expert Rev Endocrinol Metab 2022; 17:167-178. [PMID: 35404189 DOI: 10.1080/17446651.2022.2060819] [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: 01/25/2022] [Accepted: 03/29/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Rearranged during transfection (RET) gene fusions are detected in 10-20% of thyroid cancer patients. Recently, RET fusion-positive metastatic thyroid cancers have attracted much attention owing to the FDA approval of two highly selective anti-RET tyrosine kinase inhibitors, namely, selpercatinib, and pralsetinib. AREAS COVERED This review summarizes the available evidence on the biological and predictive role of RET gene fusions in thyroid carcinoma patients and the latest screening assays currently used to detect these genomic alterations in histological and cytological specimens. EXPERT OPINION Management of advanced thyroid carcinoma has significantly evolved over the last decade thanks to the approval of three multikinase inhibitors, i.e. sorafenib, lenvatinib, cabozantinib, and of two selective RET-tyrosine inhibitors, i.e. selpercatinib and pralsetinib. In this setting, the detection of RET-fusions in advanced thyroid cancer specimens through the use of next-generation sequencing has become a commonly used strategy in clinical practice to select the best treatment options.
Collapse
Affiliation(s)
- Mariantonia Nacchio
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Tommaso Porcelli
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Cristina Luongo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Antonino Iaccarino
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
4
|
Saha D, Ryan KR, Lakkaniga NR, Acharya B, Garcia NG, Smith EL, Frett B. Targeting Rearranged during Transfection in Cancer: A Perspective on Small-Molecule Inhibitors and Their Clinical Development. J Med Chem 2021; 64:11747-11773. [PMID: 34402300 DOI: 10.1021/acs.jmedchem.0c02167] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rearranged during transfection (RET) is a receptor tyrosine kinase essential for the normal development and maturation of a diverse range of tissues. Aberrant RET signaling in cancers, due to RET mutations, gene fusions, and overexpression, results in the activation of downstream pathways promoting survival, growth, and metastasis. Pharmacological manipulation of RET is effective in treating RET-driven cancers, and efforts toward developing RET-specific therapies have increased over the last 5 years. In 2020, RET-selective inhibitors pralsetinib and selpercatinib achieved clinical approval, which marked the first approvals for kinase inhibitors specifically developed to target the RET oncoprotein. This Perspective discusses current development and clinical applications for RET precision medicine by providing an overview of the incremental improvement of kinase inhibitors for use in RET-driven malignancies.
Collapse
Affiliation(s)
- Debasmita Saha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Katie Rose Ryan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Naga Rajiv Lakkaniga
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Baku Acharya
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Noemi Garcia Garcia
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Erica Lane Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 United States
| |
Collapse
|
5
|
Moccia M, Yang D, Lakkaniga NR, Frett B, McConnell N, Zhang L, Brescia A, Federico G, Zhang L, Salerno P, Santoro M, Li HY, Carlomagno F. Targeted activity of the small molecule kinase inhibitor Pz-1 towards RET and TRK kinases. Sci Rep 2021; 11:16103. [PMID: 34373541 PMCID: PMC8352932 DOI: 10.1038/s41598-021-95612-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 07/20/2021] [Indexed: 11/09/2022] Open
Abstract
We have recently described Pz-1, a benzimidazole-based type-2 RET and VEGFR2 inhibitor. Based on a kinome scan, here we show that Pz-1 is also a potent (IC50 < 1 nM) TRKA/B/C inhibitor. Pz-1 potently inhibited proliferation of human cancer cells carrying either RET- or TRKA oncoproteins (IC50 ~ 1 nM), with a negligible effect against RET- and TRKA-negative cells. By testing mutations, known to mediate resistance to other compounds, RET G810R/S, but not L730I/V, E732K, V738A and Y806N, showed some degree of resistance to Pz-1. In the case of TRKA, G595R and F589L, but not G667C, showed some degree of resistance. In xenograft models, orally administered Pz-1 almost completely inhibited RET- and TRKA-mutant tumours at 1-3 mg/kg/day but showed a reduced effect on RET/TRKA-negative cancer models. The activity, albeit reduced, on RET/TRKA-negative tumours may be justified by VEGFR2 inhibition. Tumours induced by NIH3T3 cells transfected by RET G810R and TRKA G595R featured resistance to Pz-1, demonstrating that RET or TRKA inhibition is critical for its anti-tumourigenic effect. In conclusion, Pz-1 represents a new powerful kinase inhibitor with distinct activity towards cancers induced by oncogenic RET and TRKA variants, including some mutants displaying resistance to other drugs.
Collapse
Affiliation(s)
- Marialuisa Moccia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - Donglin Yang
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - Naga Rajiv Lakkaniga
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Synactix Pharmaceuticals, Inc., Tucson, AZ, 85718, USA
| | - Nicholas McConnell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Lingtian Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Annalisa Brescia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - Giorgia Federico
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - Lingzhi Zhang
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - Paolo Salerno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
- Synactix Pharmaceuticals, Inc., Tucson, AZ, 85718, USA.
| | - Francesca Carlomagno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli "Federico II", Via S. Pansini 5, 80131, Naples, Italy.
