1
|
Chen JF, Guo SJ, He B, Zheng W, Jiang WJ, Yuan Z, Xiang Y, Peng C, Xiong W, Shi JY. Advances of dual inhibitors based on ALK for the treatment of cancer. Bioorg Chem 2025; 159:108417. [PMID: 40168884 DOI: 10.1016/j.bioorg.2025.108417] [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: 02/03/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Anaplastic lymphoma kinase (ALK), which encodes a highly conserved receptor tyrosine kinase (RTK), is important for the development and progression of many tumors, especially non-small cell lung cancer (NSCLC). Currently, third-generation ALK inhibitors are used to treat ALK-mutant NSCLC, but the rapid emergence of resistance during treatment greatly limits their efficacy in clinic. In comparison to single-target inhibitors, ALK dual inhibitors offer the benefits of reducing the emergence of drug resistance, improving treatment efficacy, and optimizing pharmacokinetic features due to the synergistic function of ALK and other associated targets involved in tumor progression. Therefore, we outline the development of ALK dual inhibitors, highlight their design approaches and structure-activity relationship (SAR), and offer insights into new challenges and potential future directions in this area.
Collapse
Affiliation(s)
- Jin-Feng Chen
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731. China; Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Shu-Jin Guo
- Department of Health Management Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bin He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Zheng
- Department of Integrative Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Wen-Jie Jiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhuo Yuan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Xiong
- Department of urology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu 610072, China.
| | - Jian-You Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
2
|
Yu N, Lee JE, Park S, Yun SK, Ryu DH, Kim JA, Kim JH, Hwang JY. Design and evaluation of anaplastic lymphoma kinase degraders using a covalent fumarate handle. Bioorg Med Chem Lett 2025; 117:130075. [PMID: 39681169 DOI: 10.1016/j.bmcl.2024.130075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/18/2024]
Abstract
Targeted protein degradation has emerged as a novel therapeutic paradigm in drug discovery. Despite the FDA approval of anaplastic lymphoma kinase (ALK) inhibitors, the pursuit of compounds with enhanced potency and prolonged efficacy remains crucial to mitigate inevitable adverse effects. In this context, we endeavored to develop ALK degraders utilizing FDA-approved ALK inhibitors-crizotinib, ceritinib, brigatinib, and alectinib-as ALK binders, along with 4-methoxyphenylfumarate as a covalent handle to bind to RNF126 E3 ligase. Among the synthesized compounds, dALK-3-derived from brigatinib-efficiently induced the proteasomal degradation of EML4-ALK and exhibited a 10-fold superior anti-proliferative effect on H3122 cells compared to brigatinib. However, the enhanced anti-proliferative activity of dALK-3 was found to be independent of RNF126, a presumed potential E3 ligase, suggesting the need for investigation of other components within the ubiquitin-proteasome system. Our findings further support the potential application of the fumarate moiety as a binder for E3 ligases in targeted protein degradation.
Collapse
Affiliation(s)
- Namsik Yu
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Department of Chemistry, Sungkyunkwan University, Jangan-Gu, Suwon, Republic of Korea
| | - Ji-Eun Lee
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seulki Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Su Kyeong Yun
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Department of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Do Hyun Ryu
- Department of Chemistry, Sungkyunkwan University, Jangan-Gu, Suwon, Republic of Korea
| | - Jung-Ae Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Republic of Korea; Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Jeong-Hoon Kim
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Republic of Korea; Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Jong Yeon Hwang
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Medicinal Chemistry and Pharmacology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
3
|
Bhagriya P, Shaikh A, Roy H. Picropodophyllotoxin alters EMT in neuroblastoma via inhibition of surface receptors IGF1R and ALK. Growth Horm IGF Res 2025; 80:101638. [PMID: 40015087 DOI: 10.1016/j.ghir.2025.101638] [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: 08/07/2024] [Revised: 10/25/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Neuroblastoma (NB) is a type of paediatric cancer that originates from embryonic sympathoadrenal cells. Despite its paediatric origin, NB is mostly treated with strategy of non-small cell lung cancer like adults due to lack of specific therapeutic approach. To improve treatment outcome for NB patients, developing drugs that specifically target the genetic mutations or molecular pathways involved in neuroblastoma is necessary. Overexpression of the insulin-like growth factor 1 receptor (IGF1R) has been linked to various malignancies, including paediatric cancers. We hypothesized that inhibiting IGF1R with ALK (NB specific mutation) by phytochemical compound could effectively treat NB while avoiding undesirable cytotoxic effects. We evaluated the efficacy of Picropodophyllotoxin (PPP) as IGF1R inhibitor, for treatment of NB. The IC50 value of PPP on SH-SY5Y, NB cells after 24 h of treatment was found to be 0.501 μM. Molecular docking studies revealed that PPP had a binding score of -7.5 kcal/mol with IGF1R and - 8.8 kcal/mol with ALK. This suggests that PPP not only binds to and inhibits IGF1R but also has a strong affinity for ALK. Gene expression studies, densitometric analysis, scratch assays, and AO/EtBr differential staining were used to evaluate the efficacy of PPP in NB cells. Transcript expression and densitometric analysis revealed that PPP could downregulate IGF1R and ALK in NB cells. Downregulation of SNAIL, a mesenchymal marker, and upregulation of E-cadherin, an epithelial marker, indicated a mesenchymal to epithelial transition in NB cells, suggesting that PPP treatment inhibited NB cell migration and proliferation. This was further supported by scratch assay results in our study. Furthermore, gene expression analysis of p53, BAX and BCL2 indicated that PPP induces apoptosis in NB cells. AO/EtBr differential staining revealed apoptotic phenomena in NB cells after 24 h of PPP treatment. Although further research is needed to explore the receptor targeting approach using PPP for IGF1R and ALK inhibition.
Collapse
Affiliation(s)
- Poonam Bhagriya
- Nutrigenomics and Cancer Biology Lab, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India; Post-Graduate Department of Biosciences, Satellite Campus, Sardar Patel University, Bakrol, Anand 388315, Gujarat, India
| | - Afridi Shaikh
- Nutrigenomics and Cancer Biology Lab, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India
| | - Hetal Roy
- Nutrigenomics and Cancer Biology Lab, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390002, Gujarat, India.
| |
Collapse
|
4
|
Ramasamy SS, Adhigaman K, Nandakumar V, Sundarasamy A, Jagadeesan S, Saravanakumar M, Malecki JG, Easwaran N, Thangaraj S. In-Silico exploration: Unraveling the anti-cancer potential of 8-Nitroquinoline hydrazides. J Mol Struct 2025; 1321:140218. [DOI: 10.1016/j.molstruc.2024.140218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Torresan S, Bortolot M, De Carlo E, Bertoli E, Stanzione B, Del Conte A, Spina M, Bearz A. Matters of the Heart: Cardiotoxicity Related to Target Therapy in Oncogene-Addicted Non-Small Cell Lung Cancer. Int J Mol Sci 2025; 26:554. [PMID: 39859270 PMCID: PMC11765312 DOI: 10.3390/ijms26020554] [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: 11/22/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The treatment of Non Small Cell Lung Cancer (NSCLC) has been revolutionised by the introduction of targeted therapies. With the improvement of response and frequently of overall survival, however, a whole new set of adverse events emerged. In fact, due to the peculiar mechanism of action of each one of the tyrosine kinase inhibitors and other targeted therapies, every drug has its own specific safety profile. In addition, this safety profile could not fully emerge from clinical trials data, as patients in clinical practice usually have more comorbidities and frailties. Cardiotoxicity is a well-known and established adverse event of anti-cancer therapies. However, only recently it has become a central topic for targeted therapies in NSCLC, due to the unknown real range and frequency. Management of this toxicity begins with prevention, and must balance the need of continuing an effective anticancer treatment versus low risk of even fatal events and the preservation of long-term quality of life. The aim of this review is to summarise the current knowledge focusing on currently used targeted therapies in NSCLC.
Collapse
Affiliation(s)
- Sara Torresan
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, 33081 Aviano, Italy
| |
Collapse
|
6
|
Liu ZY, Zhang YW, Zhuang HX, Ou YJ, Jiang QY, Li PF, He YM, Ren Y, Mao XL. Inhibiting the Otub1/phosphorylated STAT3 axis for the treatment of non-small cell lung cancer. Acta Pharmacol Sin 2025; 46:184-195. [PMID: 39198663 PMCID: PMC11697133 DOI: 10.1038/s41401-024-01366-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024]
Abstract
The transcription factor STAT3 is a promising target for the treatment of non-small cell lung cancer (NSCLC). STAT3 activity is mainly dependent on phosphorylation at tyrosine 705 (pSTAT3-Y705), but the modulation on pSTAT3-Y705 is elusive. By screening a library of deubiquitinases (Dubs), we found that the Otub1 increases STAT3 transcriptional activity. As a Dub, Otub1 binds to pSTAT3-Y705 and specifically abolishes its K48-linked ubiquitination, therefore preventing its degradation and promoting NSCLC cell survival. The Otub1/pSTAT3-Y705 axis could be a potential target for the treatment of NSCLC. To explore this concept, we screen libraries of FDA-approved drugs and natural products based on STAT3-recognition element-driven luciferase assay, from which crizotinib is found to block pSTAT3-Y705 deubiquitination and promotes its degradation. Different from its known action to induce ALK positive NSCLC cell apoptosis, crizotinib suppresses ALK-intact NSCLC cell proliferation and colony formation but not apoptosis. Furthermore, crizotinib also suppresses NSCLC xenograft growth in mice. Taken together, these findings identify Otub1 as the first deubiquitinase of pSTAT3-Y705 and provide that the Otub1/pSTAT3-Y705 axis is a promising target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Zi-Yang Liu
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University & Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ya-Wen Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Hai-Xia Zhuang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University & Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Jie Ou
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University & Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qiu-Yun Jiang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University & Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ping-Fei Li
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University & Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuan-Ming He
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University & Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ying Ren
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Xin-Liang Mao
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University & Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
7
|
Zakharova G, Suntsova M, Rabushko E, Mohammad T, Drobyshev A, Seryakov A, Poddubskaya E, Moisseev A, Smirnova A, Sorokin M, Tkachev V, Simonov A, Guguchkin E, Karpulevich E, Buzdin A. A New Approach of Detecting ALK Fusion Oncogenes by RNA Sequencing Exon Coverage Analysis. Cancers (Basel) 2024; 16:3851. [PMID: 39594806 PMCID: PMC11592821 DOI: 10.3390/cancers16223851] [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: 10/01/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND In clinical practice, various methods are used to identify ALK gene rearrangements in tumor samples, ranging from "classic" techniques, such as IHC, FISH, and RT-qPCR, to more advanced highly multiplexed approaches, such as NanoString technology and NGS panels. Each of these methods has its own advantages and disadvantages, but they share the drawback of detecting only a restricted (although sometimes quite extensive) set of preselected biomarkers. At the same time, whole transcriptome sequencing (WTS, RNAseq) can, in principle, be used to detect gene fusions while simultaneously analyzing an incomparably wide range of tumor characteristics. However, WTS is not widely used in practice due to purely analytical limitations and the high complexity of bioinformatic analysis, which requires considerable expertise. In particular, methods to detect gene fusions in RNAseq data rely on the identification of chimeric reads. However, the typically low number of true fusion reads in RNAseq limits its sensitivity. In a previous study, we observed asymmetry in the RNAseq exon coverage of the 3' partners of some fusion transcripts. In this study, we conducted a comprehensive evaluation of the accuracy of ALK fusion detection through an analysis of differences in the coverage of its tyrosine kinase exons. METHODS A total of 906 human cancer biosamples were subjected to analysis using experimental RNAseq data, with the objective of determining the extent of asymmetry in ALK coverage. A total of 50 samples were analyzed, comprising 13 samples with predicted ALK fusions and 37 samples without predicted ALK fusions. These samples were assessed by targeted sequencing with two NGS panels that were specifically designed to detect fusion transcripts (the TruSight RNA Fusion Panel and the OncoFu Elite panel). RESULTS ALK fusions were confirmed in 11 out of the 13 predicted cases, with an overall accuracy of 96% (sensitivity 100%, specificity 94.9%). Two discordant cases exhibited low ALK coverage depth, which could be addressed algorithmically to enhance the accuracy of the results. It was also important to consider read strand specificity due to the presence of antisense transcripts involving parts of ALK. In a limited patient sample undergoing ALK-targeted therapy, the algorithm successfully predicted treatment efficacy. CONCLUSIONS RNAseq exon coverage analysis can effectively detect ALK rearrangements.
