1
|
Wang Q, Wu W, Sun R, Cai L. Separation, characterization and cytotoxicity of unknown forced degradation impurity of selpercatinib using Prep-LC, HRMS and NMR. J Pharm Biomed Anal 2025; 258:116747. [PMID: 39954580 DOI: 10.1016/j.jpba.2025.116747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Selpercatinib (LOXO-292) is a newly marketed oral selective receptor tyrosine kinase inhibitor targeting rearranged during transfection (RET), demonstrating precise therapeutic effects against RET-positive non-small cell lung cancer and thyroid cancer. In this study, an unknown acid forced degradation impurity of selpercatinib, designated sel-1, was isolated and purified using semi-preparative liquid chromatography (semi-Prep-LC). The purified sel-1 showed a chromatographic purity of 99.1 % as determined by high-performance liquid chromatography (HPLC). It appeared as a white amorphous powder, with a maximum absorption peak at 235 nm and a chemical formula of C28H29N7O3. Its molecular structure was elucidated using high-resolution mass spectrometry (HRMS) and nuclear magnetic resonance (NMR). sel-1 was identified as 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-oxo-1,6-dihydropyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile. In vitro MTT assays revealed that sel-1 exhibited significant antitumor activity, particularly against HepaRG and MKN-1 cell lines, with stronger inhibition than selpercatinib. The study contributes to enhancing the quality control standards for selpercatinib and suggests that sel-1 holds potential for further drug development.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Wenyi Wu
- Department of Quality Inspection, Sinopharm Holdings Nantong Co., Ltd, Nantong, Jiangsu 226001, China
| | - Rongwei Sun
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Liangliang Cai
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
2
|
Luo Y, Li D, Yang Q, Dong Y, Chen W. Treatment of RET/ALK comutated advanced lung large cell neuroendocrine carcinoma: a case report and literature review. Anticancer Drugs 2025:00001813-990000000-00379. [PMID: 40112204 DOI: 10.1097/cad.0000000000001715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The prognosis of advanced lung large-cell neuroendocrine carcinoma is poor, and the efficacy of targeted therapy is still being explored. A case of RET fusion mutation combined with ALK rearrangement positive advanced lung complex large cell neuroendocrine carcinoma was reported. The patient developed intrapulmonary and bone metastases 8 months after chemotherapy after lung cancer surgery, RET fusion mutations were detected by genetic testing, and intracranial progression occurred 1 year after pilatinib was applied. The comutation of RET and ALK was detected by genetic testing, and the pulmonary progression occurred 2 months after the application of aletinib, after being treated with pilatinib and aletinib, he progressed again in 9 months. We point out that large cell neuroendocrine carcinoma complex patients with RET gene mutation can benefit from targeted therapy, and when drug resistance is accompanied by ALK comutation, the patient can benefit from the treatment of the aletinib combined with pilatinib targeted therapy and the side effect is slight. At the same time, we further explore the resistance mechanism of targeted therapy in lung cancer.
Collapse
Affiliation(s)
- Ying Luo
- Department of Radiation Oncology, Taizhou Central Hospital, Jiao jiang Street, Taizhou, Zhejiang, China
| | | | | | | | | |
Collapse
|
3
|
Katayama Y, Yamada T, Tanimura K, Kawachi H, Ishida M, Matsui Y, Hirai S, Nakamura R, Morimoto K, Furuya N, Arai S, Goto Y, Sakata Y, Nishino K, Tsuchiya M, Tamiya A, Saito G, Muto S, Takeda T, Date K, Fujisaka Y, Watanabe S, Fujimoto D, Uehara H, Horinaka M, Sakai T, Yano S, Tokuda S, Takayama K. YAP Regulates HER3 Signaling-Driven Adaptive Resistance to RET Inhibitors in RET-Aberrant Cancers. Clin Cancer Res 2025; 31:1127-1141. [PMID: 39495173 DOI: 10.1158/1078-0432.ccr-24-1762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/06/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE Rearranged during transfection (RET) aberrations represent a targetable oncogene in several tumor types, with RET inhibitors displaying marked efficacy. However, some patients with RET-aberrant cancer are insensitive to RET tyrosine kinase inhibitors (TKI). Recently, drug-tolerant mechanisms have attracted attention as targets for initial therapies to overcome drug resistance. The underlying mechanisms of drug-tolerant cell emergence treated with RET-TKIs derived from RET-aberrant cancer cells remain unknown. This study investigated the role of YAP-mediated HER3 signaling in the underlying mechanisms of adaptive resistance to RET-TKIs in RET-aberrant cancer cells. EXPERIMENTAL DESIGN Four RET-aberrant cancer cell lines were used to assess sensitivity to the RET-TKIs selpercatinib and pralsetinib and to elucidate the molecular mechanisms underlying adaptive resistance using RNA sequencing, phospho-receptor tyrosine kinase antibody arrays, chromatin immunoprecipitation assay, and luciferase reporter assays. Clinical specimens from patients with RET fusion-positive lung cancer were analyzed for pretreatment YAP expression and correlated with treatment outcomes. RESULTS In high YAP-expressing RET-aberrant cancer cells, YAP-mediated HER3 signaling activation maintained cell survival and induced the emergence of cells tolerant to the RET-TKIs selpercatinib and pralsetinib. The pan-ErBB inhibitor afatinib and YAP/tea domain inhibitors verteporfin and K-975 sensitized YAP-expressing RET-aberrant cancer cells to the RET-TKIs selpercatinib and pralsetinib. Pretreatment YAP expression in clinical specimens obtained from patients with RET fusion-positive lung cancer was associated with poor RET-TKI treatment outcomes. CONCLUSIONS The YAP-HER3 axis is crucial for the survival and adaptive resistance of high YAP-expressing RET-aberrant cancer cells treated with RET-TKIs. Combining YAP/HER3 inhibition with RET-TKIs represents a highly potent strategy for initial treatment. See related commentary by Ortiz-Cuaran and Leonce, p. 958.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-ret/metabolism
- Proto-Oncogene Proteins c-ret/genetics
- Proto-Oncogene Proteins c-ret/antagonists & inhibitors
- Receptor, ErbB-3/metabolism
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/antagonists & inhibitors
- Drug Resistance, Neoplasm
- Protein Kinase Inhibitors/pharmacology
- Signal Transduction/drug effects
- Cell Line, Tumor
- Transcription Factors/metabolism
- Transcription Factors/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- YAP-Signaling Proteins
- Pyrazoles/pharmacology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Pyridines/pharmacology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neoplasms/genetics
- Neoplasms/pathology
- Pyrimidines
Collapse
Affiliation(s)
- Yuki Katayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiko Tanimura
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hayato Kawachi
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaki Ishida
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yohei Matsui
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Soichi Hirai
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryota Nakamura
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji Morimoto
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of Medicine, Kawasaki, Japan
| | - Sachiko Arai
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Yasuhiro Goto
- Department of Respiratory Medicine and Allergies, Fujita Health University, Toyoake, Japan
| | - Yoshihiko Sakata
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kazumi Nishino
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Michiko Tsuchiya
- Department of Respiratory Medicine, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Akihiro Tamiya
- Department of Internal Medicine, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai, Japan
| | - Go Saito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Takayuki Takeda
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Koji Date
- Department of Pulmonary Medicine, Kyoto Chubu Medical Center, Nantan, Japan
| | - Yasuhito Fujisaka
- Department of Respiratory Medicine and Thoracic Oncology, Clinical Research Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Daichi Fujimoto
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Japan
| | - Hisanori Uehara
- Division of Pathology, Tokushima University Hospital, Tokushima, Japan
| | - Mano Horinaka
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Seiji Yano
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
- Department of Respiratory Medicine, Kanazawa Graduate School of Medical Sciences, Kanazawa, Japan
| | - Shinsaku Tokuda
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Yang R, Yao J, Ma H, Shui C, Li T, Zhang S, Li C. Celastrol promotes apoptotic cell death in thyroid cancer cells through a caspases-dependent pathway. Thyroid Res 2025; 18:9. [PMID: 40001245 PMCID: PMC11863774 DOI: 10.1186/s13044-024-00222-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/27/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Celastrol, a naturally occurring bioactive compound, has demonstrated potential in treating inflammation, obesity, and tumors, particularly in colorectal, gastric, and breast cancers. However, its therapeutic effects on thyroid cancer (TC), which have poor clinical outcomes, remain unclear. This study aimed to investigate Celastrol's potential in treating thyroid cancer using cell lines. METHODS The viability and proliferation of thyroid cancer cells treated with or without Celastrol were analyzed by CCK-8 and colony formation assay. The state of thyroid cancer cells treated with or without Celastrol were observed by microscopy. Further evidence from flow cytometry and TUNEL staining demonstrated the induction of apoptotic processes in thyroid cancer cells. The expression of PARP1, Caspase-3, Bax, BCL2 in thyroid cancer cells after indicated treatment was analyzed by Western blot and Caspase-3 expression in thyroid cancer cells after 12 and 24 h of Celastrol treatment was detected by immunofuorescence assay. Anaplastic thyroid cancer growth-limiting of Celastrol was evaluated in nude mice. RESULTS Celastrol induction promoted apoptotic in TC cells, increased the expression of PARP1, Bax and Caspase-3 and reduces expression of BCL2 by Western Blot. The expression of Caspase-3 was increased by immunofluorescence, which indicating that Celastrol may serve as an adjuvant therapeutic agent for thyroid cancer treatment by inducing apoptosis through the caspase-3 pathway. Celastrol treatment of mice implanted with anaplastic thyroid cancer cells also inhibited tumor growth, associated with reduced Ki-67 and increased Caspase-3. CONCLUSIONS Celastrol promotes apoptotic cell death in thyroid carcinoma cells by the Caspase-3 pathway.
Collapse
Affiliation(s)
- Ruoyi Yang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Guizhou Medical University, Guiyang, 550004, China
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Jie Yao
- Department of Centre for Translational Research in Cancer, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Hong Ma
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Guizhou Medical University, Guiyang, 550004, China
| | - Chunyan Shui
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Teng Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Guizhou Medical University, Guiyang, 550004, China
| | - Sicheng Zhang
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Chao Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Guizhou Medical University, Guiyang, 550004, China.
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
5
|
Hualong M, Liu J, Yin T, Cao X, Su Z, Zhao DG, Ma YY. Discovery of a Selective and Orally Bioavailable RET Degrader with Effectiveness in Various Mutations. J Med Chem 2025; 68:2657-2679. [PMID: 39772547 DOI: 10.1021/acs.jmedchem.4c01889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The rearranged during transfection (RET) mutation such as the G810C mutation has significantly restricted the clinical application of selective RET inhibitors in the treatment of RET-driven cancers. This study designed and evaluated RET proteolysis targeting chimeras (PROTACs) based on selpercatinib (LOXO-292), identifying RD-23 as a potent and selective RET PROTAC. RD-23 effectively inhibited the proliferation of BaF3 cells with various RET mutations, showing IC50 values of 2.4 to 6.5 nM. It selectively induced degradation of the RETG810C mutation via the ubiquitin-proteasome system, with a DC50 (concentration causing 50% of protein degradation) value of 11.7 nM. Additionally, RD-23 exhibited oral bioavailability and superior antitumor effects compared to LOXO-292 in a Ba/F3-KIF5B-RETG810C xenograft mouse model. These results suggested that RD-23 is a promising candidate for treating RET-driven cancers.
Collapse
Affiliation(s)
- Mo Hualong
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China
| | - JieYing Liu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China
| | - Ting Yin
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China
| | - XuXu Cao
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China
| | - ZhengXi Su
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China
| | - Deng-Gao Zhao
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China
| | - Yan-Yan Ma
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
6
|
Khan E, Hylton H, Rajan N, Bouley SJ, Siddiqui JK, Rajasekaran S, Koshre GR, Storts H, Valenciaga A, Khan M, Liyanarachchi S, Fernandez F, Zheng X, Phay J, Dedhia PH, Wang J, Walker JA, Ringel MD, Miles WO. Proteomic Profiling of Medullary Thyroid Cancer Identifies CAPN1 as a Key Regulator of NF1 and RET Fueled Growth. Thyroid 2025; 35:177-187. [PMID: 39868924 DOI: 10.1089/thy.2024.0102] [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] [Indexed: 01/28/2025]
Abstract
Background: Medullary thyroid cancer (MTC) is a frequently metastatic tumor of the thyroid that develops from the malignant transformation of C-cells. These tumors most commonly have activating mutations within the RET or RAS proto-oncogenes. Germline mutations within RET result in C-cell hyperplasia, and cause the MTC pre-disposition disorder, multiple endocrine neoplasia, type 2A (MEN2A). Single-agent therapies for MTC, including vandetanib (VAN) and cabozantinib for all MTCs and selpercatinib (SEL) for RET-mutated MTC, lead to partial responses but are not curative. Methods: To identify new therapeutic targets for MTC, we conducted proteomic profiling of normal C-cells, MTC cells, pre-malignant MEN2A patient samples, and MTC tumors. Results: From this analysis, we identified CAPN1, a member of the CALPAIN (CAPN) family endopeptidases, as widely upregulated in MTC samples. We found that short hairpin RNA-mediated depletion of CAPN1 or inhibitors of CAPN1 significantly reduced MTC cell growth, colony formation, and xenograft tumor growth in vivo. In addition, we show that CAPN1 inhibitors synergize with VAN and SEL in vitro, maximizing apoptosis. Mechanistic experiments implicate CAPN1 in inhibiting neurofibromin, encoded by NF1, and CAPN1 inhibitors stabilize NF1 protein levels and diminish downstream RAS/RET activation of AKT and ERK. Conclusions: Our data suggest that increased CAPN1 levels support RET and RAS-fueled growth by reducing NF1 levels. We find that combinatorial therapies between CAPN1 inhibitors and VAN or SEL show maximal efficacy in MTC cells.