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, 80131, Naples, Italy.
| |
Collapse
|
6
|
Liu M, Chen P, Hu HY, Ou-Yang DJ, Khushbu RA, Tan HL, Huang P, Chang S. Kinase gene fusions: roles and therapeutic value in progressive and refractory papillary thyroid cancer. J Cancer Res Clin Oncol 2021; 147:323-337. [PMID: 33387037 DOI: 10.1007/s00432-020-03491-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022]
Abstract
The incidence of papillary thyroid cancer (PTC), the major type of thyroid cancer, is increasing rapidly around the world, and its pathogenesis is still unclear. There is poor prognosis for PTC involved in rapidly progressive tumors and resistance to radioiodine therapy. Kinase gene fusions have been discovered to be present in a wide variety of malignant tumors, and an increasing number of novel types have been detected in PTC, especially progressive tumors. As a tumor-driving event, kinase fusions are constitutively activated or overexpress their kinase function, conferring oncogenic potential, and their frequency is second only to BRAFV600E mutation in PTC. Diverse forms of kinase fusions have been observed and are associated with specific pathological features of PTC (usually at an advanced stage), and clinical trials of therapeutic strategies targeting kinase gene fusions are feasible for radioiodine-resistant PTC. This review summarizes the roles of kinase gene fusions in PTC and the value of clinical therapy of targeting fusions in progressive or refractory PTC, and discusses the future perspectives and challenges related to kinase gene fusions in PTC patients.
Collapse
Affiliation(s)
- Mian Liu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Pei Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Hui-Yu Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Deng-Jie Ou-Yang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Rooh-Afza Khushbu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Hai-Long Tan
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Peng Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
7
|
Kohno T, Tabata J, Nakaoku T. REToma: a cancer subtype with a shared driver oncogene. Carcinogenesis 2020; 41:123-129. [PMID: 31711124 DOI: 10.1093/carcin/bgz184] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/17/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
RET (REarranged during Transfection), which encodes a receptor tyrosine kinase for members of the glial cell line-derived neurotrophic factor, plays a role as driver oncogene in a variety of human cancers. Fusion of RET with several partner genes has been detected in papillary thyroid, lung, colorectal, pancreatic and breast cancers, and tyrosine kinase inhibitors (TKIs) for RET (particularly RET-specific inhibitors) show promising therapeutic effects against such cancers. Oncogenic mutations within the extracellular cysteine-rich and intracellular kinase domains of RET drive medullary thyroid carcinogenesis; the same mutations are also observed in a small subset of diverse cancers such as lung, colorectal and breast cancers. Considering the oncogenic nature of RET mutants, lung, colorectal and breast cancers are predicted to respond to RET TKIs in a manner similar to medullary thyroid cancer. In summary, cancers carrying oncogenic RET alterations as a driver mutation could be collectively termed 'REToma' and treated with RET TKIs in a tissue-agnostic manner.
Collapse
Affiliation(s)
- Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Junya Tabata
- Division of Genome Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Takashi Nakaoku
- Division of Genome Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| |
Collapse
|
8
|
Moccia M, Frett B, Zhang L, Lakkaniga NR, Briggs DC, Chauhan R, Brescia A, Federico G, Yan W, Santoro M, McDonald NQ, Li HY, Carlomagno F. Bioisosteric Discovery of NPA101.3, a Second-Generation RET/VEGFR2 Inhibitor Optimized for Single-Agent Polypharmacology. J Med Chem 2020; 63:4506-4516. [PMID: 32298114 PMCID: PMC7901654 DOI: 10.1021/acs.jmedchem.9b01336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
RET receptor tyrosine kinase is a driver oncogene in human cancer. We recently identified the clinical drug candidate Pz-1, which targets RET and VEGFR2. A key in vivo metabolite of Pz-1 is its less active demethylated pyrazole analogue. Using bioisosteric substitution methods, here, we report the identification of NPA101.3, lacking the structural liability for demethylation. NPA101.3 showed a selective inhibitory profile and an inhibitory concentration 50 (IC50) of <0.003 μM for both RET and VEGFR2. NPA101.3 inhibited phosphorylation of all tested RET oncoproteins as well as VEGFR2 and proliferation of cells transformed by RET. Oral administration of NPA101.3 (10 mg/kg/day) completely prevented formation of tumors induced by RET/C634Y-transformed cells, while it weakened, but did not abrogate, formation of tumors induced by a control oncogene (HRAS/G12V). The balanced synchronous inhibition of both RET and VEGFR2, as well the resistance to demethylation, renders NPA101.3 a potential clinical candidate for RET-driven cancers.