Collapse
Affiliation(s)
- Galina Zakharova
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | - Maria Suntsova
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Endocrinology Research Center, 117292 Moscow, Russia;
| | - Elizaveta Rabushko
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | | | - Alexey Drobyshev
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | | | - Elena Poddubskaya
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Clinical Center Vitamed, 121309 Moscow, Russia
| | - Alexey Moisseev
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Oncobox LLC, 119991 Moscow, Russia;
| | - Anastasia Smirnova
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Oncobox LLC, 119991 Moscow, Russia;
| | - Maxim Sorokin
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Oncobox LLC, 119991 Moscow, Russia;
| | | | - Alexander Simonov
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
| | - Egor Guguchkin
- Institute for System Programming of RAS, 109004 Moscow, Russia; (E.G.); (E.K.)
| | - Evgeny Karpulevich
- Institute for System Programming of RAS, 109004 Moscow, Russia; (E.G.); (E.K.)
| | - Anton Buzdin
- Institute for Personalized Oncology, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (G.Z.); (M.S.); (E.R.); (A.D.); (E.P.); (A.M.); (A.S.); (M.S.); (A.S.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
8
|
Sankarapandian V, Rajendran RL, Miruka CO, Sivamani P, Maran BAV, Krishnamoorthy R, Gangadaran P, Ahn BC. A review on tyrosine kinase inhibitors for targeted breast cancer therapy. Pathol Res Pract 2024; 263:155607. [PMID: 39326367 DOI: 10.1016/j.prp.2024.155607] [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: 06/26/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
Breast cancer is a heterogeneous disease with complex molecular pathogenesis. Overexpression of several tyrosine kinase receptors is associated with poor prognosis, therefore, they can be key targets in breast cancer therapy. Tyrosine kinase inhibitors (TKIs) have emerged as leading agents in targeted cancer therapy due to their effectiveness in disrupting key molecular pathways involved in tumor growth. TKIs target various tyrosine kinases, including the human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), Vascular endothelial growth factor receptor (VEGFR), anaplastic lymphoma kinase (ALK), vascular endothelial growth factor receptor (VEGFR)-associated multi-targets, rearranged during transfection (RET), fibroblast growth factor receptor (FGFR), receptor tyrosine kinase-like orphan signal 1 (ROS1), Mitogen-activated protein kinase (MAPK), and tropomyosin receptor kinase (TRK). These drugs target the tyrosine kinase domain of receptor tyrosine kinases and play a vital role in proliferation and migration of breast cancer cells. Several TKIs, including lapatinib, neratinib, and tucatinib, have been developed and are currently used in clinical settings, often in combination with chemotherapy, endocrine therapy, or other targeted agents. TKIs have demonstrated remarkable benefits in enhancing progression-free and overall survival in patients with breast cancer and have become a standard of care for this population. This review provides an overview of TKIs currently being examined in preclinical studies and clinical trials, especially in combination with drugs approved for breast cancer treatment. TKIs have emerged as a promising therapeutic option for patients with breast cancer and hold potential for treating other breast cancer subtypes. The development of new TKIs and their integration into personalized treatment strategies will continue to shape the future of breast cancer therapy.
Collapse
Affiliation(s)
- Vidya Sankarapandian
- Department of Microbiology and Immunology, Kampala International University, Western Campus, Box 20000, Uganda
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Conrad Ondieki Miruka
- Department of Biochemistry, Kampala International University, Western Campus, Box 20000, Uganda
| | - Poornima Sivamani
- Department of Pharmacology and Clinical pharmacology, Christian Medical College, Vellore 632004, India
| | - Balu Alagar Venmathi Maran
- Graduate School of Integrated Science and Technology, Nagasaki University, 1-14 Bunkyomachi, Nagasaki 852-8521, Japan
| | - Rajapandiyan Krishnamoorthy
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea..
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea..
| |
Collapse
|
9
|
Narlapati H, Speirs C, Jones RM, Berenberg J. Successful Treatment With Lorlatinib Monotherapy for Secondary Central Nervous System Oligometastatic Disease in Refractory Anaplastic Lymphoma Kinase Positive Non-small Cell Lung Cancer. Cureus 2024; 16:e73645. [PMID: 39677236 PMCID: PMC11645476 DOI: 10.7759/cureus.73645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
The anaplastic lymphoma kinase (ALK) gene plays crucial roles in both normal brain development and oncogenesis, particularly in non-small cell lung cancer (NSCLC). Metastatic ALK-positive NSCLC is characterized by ALK tyrosine kinase domain rearrangements, prompting the use of ALK tyrosine kinase inhibitors (TKIs) to target the mutation. While first-line treatment options include alectinib, brigatinib, and lorlatinib per National Comprehensive Cancer Network (NCCN) guidelines, therapeutic challenges arise in cases of disease progression. Management strategies may involve radiation therapy, switching to alternative ALK inhibitors, or testing for resistance mutations like ALK G1202R to guide treatment selection, with lorlatinib emerging as an alternative treatment option. Here, we present the case of a 35-year-old male diagnosed with metastatic ALK-positive NSCLC. Despite initial stability on alectinib therapy, disease progression necessitated therapeutic modification, including a switch to brigatinib and subsequent treatment with lorlatinib monotherapy. Notably, the patient achieved complete remission radiologically and clinically following treatment with lorlatinib, highlighting its efficacy in refractory disease settings. While molecular research supports lorlatinib's superior central nervous system (CNS) penetrability and systemic efficacy, the absence of head-to-head clinical trials presents a significant gap in evidence. Direct comparison of second and third-generation ALK inhibitors is essential to elucidate their comparative efficacy and adverse event profiles, which could refine current management guidelines. Furthermore, if lorlatinib proves superior in terms of progression-free survival, it may offer the potential to delay or obviate the need for radiation therapy, thus mitigating the risk of neurotoxic adverse events associated with these modalities.
Collapse
Affiliation(s)
- Hema Narlapati
- Internal Medicine, Tripler Army Medical Center, Honolulu, USA
| | | | - Ryan M Jones
- Hematology and Medical Oncology, Tripler Army Medical Center, Honolulu, USA
| | - Jeffrey Berenberg
- Hematology and Medical Oncology, Tripler Army Medical Center, Honolulu, USA
| |
Collapse
|
10
|
Elkhamisy FAA, Eesa AN, Elnady OM, Elnaghi KAEA, Foda AAM. Reduced expression of SOX11 in colorectal adenocarcinoma is associated with mucinous and signet ring cell types, poor survival, and lower ALK expression. Pathol Res Pract 2024; 260:155450. [PMID: 38986363 DOI: 10.1016/j.prp.2024.155450] [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: 02/03/2024] [Revised: 06/22/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Mucinous and signet ring cell colorectal carcinoma (m/srCRC) are challenging colorectal adenocarcinoma (CRC) types with poor prognosis. This study aimed to investigate SOX11 and ALK immunohistochemical expression in the m/srCRC group, comparing the results to those of nonmucinous CRC (nmCRC) and studying their association with different clinicopathological CRC features to better understand their significance and role. Besides, the study assesses which marker has a better predictive value for clinical practice. METHODS Tissue microarrays were prepared from 150 CRC blocks distributed equally between the m/srCRC and nmCRC groups. SOX11 and ALK immunohistochemical expressions were compared between both groups. In addition, their association with CRC clinicopathological data and survival was investigated. The Receiver Operating Characteristic (ROC) Curve analysis examined the predictive ability of SOX11 and ALK IHC expression for CRC mortality. RESULTS Both SOX11 and ALK expression were significantly reduced in m/srCRC compared to nmCRC. SOX11 is significantly associated with other prognostic clinicopathological factors (tumor size, lymph node status, overall TNM stage, grade, lymphovascular and perineural invasion) and overall survival. SOX11 significantly positively correlates with ALK expression. Using the ROC analysis, SOX11 is superior to ALK in survival prediction. CONCLUSION SOX11 can be used as a prognostic marker and is a suggested therapeutic target in mucinous and signet ring cell colorectal carcinoma through upregulation modulation.
Collapse
Affiliation(s)
| | - Ahmed Naeem Eesa
- Pathology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ola Mousa Elnady
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Mansoura University, Egypt
| | - Khaled Abd Elaziz Ahmed Elnaghi
- Oncology Centre, Medical Oncology unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt; Medical Oncology Department, Oncology Center King Abdullah Medical City, Makkah, Saudi Arabia
| | - Abd AlRahman Mohammad Foda
- Anatomic Pathology department, Faculty of Medicine, Mansoura University, Egypt; Department of Pathology, General Medicine Practice Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| |
Collapse
|
11
|
Tu Y, Gong J, Mou J, Jiang H, Zhao H, Gao J. Strategies for the development of stimuli-responsive small molecule prodrugs for cancer treatment. Front Pharmacol 2024; 15:1434137. [PMID: 39144632 PMCID: PMC11322083 DOI: 10.3389/fphar.2024.1434137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Approved anticancer drugs typically face challenges due to their narrow therapeutic window, primarily because of high systemic toxicity and limited selectivity for tumors. Prodrugs are initially inactive drug molecules designed to undergo specific chemical modifications. These modifications render the drugs inactive until they encounter specific conditions or biomarkers in vivo, at which point they are converted into active drug molecules. This thoughtful design significantly improves the efficacy of anticancer drug delivery by enhancing tumor specificity and minimizing off-target effects. Recent advancements in prodrug design have focused on integrating these strategies with delivery systems like liposomes, micelles, and polymerosomes to further improve targeting and reduce side effects. This review outlines strategies for designing stimuli-responsive small molecule prodrugs focused on cancer treatment, emphasizing their chemical structures and the mechanisms controlling drug release. By providing a comprehensive overview, we aim to highlight the potential of these innovative approaches to revolutionize cancer therapy.
Collapse
Affiliation(s)
- Yuxuan Tu
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jianbao Gong
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, China
| | - Jing Mou
- Department of Neonatology, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Hongfei Jiang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Haibo Zhao
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jiake Gao
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Chiodi D, Ishihara Y. The role of the methoxy group in approved drugs. Eur J Med Chem 2024; 273:116364. [PMID: 38781921 DOI: 10.1016/j.ejmech.2024.116364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 05/25/2024]
Abstract
The methoxy substituent is prevalent in natural products and, consequently, is present in many natural product-derived drugs. It has also been installed in modern drug molecules with no remnant of natural product features because medicinal chemists have been taking advantage of the benefits that this small functional group can bestow on ligand-target binding, physicochemical properties, and ADME parameters. Herein, over 230 methoxy-containing small-molecule drugs, as well as several fluoromethoxy-containing drugs, are presented from the vantage point of the methoxy group. Biochemical mechanisms of action, medicinal chemistry SAR studies, and numerous X-ray cocrystal structures are analyzed to identify the precise role of the methoxy group for many of the drugs and drug classes. Although the methoxy substituent can be considered as the hybridization of a hydroxy and a methyl group, the combination of these functionalities often results in unique effects that can amount to more than the sum of the individual parts.
Collapse
Affiliation(s)
- Debora Chiodi
- Department of Chemistry, Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, CA, 92121, USA
| | - Yoshihiro Ishihara
- Department of Chemistry, Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
13
|
Chen S, Gu J, Wu K, Zhao X, Lu Y. Progress in clinical diagnosis and treatment of colorectal cancer with rare genetic variants. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0026. [PMID: 38940668 PMCID: PMC11208903 DOI: 10.20892/j.issn.2095-3941.2024.0026] [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: 01/14/2024] [Accepted: 05/17/2024] [Indexed: 06/29/2024] Open
Abstract
Targeted therapy is crucial for advanced colorectal cancer (CRC) positive for genetic drivers. With advances in deep sequencing technology and new targeted drugs, existing standard molecular pathological detection systems and therapeutic strategies can no longer meet the requirements for careful management of patients with advanced CRC. Thus, rare genetic variations require diagnosis and targeted therapy in clinical practice. Rare gene mutations, amplifications, and rearrangements are usually associated with poor prognosis and poor response to conventional therapy. This review summarizes the clinical diagnosis and treatment of rare genetic variations, in genes including erb-b2 receptor tyrosine kinase 2 (ERBB2), B-Raf proto-oncogene, serine/threonine kinase (BRAF), ALK receptor tyrosine kinase/ROS proto-oncogene 1, receptor tyrosine kinase (ALK/ROS1), neurotrophic receptor tyrosine kinases (NTRKs), ret proto-oncogene (RET), fibroblast growth factor receptor 2 (FGFR2), and epidermal growth factor receptor (EGFR), to enhance understanding and identify more accurate personalized treatments for patients with rare genetic variations.