Collapse
Affiliation(s)
- Eshan Khan
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Hannah Hylton
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Neel Rajan
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Stephanie J Bouley
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jalal K Siddiqui
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Swetha Rajasekaran
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ganesh R Koshre
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Hayden Storts
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Anisley Valenciaga
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Misbah Khan
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Sandya Liyanarachchi
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Francisco Fernandez
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xuguang Zheng
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - John Phay
- Division of Surgical Oncology, Ohio State University Comprehensive Cancer Center and Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Priya H Dedhia
- Division of Surgical Oncology, Ohio State University Comprehensive Cancer Center and Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jing Wang
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - James A Walker
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew D Ringel
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Molecular Medicine and Therapeutics, The Ohio State University, Columbus, Ohio, USA
| | - Wayne O Miles
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Clifton-Bligh RJ. Mechanisms of resistance to RET-directed therapies. Endocr Relat Cancer 2025; 32:e240224. [PMID: 39655713 PMCID: PMC11798414 DOI: 10.1530/erc-24-0224] [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: 08/25/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
The association between RET and multiple endocrine neoplasia type 2 was established in 1993 and remains one of the very few oncogenes for which distinct phenotypes (medullary thyroid cancer or pheochromocytoma) are associated with the same hot-spot variants occurring in either germline or somatic DNA. Somatic RET fusion events have also been described in several cancers, including papillary thyroid cancer, non-small-cell lung cancer, breast cancer, salivary gland cancer and pancreatic cancer. Highly selective RET inhibitors have improved outcomes in RET-altered cancers and have been well-tolerated. Nevertheless, primary and acquired drug resistance has been observed, arising from distinct genomic alterations either in RET (on-target resistance) or via alternate oncogenic pathways (bypass resistance). The same mechanisms of resistance have been observed across multiple cancer types, which implies RET-altered cancers evolve away from RET addiction via stochastic subclonal events. Understanding these mechanisms is crucial for identifying therapeutic opportunities to overcome resistance. Successful treatment targeting bypass oncogenes has been reported in several instances, at least for short-term outcomes; in contrast, although several compounds have been reported to overcome on-target RET alterations, none have yet been translated into routine clinical practice and this remains an area of urgent clinical need.
Collapse
Affiliation(s)
- Roderick J Clifton-Bligh
- Cancer Genetics, Kolling Institute, Royal North Shore Hospital and University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Wang Y, Hu X, Pandey S, Khatri U, Shen T, Subbiah V, Mooers BHM, Chao T, Wang S, Yu H, Sun X, Wu J, Cai J. Targeting Oncogenic RET Kinase by Simultaneously Inhibiting Kinase Activity and Degrading the Protein. J Med Chem 2025; 68:81-94. [PMID: 39723919 DOI: 10.1021/acs.jmedchem.4c01424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
The rearranged-during-transfection (RET) kinase is a validated target for the treatment of RET-altered cancers. Currently approved RET-selective kinase inhibitors, selpercatinib (LOXO-292) and pralsetinib (BLU-667), increase the oncogenic RET protein level upon treatment, which may affect their efficacy. We seek to reduce the oncogenic RET protein level and RET kinase activity simultaneously. Here, we report the development of proteolysis targeting chimera (PROTAC) degraders of oncogenic RET protein. Compound YW-N-7 exhibited dual action of selectively inhibiting and depleting RET protein both in vitro and in vivo. Proteomic analysis indicated that YW-N-7 is highly specific to RET. In cell cultures, reducing RET fusion protein potentiated the activity of LOXO-292. Furthermore, YW-N-7 showed significant activity in inhibiting KIF5B-RET-driven xenograft tumors in animals. This study exemplifies the feasibility of simultaneously inhibiting and degrading oncogenic RET kinase for cancer therapy.
Collapse
Affiliation(s)
- Yafeng Wang
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xueqing Hu
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Shriya Pandey
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Ujjwol Khatri
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Tao Shen
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, Tennessee 37203, United States
| | - Blaine H M Mooers
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Ting Chao
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Huaxuan Yu
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Jie Wu
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| |
Collapse
|
9
|
Zhang Q, He Y, Rao D, He R, Yu L, Sun Y, Lai Y, Shi Z, Peng L, Zhang Z, Xu S. Discovery of an Efficacious RET PROTAC Degrader with Enhanced Antiproliferative Activity against Resistant Cancer Cells Harboring RET Solvent-Front Mutations. J Med Chem 2025; 68:753-775. [PMID: 39731581 DOI: 10.1021/acs.jmedchem.4c02692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
Rearranged during transfection (RET) kinase is a validated therapeutic target for various cancers characterized by RET alterations. Although two selective RET inhibitors, selpercatinib and pralsetinib, have been approved by the FDA, acquired resistance through solvent-front mutations has been identified rapidly. Developing proteolysis targeting chimera (PROTAC) targeting RET mutations offers a promising strategy to combat drug resistance. Herein, we describe the design, synthesis, and evaluation of a series of RET PROTAC degraders. The representative compound QZ2135 (20) effectively degraded RET kinase and its resistant mutants, such as V804M and G810C/R. It also exhibited superior antiproliferative activity against Ba/F3 cells stably expressing oncogenic fusions of RET with solvent-front mutants, including G810C/R/S, compared to its parental inhibitor. Notably, QZ2135 demonstrated in vivo antitumor efficacy in a Ba/F3-KIF5B-RET-G810C xenograft mouse model. Together, this study provides a potential alternative strategy for overcoming acquired resistance to RET inhibitors mediated by solvent-front mutations.
Collapse
Affiliation(s)
- Qian Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yingqi He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Danni Rao
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yaoliang Sun
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuanhui Lai
- Department of Thyroid and Breast Surgery, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510310, China
| | - Zihan Shi
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lijie Peng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhang Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
- Department of Thyroid and Breast Surgery, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510310, China
| | - Shilin Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Khatri U, Gouda MA, Pandey S, Chauhan NK, Shen T, Hu X, Li M, Huang S, Subbiah V, Wu J. Selpercatinib mitigates cancer cachexia independent of anti-tumor activity in the HT1080 tumor model. Cancer Lett 2025; 611:217444. [PMID: 39778760 DOI: 10.1016/j.canlet.2025.217444] [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: 07/29/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Anorexia is a major cause of cancer cachexia and is induced by growth differentiation factor-15 (GDF15), which activates the rearranged during transfection (RET) protein tyrosine kinase in the hindbrain through GDF family receptor α-like (GFRAL), raising the possibility of targeting RET for cancer cachexia treatment. RET-altered cancer patients treated with RET-selective kinase inhibitors gain weight, however, it is unclear whether this results from tumor regression that improves the overall health of patients. Thus, the potential of using a RET inhibitor to address cancer cachexia remains unknown. Using a RET-negative tumor model, we evaluated the activity of the RET-selective inhibitor selpercatinib (LOXO-292) against cancer cachexia. In tumor-bearing animals, selpercatinib significantly increased food consumption, skeletal muscle mass and strength, adipose tissues, and body temperature, as well as reducing body weight loss, without significantly affecting tumor growth. Transcriptomes of skeletal muscle from mock-treated tumor-bearing mice were enriched in starvation and muscle atrophy genes, whereas those from selpercatinib-treated mice were enriched in myoblast proliferation, gluconeogenesis, and insulin receptor signaling genes. In parallel, retrospective analysis of weight gain in selpercatinib-treated patients showed that weight gain was not correlated with tumor response to selpercatinib. Our data demonstrate that selpercatinib could alleviate anorexia and cancer cachexia in an animal model and that weight gain in selpercatinib-treated patients is not dependent on tumor regression. These results identify a RET inhibitor as the first protein tyrosine kinase inhibitor for mitigating cancer cachexia.
Collapse
Affiliation(s)
- Ujjwol Khatri
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shriya Pandey
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Neeraj K Chauhan
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tao Shen
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xueqing Hu
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Min Li
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Suming Huang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, TN, USA.
| | - Jie Wu
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
11
|
Gil-Bernabé S, García-DeLaFuente L, García-Álvarez A, García-Rostán G, Capdevila J, Hernando J. Genomics Review of Selective RET Inhibitors Sensitivity in Thyroid Cancer Clinical Trials. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2025:e32127. [PMID: 39754491 DOI: 10.1002/ajmg.c.32127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025]
Abstract
RET gene is a driver of thyroid cancer (TC) tumorigenesis. The incidence of TC has increased worldwide in the last few decades, both in medullary and follicular-derived subtypes. Several drugs, including multikinase and selective inhibitors, have been explored. Selpercatinib and pralsetinib are selective RET inhibitors that have shown clear clinical benefits for patients in the LIBRETTO and ARROW trials, respectively. Currently, their development and application in clinical practice are ongoing. However, its efficacy in different RET pathogenic variants has not yet been well established. Although selpercatinib and pralsetinib achieved a high ORR, no data are available regarding the differences in tumor responses of both TC groups according to RET pathogenic variants. Clinical trials and literature have analyzed the efficacy of selective RET inhibitors with a special interest in the most common variants. A review of LIBRETTO and ARROW trials was made regarding the change in tumor size depending on the pathogenic variants. M918T pathogenic variant resulted in a higher complete response rate. Patients who underwent fusion had the highest ORR (objective response rate). MKi-treated patients did not exhibit significant differences from untreated patients. Different RET pathogenic variants are not biomarkers of RETi response in TC. Selpercatinib showed a tendency to achieve a complete response. All patients with RET pathogenic variants should receive treatment with selpercatinib or pralsetinib at any moment of the therapeutic schedule owing to off-target inhibition and toxicity. Therefore, new targets for drug sensitivity and resistance should be explored.
Collapse
Affiliation(s)
- Sara Gil-Bernabé
- Pathology Department, Faculty of Medicine, Valladolid University, Valladoli, Spain
- Group Pathobiology of Cancer: Inter-, Intra-Tumor Heterogeneity and Molecular Targets, Institute of Molecular Genetics and Biomedicine (IBGM), Valladoli, Spain
| | | | - Alejandro García-Álvarez
- Gastrointestinal and Endocrine Tumor Unit, Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Ginesa García-Rostán
- Pathology Department, Faculty of Medicine, Valladolid University, Valladoli, Spain
- Group Pathobiology of Cancer: Inter-, Intra-Tumor Heterogeneity and Molecular Targets, Institute of Molecular Genetics and Biomedicine (IBGM), Valladoli, Spain
| | - Jaume Capdevila
- Gastrointestinal and Endocrine Tumor Unit, Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Jorge Hernando
- Gastrointestinal and Endocrine Tumor Unit, Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
12
|
Jara MA, Castroneves LA. Overview of management and therapeutic advances in medullary thyroid cancer. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2025; 5:e240077. [PMID: 40084047 PMCID: PMC11906152 DOI: 10.1530/eo-24-0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/16/2025]
Abstract
Medullary thyroid carcinoma (MTC) is a rare cancer of the thyroid's calcitonin-producing C cells. This review covers recent advances in MTC treatment, emphasizing surgical and systemic therapies. For localized MTC, surgery remains the primary and most effective treatment, with total thyroidectomy and lymph node dissection providing the highest potential for cure. However, prognosis worsens significantly with local and distant metastases, underscoring the importance of early diagnosis and intervention. MTC can be sporadic or hereditary, with the latter associated with germline RET proto-oncogene mutations linked to multiple endocrine neoplasia types 2A and 2B. Genetic discoveries have enabled preventive measures such as prophylactic thyroidectomy, increasing the cure rate of hereditary cases. Since 2011, systemic treatment options have expanded with multikinase inhibitors (MKIs), such as vandetanib and cabozantinib, and selective RET inhibitors such as selpercatinib and pralsetinib. MKIs extend progression-free survival in advanced cases by targeting tumor growth and angiogenesis but can cause off-target effects. RET inhibitors offer precision treatment for RET-mutated tumors, showing high efficacy and fewer side effects, though resistance to these inhibitors has emerged, and current research focuses on developing next-generation inhibitors to overcome these barriers. Effective MTC management, particularly given its rarity, benefits from specialized high-volume centers. Precision medicine, standardized therapy selection and ongoing research are essential for improving outcomes in both RET-positive and RET-negative MTC patients.