Collapse
Affiliation(s)
- Marialuisa Moccia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States.,Synactix Pharmaceuticals, Inc., Tucson, Arizona 85718, United States
| | - Lingtian Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Naga Rajiv Lakkaniga
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - David C Briggs
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, U.K
| | - Rakhee Chauhan
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, U.K
| | - Annalisa Brescia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Giorgia Federico
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Wei Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Neil Q McDonald
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, U.K.,Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London WC1E 7HX, U.K
| | - Hong-Yu Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States.,Synactix Pharmaceuticals, Inc., Tucson, Arizona 85718, United States
| | - Francesca Carlomagno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy.,Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, 80131 Napoli, Italy
| |
Collapse
|
9
|
RET Gene Fusions in Malignancies of the Thyroid and Other Tissues. Genes (Basel) 2020; 11:genes11040424. [PMID: 32326537 PMCID: PMC7230609 DOI: 10.3390/genes11040424] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/05/2020] [Accepted: 04/12/2020] [Indexed: 02/07/2023] Open
Abstract
Following the identification of the BCR-ABL1 (Breakpoint Cluster Region-ABelson murine Leukemia) fusion in chronic myelogenous leukemia, gene fusions generating chimeric oncoproteins have been recognized as common genomic structural variations in human malignancies. This is, in particular, a frequent mechanism in the oncogenic conversion of protein kinases. Gene fusion was the first mechanism identified for the oncogenic activation of the receptor tyrosine kinase RET (REarranged during Transfection), initially discovered in papillary thyroid carcinoma (PTC). More recently, the advent of highly sensitive massive parallel (next generation sequencing, NGS) sequencing of tumor DNA or cell-free (cfDNA) circulating tumor DNA, allowed for the detection of RET fusions in many other solid and hematopoietic malignancies. This review summarizes the role of RET fusions in the pathogenesis of human cancer.
Collapse
|
10
|
Abstract
BACKGROUND Disease gene prediction is a critical and challenging task. Many computational methods have been developed to predict disease genes, which can reduce the money and time used in the experimental validation. Since proteins (products of genes) usually work together to achieve a specific function, biomolecular networks, such as the protein-protein interaction (PPI) network and gene co-expression networks, are widely used to predict disease genes by analyzing the relationships between known disease genes and other genes in the networks. However, existing methods commonly use a universal static PPI network, which ignore the fact that PPIs are dynamic, and PPIs in various patients should also be different. RESULTS To address these issues, we develop an ensemble algorithm to predict disease genes from clinical sample-based networks (EdgCSN). The algorithm first constructs single sample-based networks for each case sample of the disease under study. Then, these single sample-based networks are merged to several fused networks based on the clustering results of the samples. After that, logistic models are trained with centrality features extracted from the fused networks, and an ensemble strategy is used to predict the finial probability of each gene being disease-associated. EdgCSN is evaluated on breast cancer (BC), thyroid cancer (TC) and Alzheimer's disease (AD) and obtains AUC values of 0.970, 0.971 and 0.966, respectively, which are much better than the competing algorithms. Subsequent de novo validations also demonstrate the ability of EdgCSN in predicting new disease genes. CONCLUSIONS In this study, we propose EdgCSN, which is an ensemble learning algorithm for predicting disease genes with models trained by centrality features extracted from clinical sample-based networks. Results of the leave-one-out cross validation show that our EdgCSN performs much better than the competing algorithms in predicting BC-associated, TC-associated and AD-associated genes. de novo validations also show that EdgCSN is valuable for identifying new disease genes.
Collapse
Affiliation(s)
- Ping Luo
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, S7N 5A9 Canada
| | - Li-Ping Tian
- School of Information, Beijing Wuzi University, Beijing, 101149 China
| | - Bolin Chen
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710072 China
| | - Qianghua Xiao
- School of Mathematics and Physics, University of South China, HengYang, 421001 China
| | - Fang-Xiang Wu
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, S7N 5A9 Canada
- Department of Computer Science, University of Saskatchewan, Saskatoon, S7N 5C9 Canada
- School of Mathematics and Statistics, Hainan Normal University, Haikou, 571158 China
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, S7N 5A9 Canada
| |
Collapse
|
11
|
Subbiah V, Yang D, Velcheti V, Drilon A, Meric-Bernstam F. State-of-the-Art Strategies for Targeting RET-Dependent Cancers. J Clin Oncol 2020; 38:1209-1221. [PMID: 32083997 DOI: 10.1200/jco.19.02551] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Activating receptor tyrosine kinase RET (rarranged during transfection) gene alterations have been identified as oncogenic in multiple malignancies. RET gene rearrangements retaining the kinase domain are oncogenic drivers in papillary thyroid cancer, non-small-cell lung cancer, and multiple other cancers. Activating RET mutations are associated with different phenotypes of multiple endocrine neoplasia type 2 as well as sporadic medullary thyroid cancer. RET is thus an attractive therapeutic target in patients with oncogenic RET alterations. Multikinase inhibitors with RET inhibitor activity, such as cabozantinib and vandetanib, have been explored in the clinic for tumors with activating RET gene alterations with modest clinical efficacy. As a result of the nonselective nature of these multikinase inhibitors, patients had off-target adverse effects, such as hypertension, rash, and diarrhea. This resulted in a narrow therapeutic index of these drugs, limiting ability to dose for clinically effective RET inhibition. In contrast, the recent discovery and clinical validation of highly potent selective RET inhibitors (pralsetinib, selpercatinib) demonstrating improved efficacy and a more favorable toxicity profile are poised to alter the landscape of RET-dependent cancers. These drugs appear to have broad activity across tumors with activating RET alterations. The mechanisms of resistance to these next-generation highly selective RET inhibitors is an area of active research. This review summarizes the current understanding of RET alterations and the state-of-the-art treatment strategies in RET-dependent cancers.