Collapse
Affiliation(s)
- Shuyi Chen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
- School of Basic Medical Sciences, Fourth Military Medical University, Xi’an 710032, China
| | - Jing Gu
- School of Basic Medical Sciences, Fourth Military Medical University, Xi’an 710032, China
| | - Kaichun Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
14
|
Peggion S, Najem S, Kolman JP, Reinshagen K, Pagerols Raluy L. Revisiting Neuroblastoma: Nrf2, NF-κB and Phox2B as a Promising Network in Neuroblastoma. Curr Issues Mol Biol 2024; 46:3193-3208. [PMID: 38666930 PMCID: PMC11048850 DOI: 10.3390/cimb46040200] [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: 02/28/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Neuroblastoma is the most common solid extracranial tumor during childhood; it displays extraordinary heterogeneous clinical courses, from spontaneous regression to poor outcome in high-risk patients due to aggressive growth, metastasizing, and treatment resistance. Therefore, the identification and detailed analysis of promising tumorigenic molecular mechanisms are inevitable. This review highlights the abnormal regulation of NF-κB, Nrf2, and Phox2B as well as their interactions among each other in neuroblastoma. NF-κB and Nrf2 play a key role in antioxidant responses, anti-inflammatory regulation and tumor chemoresistance. Recent studies revealed a regulation of NF-κB by means of the Nrf2/antioxidant response element (ARE) system. On the other hand, Phox2B contributes to the differentiation of immature sympathetic nervous system stem cells: this transcription factor regulates the expression of RET, thereby facilitating cell survival and proliferation. As observed in other tumors, we presume striking interactions between NF-κB, Nrf2, and Phox2B, which might constitute an important crosstalk triangle, whose decompensation may trigger a more aggressive phenotype. Consequently, these transcription factors could be a promising target for novel therapeutic approaches and hence, further investigation on their regulation in neuroblastoma shall be reinforced.
Collapse
Affiliation(s)
| | | | | | | | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Centre Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
15
|
Zhou X, Lei Q, Xiao Z, Song C, Deng H. Environment-Sensitive Fluorescent Probe Enables Assessment of Anaplastic Lymphoma Kinase Activity in Nonsmall Cell Lung Cancer. Anal Chem 2024; 96:3525-3534. [PMID: 38345335 DOI: 10.1021/acs.analchem.3c05337] [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: 02/28/2024]
Abstract
Anaplastic lymphoma kinase (ALK) rearrangements have been identified as key oncogenic drivers of a subset of nonsmall cell lung cancer (NSCLC). The final chimeric protein of the fusion gene can be constitutively activated, which accounts for the growth and proliferation of ALK-rearranged tumors and thus strongly associates with cancer invasion and metastasis. Diagnostic tools enabling the visualization of ALK activity in a structure-function-based approach are highly desirable to determine ALK status and guide ALK tyrosine kinase inhibitor (ALK-TKI) treatment making. Here, we describe the design, synthesis, and application of a new environment-sensitive fluorescent probe HX16 by introducing an environment-sensitive fluorophore 4-sulfonamidebenzoxadiazole to visualize ALK activity in living cancer cells and tumor tissue slices (mouse model and human biopsy sample). HX16 is a multifunctional chemical tool based on the pharmacophore of ALK-TKI (ceritinib) and can specifically target the kinase domain of ALK with a high sensitivity. Using flow cytometry and confocal microscopy, HX16 enables visualization of ALK activity in various cancer cells with distinct ALK fusion genes, as well as xenograft mouse models. Importantly, HX16 was also applied to visualize ALK activity in a tumor biopsy from a NSCLC patient with ALK-echinoderm microtubule-associated protein-like-4 fusion gene for prediction of ALK-TKI sensitivity. These results demonstrate that strategically designed ALK-TKI-based probe allows the assessment of ALK activity in tumor tissues and hold promise as a useful diagnostic tool in predicting ALK-TKI therapy response.
Collapse
Affiliation(s)
- Xinglong Zhou
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Lei
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhaolin Xiao
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chao Song
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan 610041, China
| | - Hui Deng
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Institute of Respiratory Health, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu, Sichuan 610041, China
| |
Collapse
|
16
|
Myint KZ, Balasubramanian B, Venkatraman S, Phimsen S, Sripramote S, Jantra J, Choeiphuk C, Mingphruedhi S, Muangkaew P, Rungsakulkij N, Tangtawee P, Suragul W, Farquharson WV, Wongprasert K, Chutipongtanate S, Sanvarinda P, Ponpuak M, Poungvarin N, Janvilisri T, Suthiphongchai T, Yacqub-Usman K, Grabowska AM, Bates DO, Tohtong R. Therapeutic Implications of Ceritinib in Cholangiocarcinoma beyond ALK Expression and Mutation. Pharmaceuticals (Basel) 2024; 17:197. [PMID: 38399413 PMCID: PMC10892566 DOI: 10.3390/ph17020197] [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: 01/08/2024] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a difficult-to-treat cancer, with limited therapeutic options and surgery being the only curative treatment. Standard chemotherapy involves gemcitabine-based therapies combined with cisplatin, oxaliplatin, capecitabine, or 5-FU with a dismal prognosis for most patients. Receptor tyrosine kinases (RTKs) are aberrantly expressed in CCAs encompassing potential therapeutic opportunity. Hence, 112 RTK inhibitors were screened in KKU-M213 cells, and ceritinib, an approved targeted therapy for ALK-fusion gene driven cancers, was the most potent candidate. Ceritinib's cytotoxicity in CCA was assessed using MTT and clonogenic assays, along with immunofluorescence, western blot, and qRT-PCR techniques to analyze gene expression and signaling changes. Furthermore, the drug interaction relationship between ceritinib and cisplatin was determined using a ZIP synergy score. Additionally, spheroid and xenograft models were employed to investigate the efficacy of ceritinib in vivo. Our study revealed that ceritinib effectively killed CCA cells at clinically relevant plasma concentrations, irrespective of ALK expression or mutation status. Ceritinib modulated multiple signaling pathways leading to the inhibition of the PI3K/Akt/mTOR pathway and activated both apoptosis and autophagy. Additionally, ceritinib and cisplatin synergistically reduced CCA cell viability. Our data show ceritinib as an effective treatment of CCA, which could be potentially explored in the other cancer types without ALK mutations.
Collapse
Affiliation(s)
- Kyaw Zwar Myint
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
| | - Brinda Balasubramanian
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Simran Venkatraman
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
| | - Suchada Phimsen
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (S.P.); (C.C.)
| | - Supisara Sripramote
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Jeranan Jantra
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Chaiwat Choeiphuk
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; (S.P.); (C.C.)
| | - Somkit Mingphruedhi
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Paramin Muangkaew
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Narongsak Rungsakulkij
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Pongsatorn Tangtawee
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Wikran Suragul
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Watoo Vassanasiri Farquharson
- Hepato-Pancreatic-Biliary Surgery Unit, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (S.M.); (P.M.); (N.R.); (P.T.); (W.S.); (W.V.F.)
| | - Kanokpan Wongprasert
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Somchai Chutipongtanate
- Division of Epidemiology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Pimtip Sanvarinda
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Marisa Ponpuak
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Naravat Poungvarin
- Department of Clinical Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Tavan Janvilisri
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (K.Z.M.); (B.B.); (S.V.); (T.J.)
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Tuangporn Suthiphongchai
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| | - Kiren Yacqub-Usman
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (K.Y.-U.); (A.M.G.); (D.O.B.)
| | - Anna M. Grabowska
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (K.Y.-U.); (A.M.G.); (D.O.B.)
| | - David O. Bates
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (K.Y.-U.); (A.M.G.); (D.O.B.)
| | - Rutaiwan Tohtong
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.S.); (J.J.); (T.S.)
| |
Collapse
|
17
|
Liu C, Zhou C, Xia W, Zhou Y, Qiu Y, Weng J, Zhou Q, Chen W, Wang YN, Lee HH, Wang SC, Kuang M, Yu D, Ren N, Hung MC. Targeting ALK averts ribonuclease 1-induced immunosuppression and enhances antitumor immunity in hepatocellular carcinoma. Nat Commun 2024; 15:1009. [PMID: 38307859 PMCID: PMC10837126 DOI: 10.1038/s41467-024-45215-0] [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: 10/20/2023] [Accepted: 01/18/2024] [Indexed: 02/04/2024] Open
Abstract
Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders among a cohort of ten patients who received Nivolumab (anti-PD-1 antibody). Our findings indicate that non-responders have a high abundance of secreted RNase1, which is associated with a poor prognosis in various cancer types. Furthermore, mice implanted with HCC cells that overexpress RNase1 exhibit immunosuppressive tumor microenvironments and diminished response to anti-PD-1 therapy. RNase1 induces the polarization of macrophages towards a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway. Targeting the RNase1/ALK axis reprograms the macrophage polarization, with increased CD8+ T- and Th1- cell recruitment. Moreover, simultaneous targeting of the checkpoint protein PD-1 unleashes cytotoxic CD8+ T-cell responses. Treatment utilizing both an ALK inhibitor and an anti-PD-1 antibody exhibits enhanced tumor regression and facilitates long-term immunity. Our study elucidates the role of RNase1 in mediating tumor resistance to immunotherapy and reveals an RNase1-mediated immunosuppressive tumor microenvironment, highlighting the potential of targeting RNase1 as a promising strategy for cancer immunotherapy in HCC.
Collapse
Affiliation(s)
- Chunxiao Liu
- Department of Liver Surgery, Center of Hepato-Pancreato-biliary Surgery, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Chenhao Zhou
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Yifan Zhou
- Department of laboratory medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yufan Qiu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jialei Weng
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Qiang Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Wanyong Chen
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan
| | - Ming Kuang
- Department of Liver Surgery, Center of Hepato-Pancreato-biliary Surgery, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ning Ren
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China.
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan.
| |
Collapse
|
18
|
Boor SA, Meisel JD, Kim DH. Neuroendocrine gene expression coupling of interoceptive bacterial food cues to foraging behavior of C. elegans. eLife 2024; 12:RP91120. [PMID: 38231572 PMCID: PMC10945577 DOI: 10.7554/elife.91120] [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] [Indexed: 01/18/2024] Open
Abstract
Animal internal state is modulated by nutrient intake, resulting in behavioral responses to changing food conditions. The neural mechanisms by which internal states are generated and maintained are not well understood. Here, we show that in the nematode Caenorhabditis elegans, distinct cues from bacterial food - interoceptive signals from the ingestion of bacteria and gustatory molecules sensed from nearby bacteria - act antagonistically on the expression of the neuroendocrine TGF-beta ligand DAF-7 from the ASJ pair of sensory neurons to modulate foraging behavior. A positive-feedback loop dependent on the expression of daf-7 from the ASJ neurons acts to promote transitions between roaming and dwelling foraging states and influence the persistence of roaming states. SCD-2, the C. elegans ortholog of mammalian anaplastic lymphoma kinase (ALK), which has been implicated in the central control of metabolism of mammals, functions in the AIA interneurons to regulate foraging behavior and cell-non-autonomously control the expression of DAF-7 from the ASJ neurons. Our data establish how a dynamic neuroendocrine daf-7 expression feedback loop regulated by SCD-2 functions to couple sensing and ingestion of bacterial food to foraging behavior. We further suggest that this neuroendocrine feedback loop underlies previously characterized exploratory behaviors in C. elegans. Our data suggest that the expression of daf-7 from the ASJ neurons contributes to and is correlated with an internal state of 'unmet need' that regulates exploratory foraging behavior in response to bacterial cues in diverse physiological contexts.