Collapse
Affiliation(s)
- Mark A Jara
- Department of Endocrinology and Metabolism. University of Miami, Miami, Florida, USA
| | - Luciana Audi Castroneves
- Department of Endocrinology. Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Shen J, Liu J, Tan Z, Li A, Chen S, Li Y. Structure-Based Design of 2-Aminopyrazolpyrimidopyridone Derivatives as New Rearranged During Transfection (RET) Kinase Inhibitors. Chem Biol Drug Des 2025; 105:e70039. [PMID: 39739433 DOI: 10.1111/cbdd.70039] [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: 09/23/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
RET (Rearranged during transfection) kinase is a validated target for non-small cell lung cancer (NSCLC). In 2020, two selective RET inhibitors, selpercatinib and pralsetinib were approved by the US FDA. However, high treatment costs and clinically acquired resistance (e.g., G810C/S/R) become the new challenges for RET-based therapies. In this work, we discovered a series of 2-aminopyrazolpyrimidopyridone RET inhibitors to overcome the V804M and G810C resistant mutations. One of the compounds, 8w, exhibited inhibitory potency against the BaF3 cells harboring CCDC6-RETV804M mutation with an IC50 value of 0.715 μM. The compound also dose-dependently suppressed the activation of RET and downstream signals. Another compound, 8s suppressed BaF3 cells harboring CCDC6-RETG810C mutation with an IC50 value of 2.91 μM. However, the poor solubility of these compounds will limit their further development. Therefore, compound 8w and 8s might be promising lead compounds for the development of novel RETV804M and RETG810C inhibitors overcoming the clinically acquired resistance.
Collapse
Affiliation(s)
- Jiayi Shen
- Jiangxi Provincial Key Laboratory of Synthetic Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, China
| | - Jihu Liu
- Jiangxi Provincial Key Laboratory of Synthetic Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, China
| | - Zhiyong Tan
- Jiangxi Provincial Key Laboratory of Synthetic Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, China
| | - Anzhi Li
- Jiangxi Provincial Key Laboratory of Synthetic Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, China
| | - Sheng Chen
- Jiangxi ChiralSyn Biological Medicine Co. Ltd, Nanchang, China
| | - Yongdong Li
- Jiangxi Provincial Key Laboratory of Synthetic Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, China
| |
Collapse
|
14
|
Santillan MR, Dadu R, Gagel RF, Grubbs EG, Hu MI. Systemic Therapies for Advanced Medullary Thyroid Carcinoma. Recent Results Cancer Res 2025; 223:293-307. [PMID: 40102263 DOI: 10.1007/978-3-031-80396-3_12] [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] [Indexed: 03/20/2025]
Abstract
Medullary thyroid carcinoma (MTC) is a rare disease that is indolent in the majority of patients. In a subset of patients, the cancer is more aggressive with symptomatic or progressive disease metastasizing to cervical neck structures, lungs, liver, and/or bones. Definitive cure for metastatic MTC remains elusive. Understanding oncogenic pathways and molecular drivers of disease have led to development and approval of multikinase and highly-specific RET inhibitors for the management of progressive MTC. RET mutations are the most common drivers in MTC, followed by mutually exclusive RAS mutations. Cabozantinib and vandetanib, multikinase inhibitors (MKIs) that exert their therapeutic effect mainly through antiangiogenesis by targeting the vascular endothelial growth factor receptor, have mild anti-RET activity. Despite conveying clinical responses in MTC, MKIs have significant off-target activity causing marked toxicities limiting their effectiveness. Potent and selective RET inhibitors, selpercatinib and pralsetinib, demonstrate significant efficacy in RET-altered cancers and more tolerable side effect profiles than MKIs. However, durable responses can be limited by the acquisition of mutations which confer drug resistance to available treatments. Thus, development of more effective treatments for advanced, progressive MTC remains an urgent priority. In this chapter, we describe the current spectrum of systemic therapies for MTC, their limitations, and ongoing investigations.
Collapse
Affiliation(s)
- Marco Ruiz Santillan
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert F Gagel
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth G Grubbs
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mimi I Hu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Wu J, Mo H, An Z, Tang Z, Deng X, Zhou H, Gong Y, Zheng C, Zhuo L, Tan S. Discovery of 7-(1-methyl-1H-pyrazol-4-yl)-1,6-naphthyridine derivatives as potent inhibitors of rearranged during transfection (RET) and RET solvent-front mutants for overcoming selpercatinib resistance. Eur J Med Chem 2024; 279:116891. [PMID: 39316846 DOI: 10.1016/j.ejmech.2024.116891] [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: 07/31/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Rearranged during transfection kinase (RET) inhibition has been considered a promising therapeutic approach for treatment of a variety of cancers. However, the clinical therapeutic benefits of the second-generation RET inhibitor selpercatinib are greatly compromised by acquired resistance mediated by solvent-front mutations (e.g., RETG810 R/S/C). Herein, we report a class of 7-(1-methyl-1H-pyrazol-4-yl)-1,6-naphthyridine derivatives as potent RET and RET solvent-front mutant inhibitors for overcoming selpercatinib resistance. The representative compound 20p exhibited excellent in vitro inhibitory activities against solvent-front mutations (RETG810R, RETG810S, and RETG810C) with low nanomolar range (IC50 of 5.7-8.3 nM), which was 15-29-fold more potent than selpercatinib (IC50 of 95.3-244.1 nM). Additionally, 20p exhibited acceptable pharmacokinetic properties with oral bioavailability of 30.4 %. Importantly, 20p exhibited highly impressive antitumor potency in both a Ba/F3-KIF5B-RETWT-derived xenograft mouse model and a selpercatinib-resistant Ba/F3-KIF5B-RETG810R-positive mutant xenograft mouse model. Overall, 20p represents a novel and promising drug lead for overcoming RET solvent-front mutation-based resistance to selpercatinib.
Collapse
Affiliation(s)
- Junbo Wu
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China
| | - Hanxuan Mo
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhigang An
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zishu Tang
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xinyu Deng
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Huifang Zhou
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China
| | - Yi Gong
- National Key Laboratory of Green Pesticide, Central China Normal University, Wuhan, 430079, China
| | - Chenggong Zheng
- Pulmonary Hospital, Changsha Central Hospital, Changsha, Hunan, 410004, China
| | - Linsheng Zhuo
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Shuguang Tan
- Department of Colorectal Surgery, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China.
| |
Collapse
|
16
|
Qiao JX, Williams D, Gill P, Li L, Norris D, Tokarski JS, Wong J, Qi H, Hafeji Y, Downes DP, Degnen B, Wang YK, Locke G, Fang H, Yu F, Xu S, Naglich J, Zhang J, Nanjappa P, Dai C, Chourb L, Napoline J, Tester R, Jorge C, Li YX, Mathur A, Barbieri C, Soars MG, Venkatanarayan A, Lees E, Borzilleri RM, Gavai AV, Wichroski M, Dhar TGM. Discovery and Synthesis of Heterobifunctional Degraders of Rearranged during Transfection (RET) Kinase. J Med Chem 2024; 67:19736-19754. [PMID: 39437163 DOI: 10.1021/acs.jmedchem.4c02083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
We describe the design, synthesis, and structure-activity relationship (SAR) of heterobifunctional RET ligand-directed degraders (LDDs) derived from three different second-generation RET inhibitors. These LDDs are composed of a target binding motif (TBM) that binds to the RET protein, a linker, and a cereblon binding motif (CBM) as the E3 ligase recognition unit. This led to the identification of a series of pyrazolopyridine-based heterobifunctional LDDs, as exemplified by compound 39. LDD 39 demonstrated high in vitro inhibitory and degradation potency against both RET wild-type and the two representative mutants, V804M and G810R. Importantly, in PK/PD studies, 39 exhibited a differentiated and favorable in vivo profile compared to the corresponding tyrosine kinase inhibitor (TKI), compound 3. Robust and sustained degradation of total-RET (tRET) protein and inhibition of phospho-RET (pRET) signaling were observed in TPC-1 xenograft tumors driven by RET and the RET/G810R mutant following a single dose of LDD 39 at 15 and 75 mg/kg, respectively.
Collapse
Affiliation(s)
- Jennifer X Qiao
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - David Williams
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Patrice Gill
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Ling Li
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Derek Norris
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - John S Tokarski
- Molecular Structure & Design, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Jessica Wong
- Oncology Discovery Biology, Mechanism of Cancer Resistance, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Huilin Qi
- Oncology Discovery Biology, Mechanism of Cancer Resistance, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Yamnah Hafeji
- Oncology Discovery Biology, Mechanism of Cancer Resistance, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Daniel P Downes
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Bill Degnen
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Ying-Kai Wang
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Gregory Locke
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Hua Fang
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Fei Yu
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Songmei Xu
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Joseph Naglich
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Jun Zhang
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Purushothama Nanjappa
- Discovery Pharmacology and in vivo Biology, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Chao Dai
- Discovery Pharmacology and in vivo Biology, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Lisa Chourb
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Jonathan Napoline
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Richland Tester
- Department of Discovery Synthesis, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Christine Jorge
- Department of Discovery Synthesis, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Yi-Xin Li
- Department of Discovery Synthesis, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Arvind Mathur
- Department of Discovery Synthesis, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Christopher Barbieri
- Leads Discovery & Optimization, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Matthew G Soars
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Avinashnarayan Venkatanarayan
- Oncology Discovery Biology, Mechanism of Cancer Resistance, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Emma Lees
- Oncology Discovery Biology, Mechanism of Cancer Resistance, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - Robert M Borzilleri
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Ashvinikumar V Gavai
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| | - Michael Wichroski
- Oncology Discovery Biology, Mechanism of Cancer Resistance, Bristol Myers Squibb, Cambridge, Massachusetts 02141, United States
| | - T G Murali Dhar
- Discovery Chemistry, Bristol Myers Squibb, Princeton, New Jersey 08540, United States
| |
Collapse
|
17
|
da Silva DD, Araldi RP, Belizario MR, Rocha WG, Maciel RMDB, Cerutti JM. DLK1 Is Associated with Stemness Phenotype in Medullary Thyroid Carcinoma Cell Lines. Int J Mol Sci 2024; 25:11924. [PMID: 39595993 PMCID: PMC11594232 DOI: 10.3390/ijms252211924] [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: 09/30/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Medullary thyroid carcinoma (MTC) is a rare and aggressive tumor, often requiring systemic treatment in advanced or metastatic stages, where drug resistance presents a significant challenge. Given the role of cancer stem cells (CSCs) in cancer recurrence and drug resistance, we aimed to identify CSC subpopulations within two MTC cell lines harboring pathogenic variants in the two most common MEN2-associated codons. We analyzed 15 stemness-associated markers, along with well-established thyroid stem cell markers (CD133, CD44, and ALDH1), a novel candidate (DLK1), and multidrug resistance proteins (MRP1 and MRP3). The ability to efflux the fluorescent dye Hoechst 3342 and form spheroids, representing CSC behavior, was also assessed. MZ-CRC-1 cells (p.M918T) displayed higher expressions of canonical markers, DLK1, and MRP proteins than TT cells (p.C634W). MZ-CRC-1 cells also formed more spheroids and showed less dye accumulation (p < 0.0001). Finally, we observed that DLK1+ cells (those expressing DLK1) in both cell lines exhibited significantly higher levels of stemness markers compared to DLK1- cells (those lacking DLK1 expression). These findings underscore DLK1's role in enhancing the stemness phenotype, providing valuable insights into MTC progression and resistance and suggesting potential therapeutic implications.