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.,Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX.,MD Anderson Cancer Network, Houston, TX
| | - Dong Yang
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Alexander Drilon
- Thoracic Oncology Service, Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.,Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
12
|
de Bruin RG, Vogel G, Prins J, Duijs JMJG, Bijkerk R, van der Zande HJP, van Gils JM, de Boer HC, Rabelink TJ, van Zonneveld AJ, van der Veer EP, Richard S. Targeting the RNA-Binding Protein QKI in Myeloid Cells Ameliorates Macrophage-Induced Renal Interstitial Fibrosis. EPIGENOMES 2020; 4:epigenomes4010002. [PMID: 34968236 PMCID: PMC8594696 DOI: 10.3390/epigenomes4010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
In the pathophysiologic setting of acute and chronic kidney injury, the excessive activation and recruitment of blood-borne monocytes prompts their differentiation into inflammatory macrophages, a process that leads to progressive glomerulosclerosis and interstitial fibrosis. Importantly, this differentiation of monocytes into macrophages requires the meticulous coordination of gene expression at both the transcriptional and post-transcriptional level. The transcriptomes of these cells are ultimately determined by RNA-binding proteins such as QUAKING (QKI), that define their pre-mRNA splicing and mRNA transcript patterns. Using two mouse models, namely (1) quaking viable mice (qkv) and (2) the conditional deletion in the myeloid cell lineage using the lysozyme 2-Cre (QKIFL/FL;LysM-Cre mice), we demonstrate that the abrogation of QKI expression in the myeloid cell lineage reduces macrophage infiltration following kidney injury induced by unilateral urethral obstruction (UUO). The qkv and QKIFL/FL;LysM-Cre mice both showed significant diminished interstitial collagen deposition and fibrosis in the UUO-damaged kidney, as compared to wild-type littermates. We show that macrophages isolated from QKIFL/FL;LysM-Cre mice are associated with defects in pre-mRNA splicing. Our findings demonstrate that reduced expression of the alternative splice regulator QKI in the cells of myeloid lineage attenuates renal interstitial fibrosis, suggesting that inhibition of this splice regulator may be of therapeutic value for certain kidney diseases.
Collapse
Affiliation(s)
- Ruben G. de Bruin
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Biochemistry, Human Genetics and Medicine, McGill University, Montréal, QC H3T 1E2, Canada;
| | - Gillian Vogel
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Biochemistry, Human Genetics and Medicine, McGill University, Montréal, QC H3T 1E2, Canada;
| | - Jurrien Prins
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Jacques M. J. G. Duijs
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Roel Bijkerk
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Hendrik J. P. van der Zande
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Janine M. van Gils
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Hetty C. de Boer
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Ton J. Rabelink
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
| | - Eric P. van der Veer
- Einthoven Laboratory for Experimental Vascular Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Albinusdreef 2, C7-36, PO Box 9600, 2300RC Leiden, The Netherlands; (R.G.d.B.); (J.P.); (J.M.J.G.D.); (R.B.); (H.J.P.v.d.Z.); (J.M.v.G.); (H.C.d.B.); (T.J.R.); (A.J.v.Z.)
- Correspondence: (E.P.v.d.V.); (S.R.)
| | - Stéphane Richard
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Biochemistry, Human Genetics and Medicine, McGill University, Montréal, QC H3T 1E2, Canada;
- Correspondence: (E.P.v.d.V.); (S.R.)
| |
Collapse
|
13
|
ANKRD26-RET - A novel gene fusion involving RET in papillary thyroid carcinoma. Cancer Genet 2019; 238:10-17. [DOI: 10.1016/j.cancergen.2019.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/13/2019] [Accepted: 07/01/2019] [Indexed: 12/14/2022]
|
14
|
Jiang W, Ji M. Receptor tyrosine kinases in PI3K signaling: The therapeutic targets in cancer. Semin Cancer Biol 2019; 59:3-22. [PMID: 30943434 DOI: 10.1016/j.semcancer.2019.03.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 03/09/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway, one of the most commonly activated signaling pathways in human cancers, plays a crucial role in the regulation of cell proliferation, differentiation, and survival. This pathway is usually activated by receptor tyrosine kinases (RTKs), whose constitutive and aberrant activation is via gain-of-function mutations, chromosomal rearrangement, gene amplification and autocrine. Blockage of PI3K pathway by targeted therapy on RTKs with tyrosine kinases inhibitors (TKIs) and monoclonal antibodies (mAbs) has achieved great progress in past decades; however, there still remain big challenges during their clinical application. In this review, we provide an overview about the most frequently encountered alterations in RTKs and focus on current therapeutic agents developed to counteract their aberrant functions, accompanied with discussions of two major challenges to the RTKs-targeted therapy in cancer - resistance and toxicity.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Meiju Ji
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| |
Collapse
|
15
|