Collapse
Affiliation(s)
- Sonia A Boor
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Joshua D Meisel
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
| | - Dennis H Kim
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| |
Collapse
|
19
|
Jachowski A, Marcinkowski M, Szydłowski J, Grabarczyk O, Nogaj Z, Marcin Ł, Pławski A, Jagodziński PP, Słowikowski BK. Modern therapies of nonsmall cell lung cancer. J Appl Genet 2023; 64:695-711. [PMID: 37698765 PMCID: PMC10632224 DOI: 10.1007/s13353-023-00786-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
Lung cancer (LC), particularly nonsmall cell lung cancer (NSCLC), is one of the most prevalent types of neoplasia worldwide, regardless of gender, with the highest mortality rates in oncology. Over the years, treatment for NSCLC has evolved from conventional surgery, chemotherapy, and radiotherapy to more tailored and minimally invasive approaches. The use of personalised therapies has increased the expected efficacy of treatment while simultaneously reducing the frequency of severe adverse effects (AEs). In this review, we discuss established modern approaches, including immunotherapy and targeted therapy, as well as experimental molecular methods like clustered regularly interspaced short palindromic repeat (CRISPR) and nanoparticles. These emerging methods offer promising outcomes and shorten the recovery time for various patients. Recent advances in the diagnostic field, including imaging and genetic profiling, have enabled the implementation of these methods. The versatility of these modern therapies allows for multiple treatment options, such as single-agent use, combination with existing conventional treatments, or incorporation into new regimens. As a result, patients can survive even in the advanced stages of NSCLC, leading to increased survival indicators such as overall survival (OS) and progression-free survival (PFS).
Collapse
Affiliation(s)
- Andrzej Jachowski
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland
| | - Mikołaj Marcinkowski
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland
| | - Jakub Szydłowski
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland
| | - Oskar Grabarczyk
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland
| | - Zuzanna Nogaj
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland
| | - Łaz Marcin
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland
| | - Andrzej Pławski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32 Street, 60-479, Poznań, Poland
| | - Paweł Piotr Jagodziński
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland
| | - Bartosz Kazimierz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, Święcickiego 6 Street, 60-781, Poznań, Poland.
| |
Collapse
|
20
|
Moosburner M, Alibegovic L, Hasselmann K, Gaiderov A, Hildebrand J, Philippou-Massier J, Blum H, Fischer L, Dreyling M, Silkenstedt E. Combined treatment with crizotinib and temsirolimus is an effective strategy in mantle cell lymphoma and can overcome acquired resistance to temsirolimus. Hematol Oncol 2023; 41:858-868. [PMID: 37300279 DOI: 10.1002/hon.3194] [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: 01/20/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023]
Abstract
Constitutive activation of the PI3K/AKT/mTOR-pathway plays an important role in the pathogenesis of mantle cell lymphoma (MCL), leading to approval of the mTOR inhibitor temsirolimus for relapsed or refractory MCL. Yet, despite favorable initial response rates, early relapses under treatment have been observed. Therefore, understanding the underlying mechanisms of temsirolimus resistance and developing strategies to overcome it is highly warranted. Here, we established a new temsirolimus-resistant MCL cell line to evaluate the molecular background of resistance to this drug. Transcriptome profiling and gene set enrichment analysis comparing temsirolimus-sensitive and -resistant cell lines showed significant upregulation of PI3K/AKT/mTor-, RAS signaling- and the RTK-dependent PDGFR-, FGFR-, Met- and ALK-signaling-pathways in the resistant cells. Furthermore, MET, known as important proto-oncogene and mediator of drug resistance, was among the most upregulated genes in the resistant cells. Importantly, Met protein was overexpressed in both, MCL cells with acquired as well as intrinsic temsirolimus resistance, but could not be detected in any of the temsirolimus sensitive ones. Combined pharmacological inhibition of mTOR and Met signaling with temsirolimus and the RTK inhibitor crizotinib significantly restored sensitivity to temsirolimus. Furthermore, this combined treatment proved to be synergistic in all MCL cell lines investigated and was also active in primary MCL cells. In summary, we showed for the first time that overexpression of MET plays an important role for mediating temsirolimus resistance in MCL and combined treatment with temsirolimus and crizotinib is a very promising therapeutic approach for MCL and an effective strategy to overcome temsirolimus resistance.
Collapse
Affiliation(s)
- Marie Moosburner
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Munich, Germany
| | - Lamija Alibegovic
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Munich, Germany
| | - Korbinian Hasselmann
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Munich, Germany
| | - Anton Gaiderov
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Munich, Germany
| | - Johannes Hildebrand
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Munich, Germany
| | - Julia Philippou-Massier
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, University of Munich, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, University of Munich, Munich, Germany
| | - Luca Fischer
- Department of Medicine III, LMU University Hospital Großhadern of the Ludwig-Maximilians-University, Munich, Germany
| | - Martin Dreyling
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Munich, Germany
- Department of Medicine III, LMU University Hospital Großhadern of the Ludwig-Maximilians-University, Munich, Germany
| | - Elisabeth Silkenstedt
- Department of Medicine III, Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Ludwig-Maximilians-University, Munich, Germany
- Department of Medicine III, LMU University Hospital Großhadern of the Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
21
|
Balasundaram A, C Doss GP. Comparative Atomistic Insights on Apo and ATP-I1171N/S/T in Nonsmall-Cell Lung Cancer. ACS OMEGA 2023; 8:43856-43872. [PMID: 38027370 PMCID: PMC10666221 DOI: 10.1021/acsomega.3c05785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/21/2023] [Indexed: 12/01/2023]
Abstract
Anaplastic lymphoma kinase (ALK) rearrangements occur in about 5% of nonsmall cell lung cancer (NSCLC) patients. Despite being first recognized as EML4-ALK, fusions with several additional genes have been identified, all of which cause constitutive activation of the ALK kinase and subsequently lead to tumor development. ALK inhibitors first-line crizotinib, second-line ceritinib, and alectinib are effective against NSCLC patients with these rearrangements. Patients progressing on crizotinib had various mutations in the ALK kinase domain. ALK fusion proteins are activated by oligomerization through the fusion partner, which leads to the autophosphorylation of the kinase's domain and consequent downstream activation. The proposed computational study focuses on understanding the activation mechanism of ALK and ATP binding of wild-type (WT) and I1171N/S/T mutations. We analyzed the conformational change of ALK I1171N/S/T mutations and ATP binding using molecular docking and molecular dynamics simulation approaches. According to principal component analysis and free energy landscape, it is clear that I1171N/S/T mutations in Apo and ATP showed different energy minima/unstable structures compared to WT-Apo. The results revealed that I1171N/S/T mutations and ATP binding significantly supported a change toward an active-state conformation, whereas WT-Apo remained inactive. We demonstrated that I1171N/S/T mutations are persistent in an active state and independent of ATP. The I1171S/T mutations showed greater intermolecular H-bonds with ATP than WT-ATP. The molecular mechanics Poisson-Boltzmann surface area analysis revealed that the I1171N/S/T mutation binding energy was similar to that of WT-ATP. This study shows that I1171N/S/T can form stable bonds with ATP and may contribute to a constitutively active kinase. Based on the Y1278-C1097 H-bond and E1167-K1150 salt bridge interaction, I1171N strongly promotes the constitutively active kinase independent of ATP. This structural mechanism study will aid in understanding the oncogenic activity of ALK and the basis for improving the ALK inhibitors.
Collapse
Affiliation(s)
- Ambritha Balasundaram
- Laboratory of Integrative Genomics,
Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - George Priya C Doss
- Laboratory of Integrative Genomics,
Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
22
|
Boor SA, Meisel JD, Kim DH. Neuroendocrine Gene Expression Coupling of Interoceptive Bacterial Food Cues to Foraging Behavior of C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.15.549072. [PMID: 37503081 PMCID: PMC10369937 DOI: 10.1101/2023.07.15.549072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Animal internal state is modulated by nutrient intake, resulting in behavioral responses to changing food conditions. The neural mechanisms by which internal states are generated and maintained are not well understood. Here, we show that in the nematode Caenorhabditis elegans, distinct cues from bacterial food - interoceptive signals from the ingestion of bacteria and gustatory molecules sensed from nearby bacteria - act antagonistically on the expression of the neuroendocrine TGF-beta ligand DAF-7 from the ASJ pair of sensory neurons to modulate foraging behavior. A positive-feedback loop dependent on the expression of daf-7 from the ASJ neurons acts to promote transitions between roaming and dwelling foraging states and influence the persistence of roaming states. SCD-2, the C. elegans ortholog of mammalian Anaplastic Lymphoma Kinase (ALK), which has been implicated in the central control of metabolism of mammals, functions in the AIA interneurons to regulate foraging behavior and cell-non-autonomously control the expression of DAF-7 from the ASJ neurons. Our data establish how a dynamic neuroendocrine daf-7 expression feedback loop regulated by SCD-2 functions to couple sensing and ingestion of bacterial food to foraging behavior. We further suggest that this neuroendocrine feedback loop underlies previously characterized exploratory behaviors in C. elegans. Our data suggest that the expression of daf-7 from the ASJ neurons contributes to and is correlated with an internal state of "unmet need" that regulates exploratory foraging behavior in response to bacterial cues in diverse physiological contexts.
Collapse
Affiliation(s)
- Sonia A. Boor
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Joshua D. Meisel
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Dennis H. Kim
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
23
|
Gao H, Zhang JY, Zhao LJ, Guo YY. Synthesis and clinical application of small-molecule inhibitors and PROTACs of anaplastic lymphoma kinase. Bioorg Chem 2023; 140:106807. [PMID: 37651895 DOI: 10.1016/j.bioorg.2023.106807] [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: 06/12/2023] [Revised: 07/13/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023]
Abstract
Pharmacological interventions that specifically target protein products of oncogenes in tumors have surfaced as a propitious therapeutic approach. Among infrequent genetic alterations, rearrangements of the anaplastic lymphoma kinase (ALK) gene, typically involving a chromosome 2 inversion that culminates in a fusion with the echinoderm microtubule-associated protein like 4 (EML4), lead to anomalous expression and activation of ALK. The inhibition of autophosphorylation and subsequent blockade of signal transduction by ALK tyrosine kinase inhibitors (TKIs) has been observed to elicit anti-tumor effects. Currently, four generations of ALK-positive targeted drugs have been investigated, providing a promising outlook for patients. The aim of this review is to furnish a comprehensive survey of the synthesis and clinical application of prototypical small-molecule ALK inhibitors in both preclinical and clinical phases, offering guidance for further development of ALK inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Hua Gao
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jing-Yi Zhang
- The Rogel Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States; College of Chemistry and Chemical Engineering, Zhengzhou Normal University 450044, China.
| | - Li-Jie Zhao
- The Rogel Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Yuan-Yuan Guo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
24
|
Ploenzke M, Irizarry R. Reassessing pharmacogenomic cell sensitivity with multilevel statistical models. Biostatistics 2023; 24:901-921. [PMID: 35277956 PMCID: PMC10583722 DOI: 10.1093/biostatistics/kxac010] [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: 08/15/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2023] Open
Abstract
Pharmacogenomic experiments allow for the systematic testing of drugs, at varying dosage concentrations, to study how genomic markers correlate with cell sensitivity to treatment. The first step in the analysis is to quantify the response of cell lines to variable dosage concentrations of the drugs being tested. The signal to noise in these measurements can be low due to biological and experimental variability. However, the increasing availability of pharmacogenomic studies provides replicated data sets that can be leveraged to gain power. To do this, we formulate a hierarchical mixture model to estimate the drug-specific mixture distributions for estimating cell sensitivity and for assessing drug effect type as either broad or targeted effect. We use this formulation to propose a unified approach that can yield posterior probability of a cell being susceptible to a drug conditional on being a targeted effect or relative effect sizes conditioned on the cell being broad. We demonstrate the usefulness of our approach via case studies. First, we assess pairwise agreements for cell lines/drugs within the intersection of two data sets and confirm the moderate pairwise agreement between many publicly available pharmacogenomic data sets. We then present an analysis that identifies sensitivity to the drug crizotinib for cells harboring EML4-ALK or NPM1-ALK gene fusions, as well as significantly down-regulated cell-matrix pathways associated with crizotinib sensitivity.