Collapse
Affiliation(s)
- Danilo Dias da Silva
- Genetic Bases of Thyroid Tumour Laboratory, Division of Genetics, Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil; (D.D.d.S.); (R.P.A.); (M.R.B.); (W.G.R.)
| | - Rodrigo Pinheiro Araldi
- Genetic Bases of Thyroid Tumour Laboratory, Division of Genetics, Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil; (D.D.d.S.); (R.P.A.); (M.R.B.); (W.G.R.)
| | - Mariana Rocha Belizario
- Genetic Bases of Thyroid Tumour Laboratory, Division of Genetics, Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil; (D.D.d.S.); (R.P.A.); (M.R.B.); (W.G.R.)
| | - Welbert Gomes Rocha
- Genetic Bases of Thyroid Tumour Laboratory, Division of Genetics, Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil; (D.D.d.S.); (R.P.A.); (M.R.B.); (W.G.R.)
| | - Rui Monteiro de Barros Maciel
- Laboratório de Endocrinologia Molecular e Translacional, Disciplina de Endocrinologia e Metabologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil;
| | - Janete Maria Cerutti
- Genetic Bases of Thyroid Tumour Laboratory, Division of Genetics, Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil; (D.D.d.S.); (R.P.A.); (M.R.B.); (W.G.R.)
| |
Collapse
|
18
|
Lasolle H, Borson-Chazot F, Gauduchon T, Haissaguerre M, Illouz F, Lifante JC, Lussey-Lepoutre C, Prunier D, Sajous C, Varnier R, Hadoux J. La prise en charge des cancers médullaires de la thyroïde en 2024. Bull Cancer 2024; 111:10S53-10S63. [PMID: 39505437 DOI: 10.1016/s0007-4551(24)00408-9] [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] [Indexed: 11/08/2024]
Abstract
MANAGING MEDULLARY THYROID CARCINOMA IN 2024: Medullary thyroid carcinoma is a rare neuroendocrine thyroid cancer with a heterogeneous prognosis which has the particularity of being associated with a RET gene mutation, germline in 20-25% of cases in the context of multiple endocrine neoplasia type 2 (NEM2), and somatic in 70% of sporadic cases. It is often diagnosed on a thyroid nodule or in the context of genetic screening. Calcitonin is a biological marker, used for diagnosis, monitoring of therapeutic response and prognostic evaluation. The only curative treatment is surgery for localized disease. The extent must be carefully assessed, particularly in terms of calcitonin levels and imaging, and carried out by an expert surgeon. The prognosis of locally advanced or metastatic disease is highly heterogeneous. Histological factors, such as high grade, or biological factors, such as calcitonin doubling time, can help assess prognosis. The development of multi-kinase inhibitors cabonzantinib and vandetanib, and RET-targeted inhibitors selpercatinib, has completely changed the therapeutic arsenal for advanced disease, but their prescription is reserved to progressive disease with high tumor volume or to symptomatic disease inaccessible to local treatment in expert centers from the ENDOCAN-TUTHYREF network. Active surveillance is the alternative of choice for slowly progressing disease.
Collapse
Affiliation(s)
- Hélène Lasolle
- Service d'endocrinologie, Hôpital Louis-Pradel, Hospices civils de Lyon, Bron cedex, France; University Lyon I, Lyon, France; Inserm U 1052, CRCL.
| | - Françoise Borson-Chazot
- Service d'endocrinologie, Hôpital Louis-Pradel, Hospices civils de Lyon, Bron cedex, France; University Lyon I, Lyon, France
| | | | | | | | - Jean-Christophe Lifante
- Service de chirurgie endocrinienne, Hôpital Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, France; University Lyon I, Lyon, France
| | | | - Delphine Prunier
- Service de biochimie et biologie moléculaire, CHU d'Angers, France
| | - Christophe Sajous
- Service d'endocrinologie, Hôpital Louis-Pradel, Hospices civils de Lyon, Bron cedex, France
| | | | - Julien Hadoux
- Service d'oncologie endocrinienne, Département d'imagerie, Gustave-Roussy, F-94805 Villejuif, France
| |
Collapse
|
19
|
Gigliotti BJ, Brooks JA, Wirth LJ. Fundamentals and recent advances in the evaluation and management of medullary thyroid carcinoma. Mol Cell Endocrinol 2024; 592:112295. [PMID: 38871174 DOI: 10.1016/j.mce.2024.112295] [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: 02/11/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/15/2024]
Abstract
Medullary thyroid carcinoma (MTC) is a rare primary neuroendocrine thyroid carcinoma that is distinct from other thyroid or neuroendocrine cancers. Most cases of MTC are sporadic, although MTC exhibits a high degree of heritability as part of the multiple endocrine neoplasia syndromes. REarranged during Transfection (RET) mutations are the primary oncogenic drivers and advances in molecular profiling have revealed that MTC is enriched in druggable alterations. Surgery at an early stage is the only chance for cure, but many patients present with or develop metastases. C-cell-specific calcitonin trajectory and structural doubling times are critical biomarkers to inform prognosis, extent of surgery, likelihood of residual disease, and need for additional therapy. Recent advances in the role of active surveillance, regionally directed therapies for localized disease, and systemic therapy with multi-kinase and RET-specific inhibitors for progressive/metastatic disease have significantly improved outcomes for patients with MTC.
Collapse
Affiliation(s)
| | - Jennifer A Brooks
- Department of Otolaryngology Head & Neck Surgery, University of Rochester, Rochester, NY, USA.
| | - Lori J Wirth
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Zhang Y, Zheng WH, Zhou SH, Gu JL, Yu Q, Zhu YZ, Yan YJ, Zhu Z, Shang JB. Molecular genetics, therapeutics and RET inhibitor resistance for medullary thyroid carcinoma and future perspectives. Cell Commun Signal 2024; 22:460. [PMID: 39342195 PMCID: PMC11439284 DOI: 10.1186/s12964-024-01837-x] [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: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Medullary thyroid carcinoma (MTC) is a rare type of thyroid malignancy that accounts for approximately 1-2% of all thyroid cancers (TCs). MTC include hereditary and sporadic cases, the former derived from a germline mutation of rearrangement during transfection (RET) proto-oncogene, whereas somatic RET mutations are frequently present in the latter. Surgery is the standard treatment for early stage MTC, and the 10-year survival rate of early MTC is over 80%. While for metastatic MTC, chemotherapy showing low response rate, and there was a lack of effective systemic therapies in the past. Due to the high risk (ca. 15-20%) of distant metastasis and limited systemic therapies, the 10-year survival rate of patients with advanced MTC was only 10-40% from the time of first metastasis. Over the past decade, targeted therapy for RET has developed rapidly, bringing hopes to patients with advanced and progressive MTC. Two multi-kinase inhibitors (MKIs) including Cabozantinib and Vandetanib have been shown to increase progression-free survival (PFS) for patients with metastatic MTC and have been approved as choices of first-line treatment. However, these MKIs have not prolonged overall survival (OS) and their utility is limited due to high rates of off-target toxicities. Recently, new generation TKIs, including Selpercatinib and Pralsetinib, have demonstrated highly selective efficacy against RET and more favorable side effect profiles, and gained approval as second-line treatment options. Despite the ongoing development of RET inhibitors, the management of advanced and progressive MTC remains challenging, drug resistance remains the main reason for treatment failure, and the mechanisms are still unclear. Besides, new promising therapeutic approaches, such as novel drug combinations and next generation RET inhibitors are under development. Herein, we overview the pathogenesis, molecular genetics and current management approaches of MTC, and focus on the recent advances of RET inhibitors, summarize the current situation and unmet needs of these RET inhibitors in MTC, and provide an overview of novel strategies for optimizing therapeutic effects.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei-Hui Zheng
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shi-Hong Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jia-Lei Gu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, Zhejiang, China
| | - Qing Yu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yi-Zhou Zhu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yu-Jie Yan
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Jin-Biao Shang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China.
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Maturi A, Sastry KNV, Kumar S, Pogaku V, Kwon HJ, Ahn SM, Kim MH. Side Chain Investigation of Imidazopyridazine as a Hinge Binder for Targeting Actionable Mutations of RET Kinase. ACS Med Chem Lett 2024; 15:1566-1574. [PMID: 39291010 PMCID: PMC11403754 DOI: 10.1021/acsmedchemlett.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Actionable mutations of RET kinase have been identified as oncogenic drivers of solid tumors, including thyroid cancer, metastatic colorectal cancer, and nonsmall cell lung cancer. Although multikinase inhibitors and RET selective inhibitors are used to treat patients with RET alterations, there is insufficient research addressing certain issues: which actionable mutations arise from these therapies, how to improve the clinical response rate to RET inhibitors, and how to design new inhibitors to overcome drug resistance. Therefore, the development of sophisticated tool compounds is required to investigate the molecular mechanisms of actionable mutations and to develop breakthrough therapeutics for different RET alterations. Herein, we present our investigation into the side chains of imidazopyridazine hinge binders that are capable of inducing protein-ligand interaction patterns from the gatekeeper to the waterfront regions. Extending the substituents at the second and sixth positions enhanced the IC50 up to < 0.5 nM for diverse RET alterations.
Collapse
Affiliation(s)
- Arunkranthi Maturi
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kasinathuni Naga Visweswara Sastry
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Surendra Kumar
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Vinay Pogaku
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Yeonsu-gu, Incheon 21936, Republic of Korea
| | | | - Sung-Min Ahn
- Gachon Institute of Genome Medicine and Sciences, Gachon University Gil Medical Center, Incheon 21936, Republic of Korea
- Immunoforge, Seoul 08591, Republic of Korea
| | - Mi-Hyun Kim
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Yeonsu-gu, Incheon 21936, Republic of Korea
| |
Collapse
|
22
|
Chen MF, Repetto M, Wilhelm C, Drilon A. RET Inhibitors in RET Fusion-Positive Lung Cancers: Past, Present, and Future. Drugs 2024; 84:1035-1053. [PMID: 38997570 DOI: 10.1007/s40265-024-02040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2024] [Indexed: 07/14/2024]
Abstract
While activating RET fusions are identified in various cancers, lung cancer represents the most common RET fusion-positive tumor. The clinical drug development of RET inhibitors in RET fusion-positive lung cancers naturally began after RET fusions were first identified in patient tumor samples in 2011, and thereafter paralleled drug development in RET fusion-positive thyroid cancers. Multikinase inhibitors were initially tested with limited efficacy and substantial toxicity. RET inhibitors were then designed with improved selectivity, central nervous system penetrance, and activity against RET fusions and most RET mutations, including resistance mutations. Owing their success to these rationally designed features, the first-generation selective RET tyrosine kinase inhibitors (TKIs) had higher response rates, more durable disease control, and an improved safety profile compared to the multikinase inhibitors. This led to lung and thyroid cancer, and later tumor-agnostic regulatory approvals. While next-generation RET TKIs were designed to abrogate uncommon on-target (e.g., solvent front mutation) resistance to selpercatinib and pralsetinib, many of these drugs lacked the selectivity of the first-generation TKIs, raising the question of what the future holds for drug development in RET-dependent cancers.
Collapse
Affiliation(s)
- Monica F Chen
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Matteo Repetto
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Clare Wilhelm
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Alexander Drilon
- Thoracic Oncology, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
23
|
Spitaleri G, Trillo Aliaga P, Attili I, Del Signore E, Corvaja C, Pellizzari G, Katrini J, Passaro A, de Marinis F. Non-Small-Cell Lung Cancers (NSCLCs) Harboring RET Gene Fusion, from Their Discovery to the Advent of New Selective Potent RET Inhibitors: "Shadows and Fogs". Cancers (Basel) 2024; 16:2877. [PMID: 39199650 PMCID: PMC11352804 DOI: 10.3390/cancers16162877] [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: 07/16/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
RET fusions are relatively rare in Non-Small-Cell Lung Cancers (NSCLCs), being around 1-2% of all NSCLCs. They share the same clinical features as the other fusion-driven NSCLC patients, as follows: younger age, adenocarcinoma histology, low exposure to tobacco, and high risk of spreading to the brain. Chemotherapy and immunotherapy have a low impact on the prognosis of these patients. Multitargeted RET inhibitors have shown modest activity jeopardized by high toxicity. New potent and selective RET inhibitors (RET-Is) (pralsetinib and selpercatinib) have achieved a higher efficacy minimizing the known toxicities of the multitargeted agents. This review will describe the sensitivity of immune-checkpoint inhibitors (ICIs) in RET fusion + NSCLC patients, as well their experiences with the 'old' multi-targeted RET inhibitors. This review will focus on the advent of new potent and selective RET-Is. We will describe their efficacy as well as the main mechanisms of resistance to them. We will further proceed to deal with the new drugs and strategies proposed to overcome the resistance to RET-Is. In the last section, we will also focus on the safety profile of RET-Is, dealing with the main toxicities as well as the rare but severe adverse events.