Cui Y, Huang Y, Wu X, Zheng M, Xia Y, Fu Z, Ge H, Wang S, Xie H. Hypoxia‐induced tRNA‐derived fragments, novel regulatory factor for doxorubicin resistance in triple‐negative breast cancer. J Cell Physiol 2018; 234:8740-8751. [PMID: 30362543 DOI: 10.1002/jcp.27533] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Yangyang Cui
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| | - Yue Huang
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| | - Xiaowei Wu
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| | - Mingjie Zheng
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| | - Yiqin Xia
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| | - Ziyi Fu
- Department of Oncology The First Affiliated Hospital, Nanjing Medical University Nanjing China
- Medical Research Center, Nanjing Maternal and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital Nanjing China
- Obstetrics and Gynecology Department Northwestern University Chicago Illinois
| | - Han Ge
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| | - Shui Wang
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| | - Hui Xie
- Department of Breast Surgery The First Affiliated Hospital, Nanjing Medical University Nanjing China
| |
Collapse
|
16
|
Targeting RET-driven cancers: lessons from evolving preclinical and clinical landscapes. Nat Rev Clin Oncol 2017; 15:151-167. [PMID: 29134959 DOI: 10.1038/nrclinonc.2017.175] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The gene encoding the receptor-tyrosine kinase RET was first discovered more than three decades ago, and activating RET rearrangements and mutations have since been identified as actionable drivers of oncogenesis. Several multikinase inhibitors with activity against RET have been explored in the clinic, and confirmed responses to targeted therapy with these agents have been observed in patients with RET-rearranged lung cancers or RET-mutant thyroid cancers. Nevertheless, response rates to RET-directed therapy are modest compared with those achieved using targeted therapies matched to other oncogenic drivers of solid tumours, such as sensitizing EGFR or BRAFV600E mutations, or ALK or ROS1 rearrangements. To date, no RET-directed targeted therapeutic has received regulatory approval for the treatment of molecularly defined populations of patients with RET-mutant or RET-rearranged solid tumours. In this Review, we discuss how emerging data have informed the debate over whether the limited success of multikinase inhibitors with activity against RET can be attributed to the tractability of RET as a drug target or to the lack, until 2017, of highly specific inhibitors of this oncoprotein in the clinic. We emphasize that novel approaches to targeting RET-dependent tumours are necessary to improve the clinical efficacy of single-agent multikinase inhibition and, thus, hasten approvals of RET-directed targeted therapies.
Collapse
|
17
|
Watanabe-Smith K, Godil J, Agarwal A, Tognon C, Druker B. Analysis of acquired mutations in transgenes arising in Ba/F3 transformation assays: findings and recommendations. Oncotarget 2017; 8:12596-12606. [PMID: 28208123 PMCID: PMC5355038 DOI: 10.18632/oncotarget.15392] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/12/2017] [Indexed: 12/21/2022] Open
Abstract
The identification and functional validation of potentially oncogenic mutations in leukemia is an essential step toward a future of personalized targeted therapy. To assess the oncogenic capacity of individual mutations, reliable and scalable in vitro experimental approaches are required. Since 1988, researchers have used the IL-3 dependent Ba/F3 transformation assay to validate the oncogenic potential of mutations to drive factor-independent growth. Here we report a previously unrecognized phenomenon whereby Ba/F3 cells, engineered to express weakly transforming mutations, present with additional acquired mutations in the expressed transgene following factor withdrawal. Using four mutations with known transformative capacity in three cytokine receptors (CSF2RB, CSF3R and IL7R), we demonstrate that the mutated receptors are highly susceptible to acquiring additional mutations. These acquired mutations of unknown functional significance are selected by factor withdrawal but appear to exist prior to the removal of growth factor. This anomaly has the potential to confound efforts to both validate and characterize oncogenic mutations in leukemia, particularly when it is not standard practice to sequence validate cDNAs from transformed Ba/F3 lines. We present specific recommendations to detect and mitigate this phenomenon in future research using Ba/F3 transformation assays, along with methods to make the Ba/F3 assay more quantitative.
Collapse
Affiliation(s)
- Kevin Watanabe-Smith
- Cancer Biology Program, Oregon Health & Science University, Knight Cancer Institute, Portland, OR, USA
| | - Jamila Godil
- Honors College, College of Science, Oregon State University, Corvallis, OR, USA
| | - Anupriya Agarwal
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Knight Cancer Institute, Portland, OR, USA.,Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Cristina Tognon
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR, USA.,Howard Hughes Medical Institute, Portland, OR, USA
| | - Brian Druker
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Knight Cancer Institute, Portland, OR, USA.,Howard Hughes Medical Institute, Portland, OR, USA
| |
Collapse
|
18
|
De Falco V, Carlomagno F, Li HY, Santoro M. The molecular basis for RET tyrosine-kinase inhibitors in thyroid cancer. Best Pract Res Clin Endocrinol Metab 2017; 31:307-318. [PMID: 28911727 PMCID: PMC5624797 DOI: 10.1016/j.beem.2017.04.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RET receptor tyrosine kinase acts as a mutated oncogenic driver in several human malignancies and it is over-expressed in other cancers. Small molecule compounds with RET tyrosine kinase inhibitory activity are being investigated for the targeted treatment of these malignancies. Multi-targeted compounds with RET inhibitory concentration in the nanomolar range have entered clinical practice. This review summarizes mechanisms of RET oncogenic activity and properties of new compounds that, at the preclinical stage, have demonstrated promising anti-RET activity.