Collapse
Affiliation(s)
- Matt Ploenzke
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Building 2, 4th Floor, Boston, MA 02115
| | - Rafael Irizarry
- Department of Data Science, Dana Farber Cancer Institute, 450 Brookline Ave, CLSB 11007, Boston, MA 02215
| |
Collapse
|
25
|
Kannampuzha S, Murali R, Gopalakrishnan AV, Mukherjee AG, Wanjari UR, Namachivayam A, George A, Dey A, Vellingiri B. Novel biomolecules in targeted cancer therapy: a new approach towards precision medicine. Med Oncol 2023; 40:323. [PMID: 37804361 DOI: 10.1007/s12032-023-02168-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/18/2023] [Indexed: 10/09/2023]
Abstract
Cancer is a major threat to human life around the globe, and the discovery of novel biomolecules continue to be an urgent therapeutic need that is still unmet. Precision medicine relies on targeted therapeutic strategies. Researchers are better equipped to develop therapies that target proteins as they understand more about the genetic alterations and molecules that cause progression of cancer. There has been a recent diversification of the sorts of targets exploited in treatment. Therapeutic antibody and biotechnology advancements enabled curative treatments to reach previously inaccessible sites. New treatment strategies have been initiated for several undruggable targets. The application of tailored therapy has been proven to have efficient results in controlling cancer progression. Novel biomolecules like SMDCs, ADCs, mABs, and PROTACS has gained vast attention in the recent years. Several studies have shown that using these novel technology helps in reducing the drug dosage as well as to overcome drug resistance in different cancer types. Therefore, it is crucial to fully untangle the mechanism and collect evidence to understand the significance of these novel drug targets and strategies. This review article will be discussing the importance and role of these novel biomolecules in targeted cancer therapies.
Collapse
Affiliation(s)
- Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, India
| | - Abhijit Dey
- Department of Medical Services, MGM Cancer Institute, Chennai, Tamil Nadu, 600029, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| |
Collapse
|
26
|
Choudhary D, Kaur A, Singh P, Chaudhary G, Kaur R, Bayan MF, Chandrasekaran B, Marji SM, Ayman R. Target protein degradation by protacs: A budding cancer treatment strategy. Pharmacol Ther 2023; 250:108525. [PMID: 37696366 DOI: 10.1016/j.pharmthera.2023.108525] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023]
Abstract
Cancer is one of the most common causes of death. So, its lethal effect increases with time. Near about hundreds of cancers are known in humans. Cancer treatment is done to cure or prolonged remission, and shrinkage of the tumor. Cytotoxic agents, biological agents/targeted drugs, hormonal drugs, surgery, radiotherapy/proton therapy, chemotherapy, immunotherapy, and gene therapy are currently used in the treatment of cancer but their cost is high and cause various side effects. Seeing this, some new targeted strategies such as PROTACs are the need of the time. Proteolysis targeting chimera (PROTAC) has become one of the most discussed topics regarding cancer treatment. Few of the PROTAC molecules are in the trial phases. PROTACs have many advantages over other strategies such as modularity, compatibility, sub-stoichiometric activity, acting on undruggable targets, molecular design, and acts on intracellular targets, selectivity and specificity can be recruited for any cancer, versatility, and others. PROTACs are having some unclear questions on their pharmacokinetics, heavy-molecular weight, etc. PROTACs are anticipated to bring about a conversion in current healthcare and will emerge as booming treatments. In this review article we summarize PROTACs, their mechanism of action, uses, advantages, disadvantages, challenges, and future aspects for the successful development of potent PROTACs as a drug strategy.
Collapse
Affiliation(s)
- Diksha Choudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Amritpal Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Pargat Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Gaurav Chaudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | - Mohammad F Bayan
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | | | - Saeed M Marji
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | - Reema Ayman
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| |
Collapse
|
27
|
Oyeneyin OE, Ibrahim A, Ipinloju N, Ademoyegun AJ, Ojo ND. Insight into the corrosion inhibiting potential and anticancer activity of 1-(4-methoxyphenyl)-5-methyl-N'-(2-oxoindolin-3-ylidene)-1H-1,2,3-triazole-4-carbohydrazide via computational approaches. J Biomol Struct Dyn 2023; 42:11149-11166. [PMID: 37747068 DOI: 10.1080/07391102.2023.2260491] [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: 06/26/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023]
Abstract
Cancer is a major health concern globally. Orthodox and traditional medicine have actively been explored to manage this disease. Also, corrosion is a natural catastrophe that weakens and deteriorates metallic structures and their alloys causing major structural failures and severe economic implications. Designing and exploring multi-functional materials are beneficial since they are adaptive to different fields including engineering and pharmaceutics. In this study, we examined the anti-corrosion and anti-cancer potentials of 1-(4-methoxyphenyl)-5-methyl-N'-(2-oxoindolin-3-ylidene)-1H-1,2,3-triazole-4-carbohydrazide (MAC) using computational approaches. The molecular reactivity descriptors and charge distribution parameters of MAC were studied in gas and water at density functional theory (DFT) at B3LYP/6-311++G(d,p) theory level. The binding and mechanism of interaction between MAC and iron surface was studied using Monte Carlo (MC) and molecular dynamics (MD) simulation in hydrochloric acid medium. From the DFT, MC, and MD simulations, it was observed that MAC interacted spontaneously with iron surface essentially via van der Waal and electrostatic interactions. The near-parallel alignment of the corrosion inhibitor on iron plane facilitates its adsorption and isolation of the metal surface from the acidic solution. Further, the compound was docked in the binding pocket of anaplastic lymphoma kinase (ALK: 4FNZ) protein to assess its anti-cancer potential. The binding score, pharmacokinetics, and drug-likeness of MAC were compared with the reference drug (Crizotinib). The MAC displayed binding scores of -5.729 kcal/mol while Crizotinib has -3.904 kcal/mol. MD simulation of the complexes revealed that MAC is more stable and exhibits more favourable hydrogen bonding with the ALK receptor's active site than Crizotinib.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Oluwatoba Emmanuel Oyeneyin
- Theoretical and Computational Chemistry Unit, Adekunle Ajasin University, Akungba-Akoko, Nigeria
- School of Chemistry and Physics, University of Kwazulu-Natal, Durban, South Africa
| | - Abdulwasiu Ibrahim
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto Nigeria
| | - Nureni Ipinloju
- Theoretical and Computational Chemistry Unit, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | - Adeniyi John Ademoyegun
- Theoretical and Computational Chemistry Unit, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| | | |
Collapse
|
28
|
Wang J, Al-Majid D, Brenner JC, Smith JD. Mutant HRas Signaling and Rationale for Use of Farnesyltransferase Inhibitors in Head and Neck Squamous Cell Carcinoma. Target Oncol 2023; 18:643-655. [PMID: 37665491 DOI: 10.1007/s11523-023-00993-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are often associated with poor outcomes, due at least in part to the limited number of treatment options available for those patients who develop recurrent and/or metastatic disease (R/M HNSCC). Even with the recent validation and approval of immunotherapies in the first-line setting for these patients, the need for the development of new and alternative precision medicine strategies with survival benefit is clear. Oncogenic alterations in the HRAS (Harvey rat sarcoma virus) proto-oncogene are seen in approximately 4-8% of R/M HNSCC tumors. Recently, several preclinical and clinical advancements have been made in the implementation of small-molecule inhibitors that block post-translational farnesylation of HRas, thereby abrogating its downstream oncogenic activity. In this review, we focus on the biology of wild-type and mutant HRas signaling in HNSCC, and rationale for use and outcomes of farnesyltransferase inhibitors in patients with HRAS-mutant tumors.
Collapse
Affiliation(s)
- Jiayu Wang
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dana Al-Majid
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, MSRB III 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| | - J Chad Brenner
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, MSRB III 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Joshua D Smith
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, MSRB III 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| |
Collapse
|
29
|
Chen J, Wang J. Case report: ALK D1225N missense mutation in lung adenocarcinoma responds to tyrosine kinase inhibitors. Front Pharmacol 2023; 14:1190447. [PMID: 37529699 PMCID: PMC10387523 DOI: 10.3389/fphar.2023.1190447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023] Open
Abstract
ALK gene missense mutations are conventionally considered non-driver mutations without pathological significance, and therefore, there is a lack of effective target drugs against them. The standard treatment option for patients with ALK missense mutations is chemotherapy with or without antiangiogenic agents, which usually results in unsatisfactory outcomes. Herein, we present the case of a patient with metastatic lung adenocarcinoma harboring the only missense mutation in ALK D1225N responding to two ALK-tyrosine kinase inhibitors (TKIs), namely, crizotinib and ensartinib. Our case highlights that non-small cell lung cancer (NSCLC) patients harboring the D1225N mutation may benefit from ALK-TKIs, and therefore, ALK-TKIs should be considered candidates for further line treatment.
Collapse
Affiliation(s)
- Jianxin Chen
- Department of Medical Oncology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| | - Junhui Wang
- Department of Radiation Oncology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| |
Collapse
|
30
|
Mastini C, Campisi M, Patrucco E, Mura G, Ferreira A, Costa C, Ambrogio C, Germena G, Martinengo C, Peola S, Mota I, Vissio E, Molinaro L, Arigoni M, Olivero M, Calogero R, Prokoph N, Tabbò F, Shoji B, Brugieres L, Geoerger B, Turner SD, Cuesta-Mateos C, D’Aliberti D, Mologni L, Piazza R, Gambacorti-Passerini C, Inghirami GG, Chiono V, Kamm RD, Hirsch E, Koch R, Weinstock DM, Aster JC, Voena C, Chiarle R. Targeting CCR7-PI3Kγ overcomes resistance to tyrosine kinase inhibitors in ALK-rearranged lymphoma. Sci Transl Med 2023; 15:eabo3826. [PMID: 37379367 PMCID: PMC10804420 DOI: 10.1126/scitranslmed.abo3826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) show potent efficacy in several ALK-driven tumors, but the development of resistance limits their long-term clinical impact. Although resistance mechanisms have been studied extensively in ALK-driven non-small cell lung cancer, they are poorly understood in ALK-driven anaplastic large cell lymphoma (ALCL). Here, we identify a survival pathway supported by the tumor microenvironment that activates phosphatidylinositol 3-kinase γ (PI3K-γ) signaling through the C-C motif chemokine receptor 7 (CCR7). We found increased PI3K signaling in patients and ALCL cell lines resistant to ALK TKIs. PI3Kγ expression was predictive of a lack of response to ALK TKI in patients with ALCL. Expression of CCR7, PI3Kγ, and PI3Kδ were up-regulated during ALK or STAT3 inhibition or degradation and a constitutively active PI3Kγ isoform cooperated with oncogenic ALK to accelerate lymphomagenesis in mice. In a three-dimensional microfluidic chip, endothelial cells that produce the CCR7 ligands CCL19/CCL21 protected ALCL cells from apoptosis induced by crizotinib. The PI3Kγ/δ inhibitor duvelisib potentiated crizotinib activity against ALCL lines and patient-derived xenografts. Furthermore, genetic deletion of CCR7 blocked the central nervous system dissemination and perivascular growth of ALCL in mice treated with crizotinib. Thus, blockade of PI3Kγ or CCR7 signaling together with ALK TKI treatment reduces primary resistance and the survival of persister lymphoma cells in ALCL.