Collapse
Affiliation(s)
- Gianluca Spitaleri
- Division of Thoracic Oncology, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Pamela Trillo Aliaga
- Division of Thoracic Oncology, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Ilaria Attili
- Division of Thoracic Oncology, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Ester Del Signore
- Division of Thoracic Oncology, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Carla Corvaja
- Division of Thoracic Oncology, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Gloria Pellizzari
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Jalissa Katrini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, 20141 Milan, Italy
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Filippo de Marinis
- Division of Thoracic Oncology, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
24
|
Leighow SM, Reynolds JA, Sokirniy I, Yao S, Yang Z, Inam H, Wodarz D, Archetti M, Pritchard JR. Programming tumor evolution with selection gene drives to proactively combat drug resistance. Nat Biotechnol 2024:10.1038/s41587-024-02271-7. [PMID: 38965430 DOI: 10.1038/s41587-024-02271-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 05/06/2024] [Indexed: 07/06/2024]
Abstract
Most targeted anticancer therapies fail due to drug resistance evolution. Here we show that tumor evolution can be reproducibly redirected to engineer therapeutic opportunity, regardless of the exact ensemble of pre-existing genetic heterogeneity. We develop a selection gene drive system that is stably introduced into cancer cells and is composed of two genes, or switches, that couple an inducible fitness advantage with a shared fitness cost. Using stochastic models of evolutionary dynamics, we identify the design criteria for selection gene drives. We then build prototypes that harness the selective pressure of multiple approved tyrosine kinase inhibitors and employ therapeutic mechanisms as diverse as prodrug catalysis and immune activity induction. We show that selection gene drives can eradicate diverse forms of genetic resistance in vitro. Finally, we demonstrate that model-informed switch engagement effectively targets pre-existing resistance in mouse models of solid tumors. These results establish selection gene drives as a powerful framework for evolution-guided anticancer therapy.
Collapse
Affiliation(s)
- Scott M Leighow
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Huck Institute For The Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Joshua A Reynolds
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Ivan Sokirniy
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
- Huck Institute For The Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Shun Yao
- Huck Institute For The Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Zeyu Yang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Haider Inam
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Dominik Wodarz
- Department of Biology, University of California San Diego, San Diego, CA, USA
| | - Marco Archetti
- Huck Institute For The Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Justin R Pritchard
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA.
- Huck Institute For The Life Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
25
|
Cognigni V, Giudice GC, Bozzetti F, Milanese G, Moschini I, Casali M, Mazzaschi G, Tiseo M. Successful treatment with selpercatinib after pralsetinib-related pneumonitis and intracranial failure in a patient with RET-rearranged nonsmall cell lung cancer. Anticancer Drugs 2024; 35:559-562. [PMID: 38453158 PMCID: PMC11078287 DOI: 10.1097/cad.0000000000001590] [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: 02/03/2024] [Revised: 02/03/2024] [Indexed: 03/09/2024]
Abstract
Pralsetinib and selpercatinib are two highly potent and selective rearranged during transfection (RET) inhibitors that substantially improved the clinical outcome of patients with RET-rearranged non-small cell lung cancer. Treatment with one RET inhibitor after failure of the other is generally not recommended because of cross-resistance mechanisms. We report the case of a patient affected by metastatic RET-rearranged non-small cell lung cancer who experienced long-lasting disease control with pralsetinib. After 13 months from treatment start, the patient developed recurrent drug-related pneumonitis, requiring temporary interruptions and dose reductions and eventually failing to control the disease. Selpercatinib was then started as an off-label treatment, allowing both clinical and radiological intracranial disease control. Selpercatinib was well-tolerated at full dosage, and no pulmonary event occurred. In our case report, after pralsetinib dose reduction due to pulmonary toxicity, the therapeutic switch to selpercatinib allowed the patient to receive a full-dose treatment, eventually restoring disease control. Our case report and a few literature data suggest that switching from pralsetinib to selpercatinib may represent a therapeutic opportunity, especially for patients with brain metastases.
Collapse
Affiliation(s)
- Valeria Cognigni
- Department of Medical Oncology, Università Politecnica delle Marche, Ancona
| | | | - Francesca Bozzetti
- Department of Medicine and Surgery, University of Parma
- Neuroradiology Unit
| | - Gianluca Milanese
- Department of Medicine and Surgery, University of Parma
- Radiology Unit, University Hospital of Parma, Parma
| | | | - Miriam Casali
- Medical Oncology Unit, Azienda Socio-Sanitaria Territoriale di Lodi, Lodi, Italy
| | - Giulia Mazzaschi
- Department of Medicine and Surgery, University of Parma
- Medical Oncology Unit
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma
- Medical Oncology Unit
| |
Collapse
|
26
|
Burley SK, Wu-Wu A, Dutta S, Ganesan S, Zheng SXF. Impact of structural biology and the protein data bank on us fda new drug approvals of low molecular weight antineoplastic agents 2019-2023. Oncogene 2024; 43:2229-2243. [PMID: 38886570 PMCID: PMC11245395 DOI: 10.1038/s41388-024-03077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Open access to three-dimensional atomic-level biostructure information from the Protein Data Bank (PDB) facilitated discovery/development of 100% of the 34 new low molecular weight, protein-targeted, antineoplastic agents approved by the US FDA 2019-2023. Analyses of PDB holdings, the scientific literature, and related documents for each drug-target combination revealed that the impact of structural biologists and public-domain 3D biostructure data was broad and substantial, ranging from understanding target biology (100% of all drug targets), to identifying a given target as likely druggable (100% of all targets), to structure-guided drug discovery (>80% of all new small-molecule drugs, made up of 50% confirmed and >30% probable cases). In addition to aggregate impact assessments, illustrative case studies are presented for six first-in-class small-molecule anti-cancer drugs, including a selective inhibitor of nuclear export targeting Exportin 1 (selinexor, Xpovio), an ATP-competitive CSF-1R receptor tyrosine kinase inhibitor (pexidartinib,Turalia), a non-ATP-competitive inhibitor of the BCR-Abl fusion protein targeting the myristoyl binding pocket within the kinase catalytic domain of Abl (asciminib, Scemblix), a covalently-acting G12C KRAS inhibitor (sotorasib, Lumakras or Lumykras), an EZH2 methyltransferase inhibitor (tazemostat, Tazverik), and an agent targeting the basic-Helix-Loop-Helix transcription factor HIF-2α (belzutifan, Welireg).
Collapse
Affiliation(s)
- Stephen K Burley
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA.
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| | - Amy Wu-Wu
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Shuchismita Dutta
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| | - Steven X F Zheng
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA
| |
Collapse
|
27
|
Brandenburg T, Kroiß M. [Thyroid carcinomas: the role of systemic therapies in internal medicine]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024; 65:642-655. [PMID: 38900279 DOI: 10.1007/s00108-024-01728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/21/2024]
Abstract
The molecular pathogenesis of thyroid carcinoma is well studied and of importance for the treatment of advanced stages. Differentiated, poorly differentiated and anaplastic carcinomas originate in the follicular cells, while medullary carcinomas derive from the C‑cells. The prognosis of differentiated thyroid carcinoma is generally very favourable after surgery and radioiodine therapy. Where tumours progress and lose the ability to enrich iodine, curative treatment is usually not possible. A strategy of watchful waiting is often appropriate. Activating mutations in BRAF or gene fusions of RET and NTRK provide opportunities for targeted therapies. These may be applied with the aim of restoring iodine uptake (redifferentiation). In the absence of molecular therapy targets, multityrosine kinase inhibitors (MKI) are the therapy of choice. If anaplastic thyroid carcinoma is suspected, rapid diagnostic workup including molecular pathology is warranted. Surgery where possible and radiochemotherapy are essential components of therapy. In the presence of a BRAF mutation, inhibition of BRAF and MEK is effective, even if it is not approved in Germany. Where molecular targets are lacking, combination therapy with the MKI lenvatinib and immune checkpoint inhibition is highly effective. Mutations in RET are present in the vast majority of cases of medullary thyroid carcinoma. In aggressive advanced disease, selective RET inhibition has recently been approved as first-line therapy and often leads to an objective response and long-lasting disease stabilisation. In summary, thyroid carcinomas are among the tumour entities for which molecularly targeted therapies can be used most frequently. The involvement of specialised centres is advisable.
Collapse
Affiliation(s)
- Tim Brandenburg
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Hufelandstraße 55, 45147, Essen, Deutschland.
- Endokrines Tumorzentrum am Westdeutschen Tumorzentrum (WTZ), Universitätsklinikum Essen, Member of Endo-ERN and EURACAN, Universität Duisburg-Essen, Duisburg-Essen, Deutschland.
| | - Matthias Kroiß
- Medizinische Klinik IV, Universitätsklinikum, Member of Endo-ERN and EURACAN, Ludwig-Maximilians-Universität München, Ziemssenstr. 5, 80336, München, Deutschland.
- Comprehensive Cancer Center München, Ludwig-Maximilians-Universität München, München, Deutschland.
- Bayerisches Zentrum für Krebsforschung, München, Deutschland.
| |
Collapse
|
28
|
Abbas AA, Farghaly TA, Dawood KM. Recent advances on anticancer and antimicrobial activities of directly-fluorinated five-membered heterocycles and their benzo-fused systems. RSC Adv 2024; 14:19752-19779. [PMID: 38899036 PMCID: PMC11185950 DOI: 10.1039/d4ra01387e] [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: 02/23/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Due to the importance of the fluorinated heterocycles as main components of marketed drugs where 20% of the anticancer and antibiotic drugs contain fluorine atoms, this review describes the reported five-membered heterocycles and their benzo-fused systems having directly connected fluorine atom(s). The in vivo and in vitro anticancer and antimicrobial activities of these fluorinated heterocycles are well reported. Some fluorinated heterocycles were found to be lead structures for drug design developments where their activities were almost equal to or exceeded the potency of the reference drugs. In most cases, the fluorine-containing heterocycles showed promising safety index via their reduced cytotoxicity in non-cancerous cell lines. SAR study assigned that fluorinated heterocycles having various electron-donating or electron-withdrawing substituents significantly affected the anticancer and antimicrobial activities.
Collapse
Affiliation(s)
- Ashraf A Abbas
- Department of Chemistry, Faculty of Science, Cairo University Giza 12613 Egypt +202 35727556
| | - Thoraya A Farghaly
- Department of Chemistry, Faculty of Science, Cairo University Giza 12613 Egypt +202 35727556
- Department of Chemistry, Faculty of Science, Umm Al-Qura University Makkah Saudi Arabia
| | - Kamal M Dawood
- Department of Chemistry, Faculty of Science, Cairo University Giza 12613 Egypt +202 35727556
| |
Collapse
|
29
|
Sayyah E, Oktay L, Tunc H, Durdagi S. Developing Dynamic Structure-Based Pharmacophore and ML-Trained QSAR Models for the Discovery of Novel Resistance-Free RET Tyrosine Kinase Inhibitors Through Extensive MD Trajectories and NRI Analysis. ChemMedChem 2024; 19:e202300644. [PMID: 38523069 DOI: 10.1002/cmdc.202300644] [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: 11/19/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Activation of RET tyrosine kinase plays a critical role in the pathogenesis of various cancers, including non-small cell lung cancer, papillary thyroid cancers, multiple endocrine neoplasia type 2A and 2B (MEN2A, MEN2B), and familial medullary thyroid cancer. Gene fusions and point mutations in the RET proto-oncogene result in constitutive activation of RET signaling pathways. Consequently, developing effective inhibitors to target RET is of utmost importance. Small molecules have shown promise as inhibitors by binding to the kinase domain of RET and blocking its enzymatic activity. However, the emergence of resistance due to single amino acid changes poses a significant challenge. In this study, a structure-based dynamic pharmacophore-driven approach using E-pharmacophore modeling from molecular dynamics trajectories is proposed to select low-energy favorable hypotheses, and ML-trained QSAR models to predict pIC50 values of compounds. For this aim, extensive small molecule libraries were screened using developed ligand-based models, and potent compounds that are capable of inhibiting RET activation were proposed.
Collapse
Affiliation(s)
- Ehsan Sayyah
- Computational Biology and Molecular Simulations Lab, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul, Turkey
- Computational Drug Design Center (HITMER), Bahçeşehir University, Istanbul, Turkey
| | - Lalehan Oktay
- Computational Biology and Molecular Simulations Lab, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul, Turkey
- Computational Drug Design Center (HITMER), Bahçeşehir University, Istanbul, Turkey
| | - Huseyin Tunc
- Department of Biostatistics and Medical Informatics, School of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Lab, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul, Turkey
- Computational Drug Design Center (HITMER), Bahçeşehir University, Istanbul, Turkey
- Molecular Therapy Lab, Department of Pharmaceutical Chemistry, School of Pharmacy, Bahçeşehir University, Istanbul, Turkey
| |
Collapse
|
30
|
Halder P, Rai A, Talukdar V, Das P, Lakkaniga NR. Pyrazolopyridine-based kinase inhibitors for anti-cancer targeted therapy. RSC Med Chem 2024; 15:1452-1470. [PMID: 38784451 PMCID: PMC11110789 DOI: 10.1039/d4md00003j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/24/2024] [Indexed: 05/25/2024] Open
Abstract
The need for effective cancer treatments continues to be a challenge for the biomedical research community. In this case, the advent of targeted therapy has significantly improved therapeutic outcomes. Drug discovery and development efforts targeting kinases have resulted in the approval of several small-molecule anti-cancer drugs based on ATP-mimicking heterocyclic cores. Pyrazolopyridines are a group of privileged heterocyclic cores in kinase drug discovery, which are present in several inhibitors that have been developed against various cancers. Notably, selpercatinib, glumetinib, camonsertib and olverembatinib have either received approval or are in late-phase clinical studies. This review presents the success stories employing pyrazolopyridine scaffolds as hinge-binding cores to address various challenges in kinase-targeted drug discovery research.