Collapse
Affiliation(s)
- Valentina De Falco
- Istituto di Endocrinologia e Oncologia Sperimentale, CNR, Via S Pansini 5, 80131 Naples, Italy.
| | - Francesca Carlomagno
- Istituto di Endocrinologia e Oncologia Sperimentale, CNR, Via S Pansini 5, 80131 Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131 Naples, Italy.
| | - Hong-Yu Li
- University of Arkansas for Medical Sciences, College of Pharmacy, Division of Pharmaceutical Science, 200 South Cedar, Little Rock AR 72202, USA.
| | - Massimo Santoro
- Istituto di Endocrinologia e Oncologia Sperimentale, CNR, Via S Pansini 5, 80131 Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Via S Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
19
|
Zuo W, Wang SA, DiNardo C, Yabe M, Li S, Medeiros LJ, Tang G. Acute leukaemia and myelodysplastic syndromes with chromosomal rearrangement involving 11q23 locus, but not MLL gene. J Clin Pathol 2016; 70:244-249. [PMID: 27496968 DOI: 10.1136/jclinpath-2016-203831] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/17/2016] [Accepted: 07/18/2016] [Indexed: 12/20/2022]
Abstract
AIMS Chromosome 11q23 translocations, resulting in MLL (KMT2A) rearrangement, have been well characterised in acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL). However, little is known of haematopoietic neoplasms associated with 11q23 translocation but without MLL rearrangement (11q23+/MLL-). The aim of this study is to characterise such cases with 11q23+/MLL-. METHODS AND RESULTS We retrospectively searched our database for cases with haematopoietic malignancies with 11q23+/MLL-. We identified nine patients, two with AML, two with B-lymphoblastic leukaemia (B-ALL); two with T-lymphoblastic leukaemia (T-ALL), two with myelodysplastic syndrome (MDS) and one with chronic myelomonocytic leukaemia (CMML). The translocations included t(X;11)(p11.2;q23), t(2;11)(p21;q23), t(6;11)(q27;q23), t(8;9;11)(q13;q13;q23), t(11;11)(p15;q23), t(11;14)(q23;q24) and t(11;15)(q23;q14). Five of six patients with acute leukaemia had received chemotherapy and detection of 11q23 translocation occurred at time of disease relapse. Both patients with MDS and the patient with CMML had 11q23 translocation detected at time of initial diagnosis, all three patients progressed to AML after >1 year on hypomethylating agent therapy. All patients received risk-adapted therapies, including stem cell transplant in five patients. At the last follow-up, eight patients died with a median overall survival of 14 months. CONCLUSIONS 11q23+/MLL- occurs rarely, involving different partner chromosomes and showing clinical and pathological features and disease subtypes different from those cases with MLL rearrangement. 11q23+/MLL- appears to be associated with clonal evolution/disease progression in acute leukaemia, a high risk for AML progression in MDS/CMML and a high incidence of disease relapse.
Collapse
Affiliation(s)
- Wenli Zuo
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Hematology, Zhengzhou University Affiliated Cancer Hospital/Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mariko Yabe
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
20
|
Romei C, Ciampi R, Elisei R. A comprehensive overview of the role of the RET proto-oncogene in thyroid carcinoma. Nat Rev Endocrinol 2016; 12:192-202. [PMID: 26868437 DOI: 10.1038/nrendo.2016.11] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The rearranged during transfection (RET) proto-oncogene was identified in 1985 and, very soon thereafter, a rearrangement named RET/PTC was discovered in papillary thyroid carcinoma (PTC). After this discovery, other RET rearrangements were found in PTCs, particularly in those induced by radiation. For many years, it was thought that these genetic alterations only occurred in PTC, but, in the past couple of years, some RET/PTC rearrangements have been found in other human tumours. 5 years after the discovery of RET/PTC rearrangements in PTC, activating point mutations in the RET proto-oncogene were discovered in both hereditary and sporadic forms of medullary thyroid carcinoma (MTC). In contrast to the alterations found in PTC, the activation of RET in MTC is mainly due to activating point mutations. Interestingly, in the past year, RET rearrangements that were different to those described in PTC were observed in sporadic MTC. The identification of RET mutations is relevant to the early diagnosis of hereditary MTC and the prognosis of sporadic MTC. The diagnostic and prognostic role of the RET/PTC rearrangements in PTC is less relevant but still important in patient management, particularly for deciding if a targeted therapy should be initiated. In this Review, we discuss the pathogenic, diagnostic and prognostic roles of the RET proto-oncogene in both PTC and MTC.