Collapse
Affiliation(s)
- Cristina Mastini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Marco Campisi
- Dana Farber Cancer Institute, Boston, MA 02115, USA
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Mechanical and Aerospace Engineering, Politecnico of Torino, Torino 10129, Italy
| | - Enrico Patrucco
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Giulia Mura
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Antonio Ferreira
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Carlotta Costa
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Giulia Germena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Cinzia Martinengo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Silvia Peola
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Ines Mota
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Elena Vissio
- Department of Oncology, University of Torino, Orbassano, Torino 10043, Italy
| | - Luca Molinaro
- Department of Medical Science, University of Torino, Torino 10126, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Martina Olivero
- Department of Oncology, University of Torino, Orbassano, Torino 10043, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino 10060, Italy
| | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Nina Prokoph
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Fabrizio Tabbò
- Department of Pathology, Cornell University, New York NY 10121, USA
| | - Brent Shoji
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Laurence Brugieres
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif 94805, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, Paris-Saclay University, Villejuif 94805, France
- Université Paris-Saclay, INSERM U1015, Villejuif 94805, France
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
- Faculty of Medicine, Masaryk University, Brno 601 77, Czech Republic
| | - Carlos Cuesta-Mateos
- Department of Pre-Clinical Development, Catapult Therapeutics B.V., 8243 RC, Lelystad, Netherlands
| | - Deborah D’Aliberti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza 20900, Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza 20900, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza 20900, Italy
| | | | | | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico of Torino, Torino 10129, Italy
| | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Raphael Koch
- Dana Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- University Medical Center Göttingen, 37075 Göttingen, Germany
| | - David M. Weinstock
- Dana Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jon C. Aster
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
31
|
Berry MA, Bland AR, Ashton JC. Mechanisms of synergistic suppression of ALK-positive lung cancer cell growth by the combination of ALK and SHP2 inhibitors. Sci Rep 2023; 13:10041. [PMID: 37339995 DOI: 10.1038/s41598-023-37006-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023] Open
Abstract
Lung cancer is a major cause of cancer-related deaths. Alectinib is the first line of treatment for patients with ALK-positive lung cancer, but the survival rate beyond 2-3 years is low. Co-targeting secondary oncogenic drivers such as SHP2 is a potential strategy for improving drug efficacy. This is because SHP2 is expressed ubiquitously, but ALK expression is largely restricted to cancer cells. Thus, the combination of ALK and SHP2 inhibitors may provide a way to restrict synergistic cytotoxicity to cancer cells only, by reducing the dose of SHP2 inhibitors required for anticancer action and minimising SHP2-dependent systemic toxicity. The objective of this study was to investigate whether the combination of a SHP2 inhibitor (SHP099) with alectinib would synergistically suppress the growth of ALK-positive lung cancer cells. Our results demonstrated that the drug combination significantly and synergistically decreased cell viability at relatively low concentrations in ALK-positive H3122 and H2228 cells, due to G1 cell cycle arrest and increased apoptosis because of suppressed downstream RAS/MAPK signalling. The drug combination also induced the expression of mediators of the intrinsic apoptotic pathway, Bim and cleaved caspase-3, and modulated the expression of cell cycle mediators cyclin D1, cyclin B1, and phosphorylated CDK1.
Collapse
Affiliation(s)
- M A Berry
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - A R Bland
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - J C Ashton
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
32
|
Cai J, Sun B, Yu S, Zhang H, Zhang W. Heck Macrocyclization in Forging Non-Natural Large Rings including Macrocyclic Drugs. Int J Mol Sci 2023; 24:ijms24098252. [PMID: 37175956 PMCID: PMC10179193 DOI: 10.3390/ijms24098252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The intramolecular Heck reaction is a well-established strategy for natural product total synthesis. When constructing large rings, this reaction is also referred to as Heck macrocyclization, which has proved a viable avenue to access diverse naturally occurring macrocycles. Less noticed but likewise valuable, it has created novel macrocycles of non-natural origin that neither serve as nor derive from natural products. This review presents a systematic account of the title reaction in forging this non-natural subset of large rings, thereby addressing a topic rarely covered in the literature. Walking through two complementary sections, namely (1) drug discovery research and (2) synthetic methodology development, it demonstrates that beyond the well-known domain of natural product synthesis, Heck macrocyclization also plays a remarkable role in forming synthetic macrocycles, in particular macrocyclic drugs.
Collapse
Affiliation(s)
- Jiayou Cai
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, China
| | - Bin Sun
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, China
| | - Siqi Yu
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, China
| | - Han Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, China
| | - Weicheng Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, China
| |
Collapse
|
33
|
Kikawa C, Ketterl TG, Liu PhD YJ, Reed RC, Dahl JP. Pediatric Low-Grade Spindle Cell Neoplasm With A Novel AK5::ALK Fusion: A Case Report. Ann Otol Rhinol Laryngol 2023; 132:470-475. [PMID: 35502464 DOI: 10.1177/00034894221092207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Spindle cell neoplasms (SCN) share a single commonality of spindle-shaped cells on histopathology but are diverse in etiology. Expanding our collective knowledge of these neoplasms could further research in targeted therapies. We present a case of pediatric cutaneous SCN with a novel etiology, and the methods used to identify its origination. CASE PRESENTATION AND RESULTS A 1.5-year-old child presented with a 7-month history of a rapidly enlarging, erythematous, non-painful scalp mass without ulceration or bleeding. The child underwent ultrasound and magnetic resonance imaging, revealing a 2.9 × 3 × 2 cm vascular mass without intracranial connections. The mass was successfully resected at surgery. Subsequent histopathologic and genetic testing indicated a SCN harboring a previously undescribed gene rearrangement between adenylate kinase 5 (AK5) and anaplastic lymphoma kinase (ALK). The patient received close clinical follow-up and at 6 months post-surgery had no recurrent disease. CONCLUSIONS ALK rearrangements are common amongst many tumor types, but to our knowledge, AK5::ALK rearrangement has never been reported in SCN. Considering the rapid development of targeted clinical therapies, including those targeting ALK activity, this finding could be significant in the treatment of future patients with similar clinicopathologic and genetic presentation.
Collapse
Affiliation(s)
- Caroline Kikawa
- University of Washington School of Medicine, Seattle, WA, USA
| | - Tyler G Ketterl
- Division of Hematology-Oncology, Seattle Children's Hospital, Seattle, WA, USA
| | - Yajuan J Liu PhD
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Robyn C Reed
- Department of Laboratory Medicine, Seattle Children's Hospital, Seattle, WA, USA
| | - John P Dahl
- Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA, USA.,Division of Pediatric Otolaryngology-Head and Neck Surgery, Seattle Children's Hospital, Seattle, WA, USA
| |
Collapse
|
34
|
Navarrete-Meneses MDP, Salas-Labadía C, Juárez-Velázquez MDR, Moreno-Lorenzana D, Gómez-Chávez F, Olaya-Vargas A, Pérez-Vera P. Exposure to Insecticides Modifies Gene Expression and DNA Methylation in Hematopoietic Tissues In Vitro. Int J Mol Sci 2023; 24:6259. [PMID: 37047231 PMCID: PMC10094043 DOI: 10.3390/ijms24076259] [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: 01/30/2023] [Revised: 03/03/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
The evidence supporting the biological plausibility of the association of permethrin and malathion with hematological cancer is limited and contradictory; thus, further studies are needed. This study aimed to investigate whether in vitro exposure to 0.1 μM permethrin and malathion at 0, 24, 48 and 72 h after cell culture initiation induced changes in the gene expression and DNA methylation in mononuclear cells from bone marrow and peripheral blood (BMMCs, PBMCs). Both pesticides induced several gene expression modifications in both tissues. Through gene ontology analysis, we found that permethrin deregulates ion channels in PBMCs and BMMCs and that malathion alters genes coding proteins with nucleic acid binding capacity, which was also observed in PBMCs exposed to permethrin. Additionally, we found that both insecticides deregulate genes coding proteins with chemotaxis functions, ion channels, and cytokines. Several genes deregulated in this study are potentially associated with cancer onset and development, and some of them have been reported to be deregulated in hematological cancer. We found that permethrin does not induce DNA hypermethylation but can induce hypomethylation, and that malathion generated both types of events. Our results suggest that these pesticides have the potential to modify gene expression through changes in promoter DNA methylation and potentially through other mechanisms that should be investigated.
Collapse
Affiliation(s)
- María del Pilar Navarrete-Meneses
- Laboratorio de Genética y Cáncer, Instituto Nacional de Pediatría, Mexico City 04530, Mexico; (M.d.P.N.-M.); (C.S.-L.); (M.d.R.J.-V.); (D.M.-L.)
| | - Consuelo Salas-Labadía
- Laboratorio de Genética y Cáncer, Instituto Nacional de Pediatría, Mexico City 04530, Mexico; (M.d.P.N.-M.); (C.S.-L.); (M.d.R.J.-V.); (D.M.-L.)
| | - María del Rocío Juárez-Velázquez
- Laboratorio de Genética y Cáncer, Instituto Nacional de Pediatría, Mexico City 04530, Mexico; (M.d.P.N.-M.); (C.S.-L.); (M.d.R.J.-V.); (D.M.-L.)
| | - Dafné Moreno-Lorenzana
- Laboratorio de Genética y Cáncer, Instituto Nacional de Pediatría, Mexico City 04530, Mexico; (M.d.P.N.-M.); (C.S.-L.); (M.d.R.J.-V.); (D.M.-L.)
| | - Fernando Gómez-Chávez
- Maestría y Doctorado en Ciencia y Tecnología de Vacunas y Bioterapéuticos, Doctorado en Ciencias en Biotecnología, Laboratorio de Enfermedades Osteoarticulares e Inmunológicas, Instituto Politécnico Nacional-ENMyH, Mexico City 07738, Mexico;
| | - Alberto Olaya-Vargas
- Unidad de Trasplante de Células Hematopoyeticas y Terapia Celular, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Patricia Pérez-Vera
- Laboratorio de Genética y Cáncer, Instituto Nacional de Pediatría, Mexico City 04530, Mexico; (M.d.P.N.-M.); (C.S.-L.); (M.d.R.J.-V.); (D.M.-L.)
| |
Collapse
|
35
|
Mazzeschi M, Sgarzi M, Romaniello D, Gelfo V, Cavallo C, Ambrosi F, Morselli A, Miano C, Laprovitera N, Girone C, Ferracin M, Santi S, Rihawi K, Ardizzoni A, Fiorentino M, D’Uva G, Győrffy B, Palmer R, Lauriola M. The autocrine loop of ALK receptor and ALKAL2 ligand is an actionable target in consensus molecular subtype 1 colon cancer. J Exp Clin Cancer Res 2022; 41:113. [PMID: 35351152 PMCID: PMC8962179 DOI: 10.1186/s13046-022-02309-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/03/2022] [Indexed: 12/25/2022] Open
Abstract
Background In the last years, several efforts have been made to classify colorectal cancer (CRC) into well-defined molecular subgroups, representing the intrinsic inter-patient heterogeneity, known as Consensus Molecular Subtypes (CMSs). Methods In this work, we performed a meta-analysis of CRC patients stratified into four CMSs. We identified a negative correlation between a high level of anaplastic lymphoma kinase (ALK) expression and relapse-free survival, exclusively in CMS1 subtype. Stemming from this observation, we tested cell lines, patient-derived organoids and mice with potent ALK inhibitors, already approved for clinical use. Results ALK interception strongly inhibits cell proliferation already at nanomolar doses, specifically in CMS1 cell lines, while no effect was found in CMS2/3/4 groups. Furthermore, in vivo imaging identified a role for ALK in the dynamic formation of 3D tumor spheroids. Consistently, ALK appeares constitutively phosphorylated in CMS1, and it signals mainly through the AKT axis. Mechanistically, we found that CMS1 cells display several copies of ALKAL2 ligand and ALK-mRNAs, suggesting an autocrine loop mediated by ALKAL2 in the activation of ALK pathway, responsible for the invasive phenotype. Consequently, disruption of ALK axis mediates the pro-apoptotic action of CMS1 cell lines, both in 2D and 3D and enhanced cell-cell adhesion and e-cadherin organization. In agreement with all these findings, the ALK signature encompassing 65 genes statistically associated with worse relapse-free survival in CMS1 subtype. Finally, as a proof of concept, the efficacy of ALK inhibition was demonstrated in both patient-derived organoids and in tumor xenografts in vivo. Conclusions Collectively, these findings suggest that ALK targeting may represent an attractive therapy for CRC, and CMS classification may provide a useful tool to identify patients who could benefit from this treatment. These findings offer rationale and pharmacological strategies for the treatment of CMS1 CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02309-1.