Collapse
Affiliation(s)
- Pallabi Halder
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Anubhav Rai
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Vishal Talukdar
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Parthasarathi Das
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| | - Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad India
| |
Collapse
|
31
|
Acharya B, Saha D, Garcia Garcia N, Armstrong D, Jabali B, Hanafi M, Frett B, Ryan KR. Discovery of 9H-pyrimido[4,5-b]indole derivatives as dual RET/TRKA inhibitors. Bioorg Med Chem 2024; 106:117749. [PMID: 38744018 PMCID: PMC11144469 DOI: 10.1016/j.bmc.2024.117749] [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: 01/12/2024] [Revised: 04/14/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
Aberrant RET kinase signaling is activated in numerous cancers including lung, thyroid, breast, pancreatic, and prostate. Recent approvals of selective RET inhibitors, pralsetinib and selpercatinib, has shifted the focus of RET kinase drug discovery programs towards the development of selective inhibitors. However, selective inhibitors invariably lose efficacy as the selective nature of the inhibitor places Darwinian-like pressure on the tumor to bypass treatment through the selection of novel oncogenic drivers. Further, selective inhibitors are restricted for use in tumors with specific genetic backgrounds that do not encompass diverse patient classes. Here we report the identification of a pyrimido indole RET inhibitor found to also have activity against TRK. This selective dual RET/TRK inhibitor can be utilized in tumors with both RET and TRK genetic backgrounds and can also provide blockade of NTRK-fusions that are selected for from RET inhibitor treatments. Efforts towards developing dual RET/TRK inhibitors can be beneficial in terms of encompassing more diverse patient classes while also achieving blockade against emerging resistance mechanisms.
Collapse
Affiliation(s)
- Baku Acharya
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Debasmita Saha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Conrad Prebys Centre for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Noemi Garcia Garcia
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Daniel Armstrong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Baha'a Jabali
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maha Hanafi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11526, Egypt
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Katie Rose Ryan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
32
|
Hamidi S, Hu MI. RET kinase inhibitors for the treatment of RET-altered thyroid cancers: Current knowledge and future directions. ANNALES D'ENDOCRINOLOGIE 2024; 85:118-126. [PMID: 38342224 DOI: 10.1016/j.ando.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
RET gain-of-function mutations are the most common drivers in medullary thyroid carcinoma, while RET fusions are identified in 5-10% of papillary thyroid carcinomas. Thus, RET plays a major role in the tumorigenesis of thyroid neoplasia, making it a valuable therapeutic target. Over a decade ago, multikinase inhibitors (MKIs) were first shown to have variable degrees of anti-RET activity. Despite some clinical efficacy in RET-altered thyroid cancers, significant off-target activity of MKIs led to marked toxicities limiting their use. More recently, two potent, highly selective RET inhibitors, selpercatinib and pralsetinib, were shown to have notable efficacy in RET-altered cancers, associated with more tolerable side effect profiles than those of MKIs. However, these treatments are non-curative, and emerging evidence suggests that patients who progress on therapy acquire mutations conferring drug resistance. Thus, the quest for a more definitive treatment for advanced, RET-altered thyroid cancers continues. This year we celebrate the 30th anniversary of the association of germline mutations of the RET proto-oncogene with the multiple endocrine neoplasia (MEN) type 2 syndromes. In this timely review, we summarize the current state-of-the-art treatment strategies for RET-altered thyroid cancers, their limitations, as well as future therapeutic avenues.
Collapse
Affiliation(s)
- Sarah Hamidi
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer, Houston, TX, 77030, USA.
| | - Mimi I Hu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer, Houston, TX, 77030, USA
| |
Collapse
|
33
|
Wang ZX, Li QQ, Cai J, Wu JZ, Wang JJ, Zhang MY, Wang QX, Tong ZJ, Yang J, Wei TH, Zhou Y, Dai WC, Ding N, Leng XJ, Sun SL, Xue X, Yu YC, Yang Y, Li NG, Shi ZH. Unraveling the Promise of RET Inhibitors in Precision Cancer Therapy by Targeting RET Mutations. J Med Chem 2024; 67:4346-4375. [PMID: 38484122 DOI: 10.1021/acs.jmedchem.3c02319] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Over the past decades, the role of rearranged during transfection (RET) alterations in tumorigenesis has been firmly established. RET kinase inhibition is an essential therapeutic target in patients with RET-altered cancers. In clinical practice, initial efficacy can be achieved in patients through the utilization of multikinase inhibitors (MKIs) with RET inhibitory activity. However, the effectiveness of these MKIs is impeded by the adverse events associated with off-target effects. Recently, many RET-selective inhibitors, characterized by heightened specificity and potency, have been developed, representing a substantial breakthrough in the field of RET precision oncology. This Perspective focuses on the contemporary understanding of RET mutations, recent advancements in next-generation RET inhibitors, and the challenges associated with resistance to RET inhibitors. It provides valuable insights for the development of next-generation MKIs and selective RET inhibitors.
Collapse
Affiliation(s)
- Zi-Xuan Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Qing-Qing Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jiao Cai
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jia-Zhen Wu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jing-Jing Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Meng-Yuan Zhang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Qing-Xin Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Zhen-Jiang Tong
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jin Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Tian-Hua Wei
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yun Zhou
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Wei-Chen Dai
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Ning Ding
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Xue-Jiao Leng
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Xin Xue
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yan-Cheng Yu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| |
Collapse
|
34
|
Li D, Qian X, Wang Y, Yin Y, Sun H, Zhao H, Wu J, Qiu L. Molecular characterization and functional roles of circulating cell-free extrachromosomal circular DNA. Clin Chim Acta 2024; 556:117822. [PMID: 38325714 DOI: 10.1016/j.cca.2024.117822] [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: 12/24/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Circular DNA segments isolated from chromosomes are known as extrachromosomal circular DNA (eccDNA). Its distinct structure and characteristics, along with the variations observed in different disease states, makes it a promising biomarker. Recent studies have revealed the presence of eccDNAs in body fluids, indicating their involvement in various biological functions. This finding opens up avenues for utilizing eccDNAs as convenient and real-time biomarkers for disease diagnosis, treatment monitoring, and prognosis assessment through noninvasive analysis of body fluids. In this comprehensive review, we focused on elucidating the size profiles, potential mechanisms of formation and clearance, detection methods, and potential clinical applications of eccDNAs. We aimed to provide a valuable reference resource for future research in this field.
Collapse
Affiliation(s)
- Dandan Li
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Xia Qian
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Yingjie Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Yicong Yin
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Huishan Sun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China.
| | - Jie Wu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China.
| | - Ling Qiu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China.
| |
Collapse
|
35
|
Subbiah V, Gouda MA, Iorgulescu JB, Dadu R, Patel K, Sherman S, Cabanillas M, Hu M, Castellanos LE, Amini B, Meric-Bernstam F, Shen T, Wu J. Adaptive Darwinian off-target resistance mechanisms to selective RET inhibition in RET driven cancer. NPJ Precis Oncol 2024; 8:62. [PMID: 38438731 PMCID: PMC10912412 DOI: 10.1038/s41698-024-00563-4] [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: 10/13/2023] [Accepted: 02/23/2024] [Indexed: 03/06/2024] Open
Abstract
Patients treated with RET protein tyrosine kinase inhibitors (TKIs) selpercatinib or pralsetinib develop RET TKI resistance by secondary RET mutations or alterative oncogenes, of which alterative oncogenes pose a greater challenge for disease management because of multiple potential mechanisms and the unclear tolerability of drug combinations. A patient with metastatic medullary thyroid carcinoma (MTC) harboring a RET activation loop D898_E901del mutation was treated with selpercatinib. Molecular alterations were monitored with tissue biopsies and cfDNA during the treatment. The selpercatinib-responsive MTC progressed with an acquired ETV6::NTRK3 fusion, which was controlled by selpercatinib plus the NTRK inhibitor larotrectinib. Subsequently, tumor progressed with an acquired EML4::ALK fusion. Combination of selpercatinib with the dual NTRK/ALK inhibitor entrectinib reduced the tumor burden, which was followed by appearance of NTRK3 solvent-front G623R mutation. Preclinical experiments validated selpercatinib plus larotrectinib or entrectinib inhibited RET/NTRK3 dependent cells, whereas selpercatinib plus entrectinib was necessary to inhibit cells with RET/NTRK3/ALK triple alterations or a mixture of cell population carrying these genetic alterations. Thus, RET-altered MTC adapted to selpercatinib and larotrectinib with acquisition of ETV6::NTRK3 and EML4::ALK oncogenes can be managed by combination of selpercatinib and entrectinib providing proof-of-concept of urgency of incorporating molecular profiling in real-time and personalized N-of-1 care transcending one-size-fits-all approach.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Sarah Cannon Research Institute, Nashville, TN, USA.
| | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Bryan Iorgulescu
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur Patel
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven Sherman
- Department of Endocrine Neoplasia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Cabanillas
- Department of Endocrine Neoplasia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mimi Hu
- Department of Endocrine Neoplasia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luz E Castellanos
- Department of Endocrine Neoplasia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Behrang Amini
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tao Shen
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jie Wu
- Peggy and Charles Stephenson Cancer Center and Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
36
|
Brandenburg T, Machlah YM, Führer D. [Precision medicine in endocrinology exemplified by medullary thyroid cancer]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024; 65:202-210. [PMID: 38231404 DOI: 10.1007/s00108-023-01635-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 01/18/2024]
Abstract
Medullary thyroid cancer (MTC) is a prime example for precision medicine in endocrinology and underlines the immediate benefits of basic, translational and healthcare research for patients with a rare disease in clinical . A mutation in the rearranged during transfection (RET) proto-oncogene that codes for a transmembrane receptor protein tyrosine kinase, leads to constitutive activation of the kinase, which is the decisive pathomechanism for the disease. The MTC occurs in a sporadic (somatic RET mutation) or hereditary form (RET germline mutation, multiple endocrine neoplasia types 2 and 3). For germline mutation carriers the timing of preventive thyroidectomy depends on the RET genotype. For advanced metastasized RET-mutant MTC, selective RET kinase inhibitors are available, which are currently considered to be game changers in the treatment. Based on the specific tumor marker calcitonin, MTC can be identified at an early stage during the differential diagnosis of thyroid nodules. The preoperative calcitonin level even enables statements on the degree of dissemination of the disease and on the probability of a cure through surgery. A new development is the consideration of desmoplasia as a histopathological biomarker for the metastatic potential of a MTC, which could possibly modify the operative approach as well as the future MTC nomenclature. Furthermore, the postoperative calcitonin level and the calcitonin doubling time are highly valid prognostic markers for tumor burden and biological aggressiveness of MTC and therefore decisive for patient follow-up. Biochemical, molecular and histological markers enable a risk-adapted surgical treatment and together with new targeted systemic treatments have contributed to a paradigm shift in the diagnostics, prognosis and treatment of MTC in recent years. Endocrine precision medicine for MTC therefore enabled a change from the previous purely symptom-oriented to a modern preventive and individualized treatment.
Collapse
Affiliation(s)
- Tim Brandenburg
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Essen, Deutschland
- Endokrines Tumorzentrum am Westdeutschen Tumorzentrum (WTZ), Universitätsklinikum Essen, Universität Duisburg-Essen, Member of Endo-ERN und EURACAN, Essen, Deutschland
| | - Yara Maria Machlah
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Essen, Deutschland
- Endokrines Tumorzentrum am Westdeutschen Tumorzentrum (WTZ), Universitätsklinikum Essen, Universität Duisburg-Essen, Member of Endo-ERN und EURACAN, Essen, Deutschland
| | - Dagmar Führer
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Essen, Deutschland.
- Endokrines Tumorzentrum am Westdeutschen Tumorzentrum (WTZ), Universitätsklinikum Essen, Universität Duisburg-Essen, Member of Endo-ERN und EURACAN, Essen, Deutschland.