Collapse
Affiliation(s)
- Cristina Romei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Raffaele Ciampi
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| |
Collapse
|
21
|
Moccia M, Liu Q, Guida T, Federico G, Brescia A, Zhao Z, Choi HG, Deng X, Tan L, Wang J, Billaud M, Gray NS, Carlomagno F, Santoro M. Identification of Novel Small Molecule Inhibitors of Oncogenic RET Kinase. PLoS One 2015; 10:e0128364. [PMID: 26046350 PMCID: PMC4457528 DOI: 10.1371/journal.pone.0128364] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 04/25/2015] [Indexed: 01/08/2023] Open
Abstract
Oncogenic mutation of the RET receptor tyrosine kinase is observed in several human malignancies. Here, we describe three novel type II RET tyrosine kinase inhibitors (TKI), ALW-II-41-27, XMD15-44 and HG-6-63-01, that inhibit the cellular activity of oncogenic RET mutants at two digit nanomolar concentration. These three compounds shared a 3-trifluoromethyl-4-methylpiperazinephenyl pharmacophore that stabilizes the ‘DFG-out’ inactive conformation of RET activation loop. They blocked RET-mediated signaling and proliferation with an IC50 in the nM range in fibroblasts transformed by the RET/C634R and RET/M918T oncogenes. They also inhibited autophosphorylation of several additional oncogenic RET-derived point mutants and chimeric oncogenes. At a concentration of 10 nM, ALW-II-41-27, XMD15-44 and HG-6-63-01 inhibited RET kinase and signaling in human thyroid cancer cell lines carrying oncogenic RET alleles; they also inhibited proliferation of cancer, but not non-tumoral Nthy-ori-3-1, thyroid cells, with an IC50 in the nM range. The three compounds were capable of inhibiting the ‘gatekeeper’ V804M mutant which confers substantial resistance to established RET inhibitors. In conclusion, we have identified a type II TKI scaffold, shared by ALW-II-41-27, XMD15-44 and HG-6-63-01, that may be used as novel lead for the development of novel agents for the treatment of cancers harboring oncogenic activation of RET.
Collapse
Affiliation(s)
- Marialuisa Moccia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Naples, Italy
| | - Qingsong Liu
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Teresa Guida
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Naples, Italy
| | - Giorgia Federico
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Naples, Italy
| | - Annalisa Brescia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Naples, Italy
| | - Zheng Zhao
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, P.R. China
| | - Hwan Geun Choi
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xianming Deng
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Li Tan
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jinhua Wang
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marc Billaud
- Institut Albert Bonniot, CRI INSERM/UJF U823, La Tronche Cedex, France
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts United States of America
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Francesca Carlomagno
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Naples, Italy
| | - Massimo Santoro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Naples, Italy
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR, Naples, Italy
- * E-mail:
| |
Collapse
|
22
|
Morgensztern D, Campo MJ, Dahlberg SE, Doebele RC, Garon E, Gerber DE, Goldberg SB, Hammerman PS, Heist R, Hensing T, Horn L, Ramalingam SS, Rudin CM, Salgia R, Sequist L, Shaw AT, Simon GR, Somaiah N, Spigel DR, Wrangle J, Johnson D, Herbst RS, Bunn P, Govindan R. Molecularly targeted therapies in non-small-cell lung cancer annual update 2014. J Thorac Oncol 2015; 10:S1-63. [PMID: 25535693 PMCID: PMC4346098 DOI: 10.1097/jto.0000000000000405] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There have been significant advances in the understanding of the biology and treatment of non-small-cell lung cancer (NSCLC) during the past few years. A number of molecularly targeted agents are in the clinic or in development for patients with advanced NSCLC. We are beginning to understand the mechanisms of acquired resistance after exposure to tyrosine kinase inhibitors in patients with oncogene addicted NSCLC. The advent of next-generation sequencing has enabled to study comprehensively genomic alterations in lung cancer. Finally, early results from immune checkpoint inhibitors are very encouraging. This review summarizes recent advances in the area of cancer genomics, targeted therapies, and immunotherapy.
Collapse
Affiliation(s)
- Daniel Morgensztern
- Department of Medical Oncology, Washington University School of Medicine, Saint Louis, MO
| | - Meghan J. Campo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Suzanne E. Dahlberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston MA
| | - Robert C. Doebele
- Department of Medical Oncology, University of Colorado School of Medicine and University of Colorado Cancer Center, Aurora, CO
| | - Edward Garon
- UCLA Santa Monica Hematology Oncology, Santa Monica, CA
| | - David E. Gerber
- Division of Hematology-Oncology, Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Sarah B. Goldberg
- Department of Medical Oncology, Yale School of Medicine and Cancer Center, New Haven, CT
| | | | - Rebecca Heist
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - Thomas Hensing
- Department of Oncology, The University of Chicago Medicine, Chicago, IL
| | - Leora Horn
- Division of Hematology-Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Suresh S. Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA
| | | | - Ravi Salgia
- Department of Oncology, The University of Chicago Medicine, Chicago, IL
| | - Lecia Sequist
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - Alice T. Shaw
- Department of Oncology, Massachusetts General Hospital, Boston, MA
| | - George R. Simon
- Division of Hematology-Oncology, Medical University of South Carolina, Charleston, SC
| | - Neeta Somaiah
- Division of Hematology-Oncology, Medical University of South Carolina, Charleston, SC
| | | | - John Wrangle