Collapse
|
36
|
Chu YY, Chen MK, Wei Y, Lee HH, Xia W, Wang YN, Yam C, Hsu JL, Wang HL, Chang WC, Yamaguchi H, Jiang Z, Liu C, Li CF, Nie L, Chan LC, Gao Y, Wang SC, Liu J, Westin SN, Lee S, Sood AK, Yang L, Hortobagyi GN, Yu D, Hung MC. Targeting the ALK-CDK9-Tyr19 kinase cascade sensitizes ovarian and breast tumors to PARP inhibition via destabilization of the P-TEFb complex. NATURE CANCER 2022; 3:1211-1227. [PMID: 36253486 PMCID: PMC9586872 DOI: 10.1038/s43018-022-00438-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/24/2022] [Indexed: 12/28/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors have demonstrated promising clinical activity in multiple cancers. However, resistance to PARP inhibitors remains a substantial clinical challenge. In the present study, we report that anaplastic lymphoma kinase (ALK) directly phosphorylates CDK9 at tyrosine-19 to promote homologous recombination (HR) repair and PARP inhibitor resistance. Phospho-CDK9-Tyr19 increases its kinase activity and nuclear localization to stabilize positive transcriptional elongation factor b and activate polymerase II-dependent transcription of HR-repair genes. Conversely, ALK inhibition increases ubiquitination and degradation of CDK9 by Skp2, an E3 ligase. Notably, combination of US Food and Drug Administration-approved ALK and PARP inhibitors markedly reduce tumor growth and improve survival of mice in PARP inhibitor-/platinum-resistant tumor xenograft models. Using human tumor biospecimens, we further demonstrate that phosphorylated ALK (p-ALK) expression is associated with resistance to PARP inhibitors and positively correlated with p-Tyr19-CDK9 expression. Together, our findings support a biomarker-driven, combinatorial treatment strategy involving ALK and PARP inhibitors to induce synthetic lethality in PARP inhibitor-/platinum-resistant tumors with high p-ALK-p-Tyr19-CDK9 expression.
Collapse
Affiliation(s)
- Yu-Yi Chu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mei-Kuang Chen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clinton Yam
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hung-Ling Wang
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chao Chang
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Hirohito Yamaguchi
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Zhou Jiang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chunxiao Liu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Fei Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lei Nie
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li-Chuan Chan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuan Gao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Jinsong Liu
- Department of Anatomic Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanghoon Lee
- Department of Systems Biology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan.
- Department of Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
37
|
Inhibiting ALK-TOPK signaling pathway promotes cell apoptosis of ALK-positive NSCLC. Cell Death Dis 2022; 13:828. [PMID: 36167821 PMCID: PMC9515217 DOI: 10.1038/s41419-022-05260-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 01/23/2023]
Abstract
T-LAK cell-oriented protein kinase (TOPK) is a potential therapeutic target in tumors. However, its role in anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC) has not been reported. Here, we found that TOPK was highly expressed in ALK-positive NSCLC. Additionally, ALK was identified as another upstream kinase of TOPK by in vitro kinase assay screening. Then, it was proven that ALK phosphorylated TOPK at Y74 in vitro and ex vivo, and the pathways downstream of ALK-TOPK were explored by phosphoproteomic analysis. Subsequently, we demonstrated that inhibiting TOPK enhanced tumor sensitivity to alectinib (an ALK inhibitor). The combination of alectinib and HI-032 (a TOPK inhibitor) suppressed the growth and promoted the apoptosis of ALK-positive NSCLC cells ex vivo and in vivo. Our findings reveal a novel ALK-TOPK signaling pathway in ALK-positive NSCLC. The combination of alectinib and HI-032 might be a promising therapeutic strategy for improving the sensitivity of ALK-positive NSCLC to targeted therapy.
Collapse
|
38
|
Mesquita FP, Souza PFN, da Silva EL, Lima LB, de Oliveira LLB, Moreira-Nunes CA, Zuercher WJ, Burbano RMR, de Moraes MEA, Montenegro RC. Kinase Inhibitor Screening Displayed ALK as a Possible Therapeutic Biomarker for Gastric Cancer. Pharmaceutics 2022; 14:pharmaceutics14091841. [PMID: 36145589 PMCID: PMC9501214 DOI: 10.3390/pharmaceutics14091841] [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: 07/20/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022] Open
Abstract
Despite advances in cancer chemotherapy, gastric cancer (GC) continues to have high recurrence rates and poor prognosis with limited treatment options. Understanding the etiology of GC and developing more effective, less harmful therapeutic approaches are vital and urgent. Therefore, this work describes a novel kinase target in malignant gastric cells as a potential therapeutic strategy. Our results demonstrate that among 147 kinase inhibitors (KI), only three molecules were significantly cytotoxic for the AGP-01 cell line. Hence, these three molecules were further characterized in their cellular mode of action. There was significant cell cycle impairment due to the expression modulation of genes such as TP53, CDKN1A, CDC25A, MYC, and CDK2 with subsequent induction of apoptosis. In fact, the Gene Ontology analysis revealed a significant enrichment of pathways related to cell cycle regulation (GO:1902749 and GO:1903047). Moreover, the three selected KIs significantly reduced cell migration and Vimentin mRNA expression after treatment. Surprisingly, the three KIs share the same target, ALK and INSR, but only the ALK gene was found to have a high expression level in the gastric cancer cell line. Additionally, lower survival rates were observed for patients with high ALK expression in TCGA-STAD analysis. In summary, we hypothesize that ALK gene overexpression can be a promising biomarker for prognosis and therapeutic management of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Felipe Pantoja Mesquita
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Pedro Filho Noronha Souza
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Emerson Lucena da Silva
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Luina Benevides Lima
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Lais Lacerda Brasil de Oliveira
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Caroline Aquino Moreira-Nunes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
- Correspondence: (C.A.M.-N.); (R.C.M.)
| | - William J. Zuercher
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rommel Mario Rodríguez Burbano
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém 66073-005, PA, Brazil
- Molecular Biology Laboratory, Ophir Loyola Hospital, Belém 66073-005, PA, Brazil
| | - Maria Elisabete Amaral de Moraes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Raquel Carvalho Montenegro
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
- Correspondence: (C.A.M.-N.); (R.C.M.)
| |
Collapse
|
39
|
Nian R, Jiang H, Zhao J, Hou W, Zhang H, Ma J, Lv P, Jiang L, Wang Y, Xu Y, Wu S, Lou J, Li W. Differences in actionable genomic alterations between brain metastases and non‑brain metastases in patients with non‑small cell lung cancer. Int J Oncol 2022; 61:100. [PMID: 35796015 PMCID: PMC9291252 DOI: 10.3892/ijo.2022.5390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/15/2022] [Indexed: 11/06/2022] Open
Abstract
Brain metastases (BM) have been closely associated with increased morbidity and poor survival outcomes in patients with non‑small cell lung cancer (NSCLC). Excluding risk factors in histological subtypes, genomic alterations, including epidermal growth factor receptor mutations and anaplastic lymphoma kinase (ALK) rearrangements have been also regarded as greater risk factors for BM in the aspect of molecular subtypes. In the present study, 69 tumor tissues and 51 peripheral blood samples from patients with NSCLC were analyzed using a hybridization capture‑based next‑generation sequencing (NGS) panel, including 95 known cancer genes. Among the 90 patients with stage IV NSCLC, 26 cases suffered from BM and 64 cases did not. In total, 174 somatic mutations in 35 mutated genes were identified, and 12 of these genes were concurrently present in the BM group and the non‑BM group. Importantly, five mutated genes including ALK, cytidine deaminase (CDA), SMAD family member 4 (SMAD4), superoxide dismutase 2 (SOD2) and Von Hippel‑Lindau tumor suppressor (VHL) genes were uniquely detected in the BM group, and they were enriched in the Hippo signaling pathway, pyrimidine metabolism and pantothenate and co‑enzyme A (CoA) biosynthesis, as demonstrated using Kyoto Encyclopedia of Genes and Genomes enrichment analysis. RNA polymerase II transcription regulator complex and promyelocytic leukemia nuclear body were the top functional categories according to the Gene Ontology enrichment analysis in the BM group and non‑BM group, respectively. Furthermore, 43.33% (13/30) of mutated genes were detected by both tumor tissue deoxyribonucleic acid (DNA) and plasma‑derived circulating tumor DNA (ctDNA) in the non‑BM group, while this percentage was only limited to 29.41% (5/17) in the BM group. To summarize, significant differences in somatic mutations, somatic interactions, key signaling pathways, functional biological information, and clinical actionability for the therapy of targeted agents were founded between the BM group and the non‑BM group, and ctDNA analysis may by applied as a more credible alternative for genomic profiling in patients with advanced NSCLC without BM, due to its higher consistency for genomic profiling between ctDNA analysis and tissue DNA analysis.
Collapse
Affiliation(s)
- Rui Nian
- Department of Pathology, Affiliated 3201 Hospital of Xi'an Jiaotong University, Shaanxi, Hanzhong 723000, P.R. China
| | - Huihui Jiang
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 200135, P.R. China
| | - Jiangman Zhao
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 200135, P.R. China
| | - Wanle Hou
- Department of Laboratory Medicine, First Hospital of Laohekou City, Hubei, Xiangyang 441800, P.R. China
| | - Hua Zhang
- Department of Pathology, Hanzhong People's Hospital, Shaanxi, Hanzhong 723000, P.R. China
| | - Jiangtao Ma
- Medical Laboratory Science, Hanzhong Railway Central Hospital, Shaanxi, Hanzhong 723000, P.R. China
| | - Pengbiao Lv
- Department of Surgical Oncology, Hanzhong People's Hospital, Shaanxi, Hanzhong 723000, P.R. China
| | - Lisha Jiang
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 200135, P.R. China
| | - Yongpan Wang
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 200135, P.R. China
| | - Yue Xu
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 200135, P.R. China
| | - Shouxin Wu
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 200135, P.R. China
| | - Jingwei Lou
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai 200135, P.R. China
- Dr Jingwei Lou, Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., 180 Zhangheng Road, Shanghai 200135, P.R. China, E-mail:
| | - Wanjun Li
- Department of Pathology, Affiliated 3201 Hospital of Xi'an Jiaotong University, Shaanxi, Hanzhong 723000, P.R. China
- Correspondence to: Professor Wanjun Li, Department of Pathology, Affiliated 3201 Hospital of Xi'an Jiaotong University, 783 Tianhan Avenue, Shaanxi, Hanzhong 723000, P.R. China, E-mail:
| |
Collapse
|
40
|
Liu ZH, Zhu BW, Shi M, Qu YR, He XJ, Yuan HL, Ma J, Li W, Zhao DD, Liu ZC, Wang BM, Wang CY, Tao HQ, Ma TH. Profiling of gene fusion involving targetable genes in Chinese gastric cancer. World J Gastrointest Oncol 2022; 14:1528-1539. [PMID: 36160735 PMCID: PMC9412921 DOI: 10.4251/wjgo.v14.i8.1528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/14/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Approximately half of all new cases of gastric cancer (GC) and related deaths occur in China. More than 80% of patients with GC are diagnosed at an advanced stage, which results in poor prognosis. Although HER2-directed therapy and immune checkpoint inhibitors have been somewhat successful, new drugs are still needed for the treatment of GC. Notably, several gene fusion-targeted drugs have been approved by the United States Food and Drug Administration for solid tumors, including GC, such as larotrectinib for NTRK fusion-positive cancers and zenocutuzumab for NRG1 fusion-positive cancers. However, gene fusions involving targetable genes have not been well characterized in Chinese patients with GC.
AIM To identify the profile of fusions involving targetable genes in Chinese patients with GC using clinical specimens and determine the distribution of patients with gene fusion variants among the molecular subtypes of GC.
METHODS We retrospectively analyzed gene fusion events in tumor tissue samples from 954 Chinese patients with GC. Clinicopathological characteristics were obtained from their medical records. Genetic alterations, such as single nucleotide variants, indels, amplifications, and gene fusions, were identified using a targeted sequencing panel containing 825 genes. Fusions were validated by fluorescence in situ hybridization (FISH) using break-apart probes. The microsatellite instability (MSI) status was evaluated using MSIsensor from the targeted sequencing panel data. Tumor mutational burden (TMB) was calculated using the total number of nonsynonymous mutations divided by the total genomic targeted region. Chi-square analysis was used to determine the enrichment of gene fusions associated with the molecular subtypes of GC.