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Zentrallabor - Bereich Forschung und Lehre, Universitätsklinikum Essen, Hufelandstr. 55, 45177, Essen, Deutschland.
| |
Collapse
|
37
|
Chen W, Dream S, Leung PY, Wu PK, Wong S, Park JI. Selpercatinib combination with the mitochondria-targeted antioxidant MitoQ effectively suppresses RET-mutant thyroid cancer. NPJ Precis Oncol 2024; 8:39. [PMID: 38378752 PMCID: PMC10879150 DOI: 10.1038/s41698-024-00536-7] [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: 06/23/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024] Open
Abstract
Genetic alternation of REarranged during Transfection (RET) that leads to constitutive RET activation is a crucial etiological factor for thyroid cancer. RET is known to regulate mitochondrial processes, although the underlying molecular mechanisms remain unclear. We previously showed that the multi-kinase inhibitors vandetanib and cabozantinib increase the mitochondrial membrane potential (Δψm) in RET-mutated thyroid tumor cells and that this effect can be exploited to increase mitochondrial enrichment of Δψm-sensitive agents in the tumor cells. In this study, we hypothesized that the RET-selective inhibitor, selpercatinib, can increase Δψm and, subsequently, tumor cell uptake of the mitochondria-targeted ubiquinone (MitoQ) to the level to break the mitochondrial homeostasis and induce lethal responses in RET-mutated thyroid tumor cells. We show that selpercatinib significantly increased Δψm, and its combination with MitoQ synergistically suppressed RET-mutated human thyroid tumor cells, which we validated using RET-targeted genetic approaches. Selpercatinib and MitoQ, in combination, also suppressed CCDC6-RET fusion cell line xenografts in mice and prolonged animal survival more effectively than single treatments of each agent. Moreover, we treated two patients with CCDC6-RET or RETM918T thyroid cancer, who could not take selpercatinib at regular doses due to adverse effects, with a dose-reduced selpercatinib and MitoQ combination. In response to this combination therapy, both patients showed tumor reduction. The quality of life of one patient significantly improved over a year until the tumor relapsed. This combination of selpercatinib with MitoQ may have therapeutic potential for patients with RET-mutated tumors and intolerant to regular selpercatinib doses.
Collapse
Affiliation(s)
- Wenjing Chen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sophie Dream
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Pui-Yin Leung
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Pui-Kei Wu
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Stuart Wong
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
38
|
Landa I, Cabanillas ME. Genomic alterations in thyroid cancer: biological and clinical insights. Nat Rev Endocrinol 2024; 20:93-110. [PMID: 38049644 DOI: 10.1038/s41574-023-00920-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 12/06/2023]
Abstract
Tumours can arise from thyroid follicular cells if they acquire driver mutations that constitutively activate the MAPK signalling pathway. In addition, a limited set of additional mutations in key genes drive tumour progression towards more aggressive and less differentiated disease. Unprecedented insights into thyroid tumour biology have come from the breadth of thyroid tumour sequencing data from patients and the wide range of mutation-specific mechanisms identified in experimental models, in combination with the genomic simplicity of thyroid cancers. This knowledge is gradually being translated into refined strategies to stratify, manage and treat patients with thyroid cancer. This Review summarizes the biological underpinnings of the genetic alterations involved in thyroid cancer initiation and progression. We also provide a rationale for and discuss specific examples of how to implement genomic information to inform both recommended and investigational approaches to improve thyroid cancer prognosis, redifferentiation strategies and targeted therapies.
Collapse
Affiliation(s)
- Iñigo Landa
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
39
|
Shen C, Shi X, Wen D, Zhang Y, Du Y, Zhang Y, Ma B, Tang H, Yin M, Huang N, Liao T, Zhang TT, Kong C, Wei W, Ji Q, Wang Y. Comprehensive DNA Methylation Profiling of Medullary Thyroid Carcinoma: Molecular Classification, Potential Therapeutic Target, and Classifier System. Clin Cancer Res 2024; 30:127-138. [PMID: 37931242 DOI: 10.1158/1078-0432.ccr-23-2142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Medullary thyroid carcinoma (MTC) presents a distinct biological context from other thyroid cancers due to its specific cellular origin. This heterogeneous and rare tumor has a high prevalence of advanced diseases, making it crucial to address the limited therapeutic options and enhance complex clinical management. Given the high clinical accessibility of methylation information, we construct the largest MTC methylation cohort to date. EXPERIMENTAL DESIGN Seventy-eight fresh-frozen MTC samples constituted our methylation cohort. The comprehensive study process incorporated machine learning, statistical analysis, and in vitro experiments. RESULTS Our study pioneered the identification of a three-class clustering system for risk stratification, exhibiting pronounced epigenomic heterogeneity. The elevated overall methylation status in MTC-B, combined with the "mutual exclusivity" of hypomethylated sites displayed by MTC-A and MTC-C, distinctively characterized the MTC-specific methylation pattern. Integrating with the transcriptome, we further depicted the features of these three clusters to scrutinize biological properties. Several MTC-specific aberrant DNA methylation events were emphasized in our study. NNAT expression was found to be notably reduced in poor-prognostic MTC-C, with its promoter region overlapping with an upregulated differentially methylated region. In vitro experiments further affirmed NNAT's therapeutic potential. Moreover, we built an elastic-net logistic regression model with a relatively high AUC encompassing 68 probes, intended for future validation and systematic clinical application. CONCLUSIONS Conducting research on diseases with low incidence poses significant challenges, and we provide a robust resource and comprehensive research framework to assist in ongoing MTC case inclusion and facilitate in-depth dissection of its molecular biological features.
Collapse
Affiliation(s)
- Cenkai Shen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Duo Wen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yuqing Zhang
- School of Data Science, Fudan University, Shanghai, P.R. China
| | - Yuxin Du
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yu Zhang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ben Ma
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Haitao Tang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Min Yin
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Naisi Huang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Tian Liao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ting-Ting Zhang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Chang'e Kong
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Wenjun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
40
|
Romanelli MN, Braconi L, Gabellini A, Manetti D, Marotta G, Teodori E. Synthetic Approaches to Piperazine-Containing Drugs Approved by FDA in the Period of 2011-2023. Molecules 2023; 29:68. [PMID: 38202651 PMCID: PMC10780301 DOI: 10.3390/molecules29010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
The piperazine moiety is often found in drugs or in bioactive molecules. This widespread presence is due to different possible roles depending on the position in the molecule and on the therapeutic class, but it also depends on the chemical reactivity of piperazine-based synthons, which facilitate its insertion into the molecule. In this paper, we take into consideration the piperazine-containing drugs approved by the Food and Drug Administration between January 2011 and June 2023, and the synthetic methodologies used to prepare the compounds in the discovery and process chemistry are reviewed.
Collapse
Affiliation(s)
- Maria Novella Romanelli
- Section of Pharmaceutical and Nutraceutical Science, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Via Ugo Schiff, 6, Sesto Fiorentino, 50019 Florence, Italy; (L.B.); (A.G.); (D.M.); (G.M.); (E.T.)
| | | | | | | | | | | |
Collapse
|
41
|
Clark L, Fisher G, Brook S, Patel S, Arkenau HT. Selective RET Inhibitors (SRIs) in Cancer: A Journey from Multi-Kinase Inhibitors to the Next Generation of SRIs. Cancers (Basel) 2023; 16:31. [PMID: 38201460 PMCID: PMC10778005 DOI: 10.3390/cancers16010031] [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: 11/19/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
RET is a receptor tyrosine kinase that plays an important role in the development of neurons and kidneys. The gene encoding the rearranged-during-transfection (RET) receptor tyrosine kinase was first discovered in the 1980s. Activating RET mutations and rearrangements have since been identified as actionable drivers of oncogenesis in numerous cancer types and are most prevalent in thyroid and non-small-cell lung cancer. Following the modest success of repurposed RET-active multikinase inhibitors, the first selective RET inhibitors (SRIs), selpercatinib and pralsetinib, received regulatory approval in 2020. Now, thousands of patients with RET-altered cancers have benefited from first-generation SRIs, with impressive deep and durable responses. However, following prolonged treatment with these SRIs, a number of acquired on-target resistance mutations have been identified together with other non-RET-dependent resistance mechanisms. Today, the focus is on how we can further evolve and improve the treatment of RET-altered tumors with next-generation SRIs, and a number of candidate drugs are in development. The ideal next-generation SRIs will be active against on-target acquired resistance alterations, including those that emerge in the CNS, and will have improved safety and tolerability relative to first-generation SRIs. In this review, we will provide an update on these candidates and their potential to meet the unmet clinical need for patients who progress on first-generation SRIs.
Collapse
|
42
|
Li J, Gu A, Nong XM, Zhai S, Yue ZY, Li MY, Liu Y. Six-Membered Aromatic Nitrogen Heterocyclic Anti-Tumor Agents: Synthesis and Applications. CHEM REC 2023; 23:e202300293. [PMID: 38010365 DOI: 10.1002/tcr.202300293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Indexed: 11/29/2023]
Abstract
Cancer stands as a serious malady, posing substantial risks to human well-being and survival. This underscores the paramount necessity to explore and investigate novel antitumor medications. Nitrogen-containing compounds, especially those derived from natural sources, form a highly significant category of antitumor agents. Among these, antitumor agents with six-membered aromatic nitrogen heterocycles have consistently attracted the attention of chemists and pharmacologists. Accordingly, we present a comprehensive summary of synthetic strategies and clinical implications of these compounds in this review. This entails an in-depth analysis of synthesis pathways for pyridine, quinoline, pyrimidine, and quinazoline. Additionally, we explore the historical progression, targets, mechanisms of action, and clinical effectiveness of small molecule inhibitors possessing these structural features.
Collapse
Affiliation(s)
- Jiatong Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ao Gu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Xiao-Mei Nong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Shuyang Zhai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Zhu-Ying Yue
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Meng-Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
43
|
Mulligan L. Selective RET Kinase Inhibitors and Lung Cancer. N Engl J Med 2023; 389:1913-1916. [PMID: 37966290 DOI: 10.1056/nejme2311295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Affiliation(s)
- Lois Mulligan
- From the Cancer Research Institute, Queen's University, Kingston, ON, Canada
| |
Collapse
|
44
|
Fricke J, Wang J, Gallego N, Mambetsariev I, Kim P, Babikian R, Chen BT, Afkhami M, Subbiah V, Salgia R. Selpercatinib and Pralsetinib Induced Chylous Ascites in RET-Rearranged Lung Adenocarcinoma: A Case Series. Clin Lung Cancer 2023; 24:666-671. [PMID: 37580188 PMCID: PMC10840632 DOI: 10.1016/j.cllc.2023.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023]
Affiliation(s)
- Jeremy Fricke
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA
| | - Joshua Wang
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA
| | | | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA
| | - Pauline Kim
- Department of Pharmacy, City of Hope National Medical Center, Duarte, CA
| | - Razmig Babikian
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA
| | - Bihong T Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA
| | | | - Vivek Subbiah
- Department of Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, TN
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA.
| |
Collapse
|
45
|
Meyer C, McCoy M, Li L, Posner B, Westover KD. LIMS-Kinase provides sensitive and generalizable label-free in vitro measurement of kinase activity using mass spectrometry. CELL REPORTS. PHYSICAL SCIENCE 2023; 4:101599. [PMID: 38213501 PMCID: PMC10783653 DOI: 10.1016/j.xcrp.2023.101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Measurements of kinase activity are important for kinase-directed drug development, analysis of inhibitor structure and function, and understanding mechanisms of drug resistance. Sensitive, accurate, and miniaturized assay methods are crucial for these investigations. Here, we describe a label-free, high-throughput mass spectrometry-based assay for studying individual kinase enzymology and drug discovery in a purified system, with a focus on validated drug targets as benchmarks. We demonstrate that this approach can be adapted to many known kinase substrates and highlight the benefits of using mass spectrometry to measure kinase activity in vitro, including increased sensitivity. We speculate that this approach to measuring kinase activity will be generally applicable across most of the kinome, enabling research on understudied kinases and kinase drug discovery.
Collapse
Affiliation(s)
- Cynthia Meyer
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Melissa McCoy
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Lianbo Li
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Bruce Posner
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Kenneth D. Westover
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
- X (formerly Twitter): @KENWESTOVER
- Lead contact
| |
Collapse
|
46
|
Sahakian N, Castinetti F, Romanet P. Molecular Basis and Natural History of Medullary Thyroid Cancer: It is (Almost) All in the RET. Cancers (Basel) 2023; 15:4865. [PMID: 37835559 PMCID: PMC10572078 DOI: 10.3390/cancers15194865] [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: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Medullary thyroid cancer (MTC) is a rare disease, which can be either sporadic (roughly 75% of cases) or genetically determined (multiple endocrine neoplasia type 2, due to REarranged during Transfection RET germline mutations, 25% of cases). Interestingly, RET pathogenic variants (mainly M918T) have also been reported in aggressive forms of sporadic MTC, suggesting the importance of RET signalling pathways in the pathogenesis of MTC. The initial theory of RET codon-related MTC aggressiveness has been recently questioned by studies suggesting that this would only define the age at disease onset rather than the aggressiveness of MTC. Other factors might however impact the natural history of the disease, such as RET polymorphisms, epigenetic factors, environmental factors, MET (mesenchymal-epithelial transition) alterations, or even other genetic alterations such as RAS family (HRAS, KRAS, NRAS) genetic alterations. This review will detail the molecular bases of MTC, focusing on RET pathways, and the potential mechanisms that explain the phenotypic intra- and interfamilial heterogeneity.