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - David Johnson
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Roy S. Herbst
- Department of Medical Oncology, Yale School of Medicine and Cancer Center, New Haven, CT
| | - Paul Bunn
- Division of Medical Oncology, University of Colorado Denver School of Medicine, Denver, CO
| | - Ramaswamy Govindan
- Department of Medical Oncology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
23
|
Uversky VN. Wrecked regulation of intrinsically disordered proteins in diseases: pathogenicity of deregulated regulators. Front Mol Biosci 2014; 1:6. [PMID: 25988147 PMCID: PMC4428494 DOI: 10.3389/fmolb.2014.00006] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/06/2014] [Indexed: 12/14/2022] Open
Abstract
Biologically active proteins without stable tertiary structure are common in all known proteomes. Functions of these intrinsically disordered proteins (IDPs) are typically related to regulation, signaling, and control. Cellular levels of these important regulators are tightly regulated by a variety mechanisms ranging from firmly controlled expression to precisely targeted degradation. Functions of IDPs are controlled by binding to specific partners, alternative splicing, and posttranslational modifications among other means. In the norm, right amounts of precisely activated IDPs have to be present in right time at right places. Wrecked regulation brings havoc to the ordered world of disordered proteins, leading to protein misfolding, misidentification, and missignaling that give rise to numerous human diseases, such as cancer, cardiovascular disease, neurodegenerative diseases, and diabetes. Among factors inducing pathogenic transformations of IDPs are various cellular mechanisms, such as chromosomal translocations, damaged splicing, altered expression, frustrated posttranslational modifications, aberrant proteolytic degradation, and defective trafficking. This review presents some of the aspects of deregulated regulation of IDPs leading to human diseases.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida Tampa, FL, USA ; Biology Department, Faculty of Science, King Abdulaziz University Jeddah, Saudi Arabia ; Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences Moscow, Russia
| |
Collapse
|
24
|
Bossi D, Carlomagno F, Pallavicini I, Pruneri G, Trubia M, Raviele PR, Marinelli A, Anaganti S, Cox MC, Viale G, Santoro M, Di Fiore PP, Minucci S. Functional characterization of a novel FGFR1OP-RET rearrangement in hematopoietic malignancies. Mol Oncol 2013; 8:221-31. [PMID: 24315414 DOI: 10.1016/j.molonc.2013.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 10/29/2013] [Accepted: 11/11/2013] [Indexed: 01/08/2023] Open
Abstract
The RET (REarranged during Transfection) receptor tyrosine kinase is targeted by oncogenic rearrangements in thyroid and lung adenocarcinoma. Recently, a RET (exon 12) rearrangement with FGFR1OP [fibroblast growth factor receptor 1 (FGFR1) oncogene partner] (exon 12) was identified in one chronic myelomonocytic leukemia (CMML) patient. We report the molecular cloning and functional characterization of a novel FGFR1OP (exon 11)-RET (exon 11) gene fusion event (named FGFR1OP-RET), mediated by a reciprocal translocation t(6; 10)(q27; q11), in a patient affected by primary myelofibrosis (PMF) with secondary acute myeloid leukemia (AML). The FGFR1OP-RET fusion protein displayed constitutive tyrosine kinase and transforming activity in NIH3T3 fibroblasts, and induced IL3-independent growth and activation of PI3K/STAT signaling in hematopoietic Ba/F3 cells. FGFR1OP-RET supported cytokine-independent growth, protection from stress and enhanced self-renewal of primary murine hematopoietic progenitor and stem cells in vitro. In vivo, FGFR1OP-RET caused a spectrum of disease phenotypes, with >50% of mice showing a fatal myeloproliferative disorder (MPD). Other phenotypes were leukemia transplantable in secondary recipients, dramatic expansion of the mast cell lineage, and reduction of repopulating activity upon lethal irradiation. In conclusion, FGFR1OP-RET chimeric oncogenes are endowed with leukemogenic potential and associated to myeloid neoplasms (CMML and PMF/AML).
Collapse
MESH Headings
- Animals
- Chromosomes, Human, Pair 10/genetics
- Chromosomes, Human, Pair 10/metabolism
- Chromosomes, Human, Pair 6/genetics
- Chromosomes, Human, Pair 6/metabolism
- Hematologic Neoplasms/genetics
- Hematologic Neoplasms/metabolism
- Hematologic Neoplasms/pathology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive
- Mice
- NIH 3T3 Cells
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-ret/genetics
- Proto-Oncogene Proteins c-ret/metabolism
- Translocation, Genetic
Collapse
Affiliation(s)
- Daniela Bossi
- Department of Experimental Oncology, IEO, European Institute of Oncology, Milan, Italy
| | - Francesca Carlomagno
- Department of Biology and Cellular and Molecular Pathology, University Federico II, Naples, Italy
| | - Isabella Pallavicini
- Department of Experimental Oncology, IEO, European Institute of Oncology, Milan, Italy
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Maurizio Trubia
- Department of Experimental Oncology, IEO, European Institute of Oncology, Milan, Italy
| | | | | | - Suresh Anaganti
- Department of Biology and Cellular and Molecular Pathology, University Federico II, Naples, Italy
| | - Maria Christina Cox
- Department of Hematology, A.O. Sant'Andrea, University La Sapienza, Rome, Italy
| | - Giuseppe Viale
- Department of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Massimo Santoro
- Department of Biology and Cellular and Molecular Pathology, University Federico II, Naples, Italy.
| | - Pier Paolo Di Fiore
- Department of Experimental Oncology, IEO, European Institute of Oncology, Milan, Italy; Department of Scienze della Salute, University of Milan, Via di Rudinì 8, 20122 Milan, Italy; IFOM, FIRC Institute of Molecular Oncology, Milan, Italy.
| | - Saverio Minucci
- Department of Experimental Oncology, IEO, European Institute of Oncology, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy.
| |
Collapse
|