Collapse
Affiliation(s)
- Nicolas Sahakian
- Aix Marseille Univ, APHM, INSERM, MMG, La Conception University Hospital, Department of Endocrinology, Marseille, France; (N.S.); (F.C.)
| | - Frédéric Castinetti
- Aix Marseille Univ, APHM, INSERM, MMG, La Conception University Hospital, Department of Endocrinology, Marseille, France; (N.S.); (F.C.)
| | - Pauline Romanet
- Aix Marseille Univ, APHM, INSERM, MMG, La Conception University Hospital, Laboratory of Molecular Biology, Marseille, France
| |
Collapse
|
47
|
Gou X, Kim BJ, Anurag M, Lei JT, Young MN, Holt MV, Fandino D, Vollert CT, Singh P, Alzubi MA, Malovannaya A, Dobrolecki LE, Lewis MT, Li S, Foulds CE, Ellis MJ. Kinome Reprogramming Is a Targetable Vulnerability in ESR1 Fusion-Driven Breast Cancer. Cancer Res 2023; 83:3237-3251. [PMID: 37071495 PMCID: PMC10543968 DOI: 10.1158/0008-5472.can-22-3484] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/20/2023] [Accepted: 04/12/2023] [Indexed: 04/19/2023]
Abstract
Transcriptionally active ESR1 fusions (ESR1-TAF) are a potent cause of breast cancer endocrine therapy (ET) resistance. ESR1-TAFs are not directly druggable because the C-terminal estrogen/anti-estrogen-binding domain is replaced with translocated in-frame partner gene sequences that confer constitutive transactivation. To discover alternative treatments, a mass spectrometry (MS)-based kinase inhibitor pulldown assay (KIPA) was deployed to identify druggable kinases that are upregulated by diverse ESR1-TAFs. Subsequent explorations of drug sensitivity validated RET kinase as a common therapeutic vulnerability despite remarkable ESR1-TAF C-terminal sequence and structural diversity. Organoids and xenografts from a pan-ET-resistant patient-derived xenograft model that harbors the ESR1-e6>YAP1 TAF were concordantly inhibited by the selective RET inhibitor pralsetinib to a similar extent as the CDK4/6 inhibitor palbociclib. Together, these findings provide preclinical rationale for clinical evaluation of RET inhibition for the treatment of ESR1-TAF-driven ET-resistant breast cancer. SIGNIFICANCE Kinome analysis of ESR1 translocated and mutated breast tumors using drug bead-based mass spectrometry followed by drug-sensitivity studies nominates RET as a therapeutic target. See related commentary by Wu and Subbiah, p. 3159.
Collapse
Affiliation(s)
- Xuxu Gou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston Texas
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Jonathan T. Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Meggie N. Young
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Matthew V. Holt
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Diana Fandino
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Craig T. Vollert
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, Los Angeles
| | - Purba Singh
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Mohammad A. Alzubi
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, Los Angeles
| | - Anna Malovannaya
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| | - Shunqiang Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Charles E. Foulds
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew J. Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
48
|
Zhou Y, Wu X, Zhang Y, Li Z, Ge X, Chen H, Mao Y, Ding W. Performance of multigene testing in cytologically indeterminate thyroid nodules and molecular risk stratification. PeerJ 2023; 11:e16054. [PMID: 37744220 PMCID: PMC10512961 DOI: 10.7717/peerj.16054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/16/2023] [Indexed: 09/26/2023] Open
Abstract
Objective Thyroid cancer is the third most prevalent cancer among females. Genetic testing based on next-generation sequencing may provide an auxiliary diagnosis to reduce cytologically diagnostic uncertainty. However, commercial multigene tests are not widely available and are not well-tested in the Chinese population. Methods In this study, we designed a multigene testing panel and evaluated its performance in 529 cytologically indeterminate thyroid nodules (Bethesda III, IV and V). The molecular data of the DNA mutations and RNA fusions of fine needle aspiration samples were reviewed in conjunction with a clinical diagnosis, pathological reports, and definitive surgery for retrospective analysis. Then, the molecular risk stratification was investigated for its accuracy in malignant risk prediction. Results The overall combined consistency revealed substantial agreement (Kappa = 0.726) with the sensitivity, specificity, positive predictive value, and negative predictive values of 97.80%, 82.14%, 98.99%, and 67.65%, respectively. The most common aberration was BRAFV600E (82.59%), followed by NRAS mutants (4.07%), RET fusions (3.70%), and KRAS mutants (3.15%). Two cases (0.44%) were categorized into a high-risk group, 426 cases (94.67%) were categorized into a BRAF-like group with totally histopathologic papillary patterned tumors, and 22 cases (4.89%) were categorized into a RAS-like group with 14 papillary and eight follicular patterned tumors when the cohort concurrent aberrations were excluded. Potentially aggressive features may be related to concurrent molecular alterations of BRAFV600E with TERTQ302R, and AKT1L52R, NRASG12C, NRASQ61R, and CCDC6-RET fusions. Conclusions This study provided a multigene panel for identifying benign nodules from cytologically indeterminate thyroid nodules to avoid unnecessary surgery. We provide further evidence for using molecular risk stratification as a promising predictor of disease outcomes. The results of this study may be limited by the extremely high prevalence of cancer in the cohort for clinical reference.
Collapse
Affiliation(s)
- Yuanyuan Zhou
- Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Hefei, Anhui Province, China
| | - Xinping Wu
- Department of Ultrasound, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yuzhi Zhang
- Department of Ultrasound, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Zhiqiang Li
- Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Hefei, Anhui Province, China
| | - Xia Ge
- Department of Pathological Diagnosis, KingMed Center for Clinical Laboratory Co., Ltd, Hefei, Anhui Province, China
| | - Hao Chen
- Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Hefei, Anhui Province, China
| | - Yuan Mao
- Genome Center, KingMed Center for Clinical Laboratory Co., Ltd, Hefei, Anhui Province, China
| | - Wenbo Ding
- Department of Ultrasound, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
49
|
Gild ML, Clifton-Bligh RJ, Wirth LJ, Robinson BG. Medullary Thyroid Cancer: Updates and Challenges. Endocr Rev 2023; 44:934-946. [PMID: 37204852 PMCID: PMC10656709 DOI: 10.1210/endrev/bnad013] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/19/2023] [Accepted: 05/18/2023] [Indexed: 05/20/2023]
Abstract
A personalized approach to the management of medullary thyroid cancer (MTC) presents several challenges; however, in the past decade significant progress has been made in both diagnostic and treatment modalities. Germline rearranged in transfection (RET) testing in multiple endocrine neoplasia 2 and 3, and somatic RET testing in sporadic MTC have revolutionized the treatment options available to patients. Positron emission tomography imaging with novel radioligands has improved characterization of disease and a new international grading system can predict prognosis. Systemic therapy for persistent and metastatic disease has evolved significantly with targeted kinase therapy especially for those harboring germline or somatic RET variants. Selpercatinib and pralsetinib are highly selective RET kinase inhibitors that have shown improved progression-free survival with better tolerability than outcomes seen in earlier multikinase inhibitor studies. Here we discuss changes in paradigms for MTC patients: from determining RET alteration status upfront to novel techniques for the evaluation of this heterogenous disease. Successes and challenges with kinase inhibitor use will illustrate how managing this rare malignancy continues to evolve.
Collapse
Affiliation(s)
- Matti L Gild
- Faculty of Health and Medicine, University of Sydney, Sydney 2006, Australia
- Department of Diabetes and Endocrinology, Royal North Shore Hospital, Sydney 2065, Australia
- Cancer Genetics, Kolling Institute of Medical Research, Sydney 2065, Australia
| | - Roderick J Clifton-Bligh
- Faculty of Health and Medicine, University of Sydney, Sydney 2006, Australia
- Department of Diabetes and Endocrinology, Royal North Shore Hospital, Sydney 2065, Australia
- Cancer Genetics, Kolling Institute of Medical Research, Sydney 2065, Australia
| | - Lori J Wirth
- Department of Medicine, Massachusetts General Hospital, & Harvard Medical School, Boston 02114, USA
| | - Bruce G Robinson
- Faculty of Health and Medicine, University of Sydney, Sydney 2006, Australia
- Department of Diabetes and Endocrinology, Royal North Shore Hospital, Sydney 2065, Australia
- Cancer Genetics, Kolling Institute of Medical Research, Sydney 2065, Australia
| |
Collapse
|
50
|
Hadoux J, Al Ghuzlan A, Lamartina L, Bani MA, Moog S, Attard M, Scoazec JY, Hartl D, Aldea M, Friboulet L, Jules-Clement G, Italiano A, Besse B, Lacroix L, Baudin E. Patterns of Treatment Failure After Selective Rearranged During Transfection (RET) Inhibitors in Patients With Metastatic Medullary Thyroid Carcinoma. JCO Precis Oncol 2023; 7:e2300053. [PMID: 38127829 DOI: 10.1200/po.23.00053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/13/2023] [Accepted: 09/20/2023] [Indexed: 12/23/2023] Open
Abstract
PURPOSE Medullary thyroid cancer (MTC) harbors frequent mutations in RET oncogene. Selective RET inhibitors (RETi) have emerged as effective treatments. However, resistance almost invariably occurs. METHODS MTC patients who were initiated on RETi between 2018 and 2022 were included. Baseline characteristics, RET mutational status, RETi response, available tumor tissue and molecular profiles sampled pre- and post-RETi were analyzed. RESULTS Among 46 MTC patients on RETi during the study period, 26 patients had discontinued at data cut-off because of progression (n = 16), death (n = 4), and toxicity (n = 6). The most frequent RET mutations at baseline were p.M918T (n = 29), and p.C634X (n = 6). Pre- and post-RETi molecular profiles were available in 14 patients. There was no primary resistance on pre-RETi samples. Post-RETi profiles revealed a bypass mechanism of resistance in 75% of the cases including RAS genes mutations (50%), FGFR2 and ALK fusions and and MYC p.P44L. RET solvent from and hinge region mutations was the only resistance mechanisms in 25% of the cases. Tumor samples from initial thyroidectomy, pre- and post-RETi, from six patients, showed an increase of the mean Ki 67-index of 7%, 17% and 40% respectively (P = 0.037) and a more aggressive poorly differentiated histology in three patients. DISCUSSION Bypass resistance may be the most frequent mechanism of progression under RETi. A more aggressive histology may arise following RETi and warrants further investigation.
Collapse
Affiliation(s)
- Julien Hadoux
- Département d'imagerie, Service d'oncologie endocrinienne, Gustave Roussy, Villejuif, France
| | - Abir Al Ghuzlan
- Département de biologie et pathologie médicale, Gustave Roussy, Villejuif, France
| | - Livia Lamartina
- Département d'imagerie, Service d'oncologie endocrinienne, Gustave Roussy, Villejuif, France
| | - Mohamed-Amine Bani
- Département de biologie et pathologie médicale, Gustave Roussy, Villejuif, France
| | - Sophie Moog
- Département d'imagerie, Service d'oncologie endocrinienne, Gustave Roussy, Villejuif, France
| | - Marie Attard
- Département d'imagerie, Gustave Roussy, Villejuif, France
| | - Jean Yves Scoazec
- Département de biologie et pathologie médicale, Gustave Roussy, Villejuif, France
- Faculté de Médecine, Paris-Saclay Université, Le Kremlin-Bicêtre, France
| | - Dana Hartl
- Département de chirurgie et anesthésie, Gustave Roussy, Villejuif, France
| | - Mihaela Aldea
- Faculté de Médecine, Paris-Saclay Université, Le Kremlin-Bicêtre, France
- Département de médecine, Gustave Roussy, Villejuif, France
| | - Luc Friboulet
- Inserm U981, Gustave Roussy, Paris-Saclay Université, Villejuif, France
| | | | - Antoine Italiano
- Département d'innovation thérapeutique et essais précoces, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Faculté de Médecine, Paris-Saclay Université, Le Kremlin-Bicêtre, France
- Département de médecine, Gustave Roussy, Villejuif, France
| | - Ludovic Lacroix
- Département de biologie et pathologie médicale, Gustave Roussy, Villejuif, France
| | - Eric Baudin
- Département d'imagerie, Service d'oncologie endocrinienne, Gustave Roussy, Villejuif, France
| |
Collapse
|