RESULTS We found that 1.68% (16/954) of patients harbored 20 fusion events involving targetable genes. RARA fusions (n = 5) were the most common, followed by FGFR2, BRAF, MET, FGFR3, RET, ALK, EGFR, NTRK2, and NRG1 fusions. Two of the RARA fusions, EML4-ALK (E6:E20) and EGFR-SEPTIN14 (E7:E10), have been identified in other tumors but not in GC. Surprisingly, 18 gene fusion events were previously not reported in any cancer types. Twelve of the eighteen novel gene fusions included complete exons encoding functional domains of targetable genes, such as the tyrosine kinase domain of receptor tyrosine kinases and the DNA- and ligand-binding domains of RARA. Consistent with the results of detection using the targeted sequencing fusion panel, the results of FISH (fluorescence in situ hybridization) confirmed the rearrangement of FGFR2 and BRAF in tumors from patients 04 and 09, respectively. Genetic analysis indicated that the fusion genes were significantly enriched in patients with ERBB2 amplification (P = 0.02); however, there were no significant differences between fusion-positive and fusion-negative patients in age, sex, MSI status, and TMB.
CONCLUSION We characterized the landscape of fusions involving targetable genes in a Chinese GC cohort and found that 1.68% of patients with GC harbor potential targetable gene fusions, which were enriched in patients with ERBB2 amplification. Gene fusion detection may provide a potential treatment strategy for patients with GC with disease progression following standard therapy.
Collapse
Affiliation(s)
- Zhen-Hua Liu
- Department of Medical Oncology, Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Bo-Wen Zhu
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Min Shi
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Yu-Rong Qu
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Xun-Jun He
- Department of Genetics and Genomic Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Hong-Ling Yuan
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Jie Ma
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Wei Li
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Dan-Dan Zhao
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Zheng-Chuang Liu
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Department of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Bao-Ming Wang
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Chun-Yang Wang
- Medical Center, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| | - Hou-Quan Tao
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
- Department of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Tong-Hui Ma
- Department of Translational Medicine, Genetron Health (Beijing) Technology, Co. Ltd., Beijing 102200, China
| |
Collapse
|
41
|
Jawarkar RD, Sharma P, Jain N, Gandhi A, Mukerjee N, Al-Mutairi AA, Zaki MEA, Al-Hussain SA, Samad A, Masand VH, Ghosh A, Bakal RL. QSAR, Molecular Docking, MD Simulation and MMGBSA Calculations Approaches to Recognize Concealed Pharmacophoric Features Requisite for the Optimization of ALK Tyrosine Kinase Inhibitors as Anticancer Leads. Molecules 2022; 27:molecules27154951. [PMID: 35956900 PMCID: PMC9370430 DOI: 10.3390/molecules27154951] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 12/04/2022] Open
Abstract
ALK tyrosine kinase ALK TK is an important target in the development of anticancer drugs. In the present work, we have performed a QSAR analysis on a dataset of 224 molecules in order to quickly predict anticancer activity on query compounds. Double cross validation assigns an upward plunge to the genetic algorithm−multi linear regression (GA-MLR) based on robust univariate and multivariate QSAR models with high statistical performance reflected in various parameters like, fitting parameters; R2 = 0.69−0.87, F = 403.46−292.11, etc., internal validation parameters; Q2LOO = 0.69−0.86, Q2LMO = 0.69−0.86, CCCcv = 0.82−0.93, etc., or external validation parameters Q2F1 = 0.64−0.82, Q2F2 = 0.63−0.82, Q2F3 = 0.65−0.81, R2ext = 0.65−0.83 including RMSEtr < RMSEcv. The present QSAR evaluation successfully identified certain distinct structural features responsible for ALK TK inhibitory potency, such as planar Nitrogen within four bonds from the Nitrogen atom, Fluorine atom within five bonds beside the non-ring Oxygen atom, lipophilic atoms within two bonds from the ring Carbon atoms. Molecular docking, MD simulation, and MMGBSA computation results are in consensus with and complementary to the QSAR evaluations. As a result, the current study assists medicinal chemists in prioritizing compounds for experimental detection of anticancer activity, as well as their optimization towards more potent ALK tyrosine kinase inhibitor.
Collapse
Affiliation(s)
- Rahul D. Jawarkar
- Faculty of Pharmacy, Oriental University, Indore 453555, Madhya Pradesh, India; (P.S.); (N.J.)
- Correspondence: (R.D.J.); (M.E.A.Z.); Tel.: +91-7385178762 (R.D.J.)
| | - Praveen Sharma
- Faculty of Pharmacy, Oriental University, Indore 453555, Madhya Pradesh, India; (P.S.); (N.J.)
| | - Neetesh Jain
- Faculty of Pharmacy, Oriental University, Indore 453555, Madhya Pradesh, India; (P.S.); (N.J.)
| | - Ajaykumar Gandhi
- Department of Chemistry, Government College of Arts and Science, Aurangabad 431004, Maharashtra, India;
| | - Nobendu Mukerjee
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Kolkata 700118, West Bengal, India;
| | - Aamal A. Al-Mutairi
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 13318, Saudi Arabia; (A.A.A.-M.); (S.A.A.-H.)
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 13318, Saudi Arabia; (A.A.A.-M.); (S.A.A.-H.)
- Correspondence: (R.D.J.); (M.E.A.Z.); Tel.: +91-7385178762 (R.D.J.)
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 13318, Saudi Arabia; (A.A.A.-M.); (S.A.A.-H.)
| | - Abdul Samad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tishk International University, Erbil 44001, Kurdistan Region, Iraq;
| | - Vijay H. Masand
- Department of Chemistry, Vidyabharati Mahavidyalalya, Camp Road, Amravati 444602, Maharashtra, India;
| | - Arabinda Ghosh
- Microbiology Division, Department of Botany, Gauhati University, Guwahati 781014, Assam, India;
| | - Ravindra L. Bakal
- Department of Medicinal Chemistry, Dr. Rajendra Gode Institute of Pharmacy, University-Mardi Road, Amravati 444603, Maharashtra, India;
| |
Collapse
|
42
|
Jenkins KJ, Fineman JR. Progress in Pulmonary Vein Stenosis: Lessons from Success in Treating Pulmonary Arterial Hypertension. CHILDREN 2022; 9:children9060799. [PMID: 35740736 PMCID: PMC9222029 DOI: 10.3390/children9060799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022]
Abstract
Pulmonary vein stenosis (PVS) is a rare and poorly understood condition that can be classified as primary, acquired, status-post surgical repair of PVS, and/or associated with developmental lung disease. Immunohistochemical studies demonstrate that obstruction of the large (extrapulmonary) pulmonary veins is associated with the neointimal proliferation of myofibroblasts. This rare disorder is likely multifactorial with a spectrum of pathobiology. Treatments have been historically surgical, with an increasing repetitive interventional approach. Understanding the biology of these disorders is in its infancy; thus, medical management has lagged behind. Throughout medical history, an increased understanding of the underlying biology of a disorder has led to significant improvements in care and outcomes. One example is the treatment of pulmonary arterial hypertension (PAH). PAH shares several common themes with PVS. These include the spectrum of disease and biological alterations, such as vascular remodeling and vasoconstriction. Over the past two decades, an exponential increase in the understanding of the pathobiology of PAH has led to a dramatic increase in medical therapies that have changed the landscape of the disease. We believe that a similar approach to PVS can generate novel medical therapeutic targets that will markedly improve the outcome of these vulnerable patients.
Collapse
Affiliation(s)
- Kathy J. Jenkins
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA;
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA
- Correspondence:
| |
Collapse
|
43
|
Lee CJ, Modave E, Boeckx B, Kasper B, Aamdal S, Leahy MG, Rutkowski P, Bauer S, Debiec-Rychter M, Sciot R, Lambrechts D, Wozniak A, Schöffski P. Correlation of Immunological and Molecular Profiles with Response to Crizotinib in Alveolar Soft Part Sarcoma: An Exploratory Study Related to the EORTC 90101 "CREATE" Trial. Int J Mol Sci 2022; 23:ijms23105689. [PMID: 35628499 PMCID: PMC9145625 DOI: 10.3390/ijms23105689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023] Open
Abstract
Alveolar soft part sarcoma (ASPS) is a rare subtype of soft tissue sarcoma characterized by an unbalanced translocation, resulting in ASPSCR1-TFE3 fusion that transcriptionally upregulates MET expression. The European Organization for Research and Treatment of Cancer (EORTC) 90101 “CREATE” phase II trial evaluated the MET inhibitor crizotinib in ASPS patients, achieving only limited antitumor activity. We performed a comprehensive molecular analysis of ASPS tissue samples collected in this trial to identify potential biomarkers correlating with treatment outcome. A tissue microarray containing 47 ASPS cases was used for the characterization of the tumor microenvironment using multiplex immunofluorescence. DNA isolated from 34 available tumor samples was analyzed to detect recurrent gene copy number alterations (CNAs) and mutations by low-coverage whole-genome sequencing and whole-exome sequencing. Pathway enrichment analysis was used to identify diseased-associated pathways in ASPS sarcomagenesis. Kaplan–Meier estimates, Cox regression, and the Fisher’s exact test were used to correlate histopathological and molecular findings with clinical data related to crizotinib treatment, aiming to identify potential factors associated with patient outcome. Tumor microenvironment characterization showed the presence of PD-L1 and CTLA-4 in 10 and 2 tumors, respectively, and the absence of PD-1 in all specimens. Apart from CD68, other immunological markers were rarely expressed, suggesting a low level of tumor-infiltrating lymphocytes in ASPS. By CNA analysis, we detected a number of broad and focal alterations. The most common alteration was the loss of chromosomal region 1p36.32 in 44% of cases. The loss of chromosomal regions 1p36.32, 1p33, 1p22.2, and 8p was associated with shorter progression-free survival. Using whole-exome sequencing, 13 cancer-associated genes were found to be mutated in at least three cases. Pathway enrichment analysis identified genetic alterations in NOTCH signaling, chromatin organization, and SUMOylation pathways. NOTCH4 intracellular domain dysregulation was associated with poor outcome, while inactivation of the beta-catenin/TCF complex correlated with improved outcome in patients receiving crizotinib. ASPS is characterized by molecular heterogeneity. We identify genetic aberrations potentially predictive of treatment outcome during crizotinib therapy and provide additional insights into the biology of ASPS, paving the way to improve treatment approaches for this extremely rare malignancy.
Collapse
Affiliation(s)
- Che-Jui Lee
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; (C.-J.L.); (A.W.)
| | - Elodie Modave
- VIB Center for Cancer Biology, VIB and Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (E.M.); (B.B.); (D.L.)
| | - Bram Boeckx
- VIB Center for Cancer Biology, VIB and Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (E.M.); (B.B.); (D.L.)
| | - Bernd Kasper
- Sarcoma Unit, Interdisciplinary Tumor Center, Mannheim University Medical Center, 68167 Mannheim, Germany;
| | - Steinar Aamdal
- Department of Oncology, Oslo University Hospital, 0315 Oslo, Norway;
| | | | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 00-001 Warsaw, Poland;
| | - Sebastian Bauer
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany;
| | - Maria Debiec-Rychter
- Department of Human Genetics, University Hospitals Leuven, KU Leuven, 3000 Leuven, Belgium;
| | - Raf Sciot
- Department of Pathology, University Hospitals Leuven, KU Leuven, 3000 Leuven, Belgium;
| | - Diether Lambrechts
- VIB Center for Cancer Biology, VIB and Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (E.M.); (B.B.); (D.L.)
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; (C.-J.L.); (A.W.)
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; (C.-J.L.); (A.W.)
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, KU Leuven, 3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-1634-1019
| |
Collapse
|
44
|
Activity of ALK Inhibitors in Renal Cancer with ALK Alterations: A Systematic Review. Int J Mol Sci 2022; 23:ijms23073995. [PMID: 35409355 PMCID: PMC8999731 DOI: 10.3390/ijms23073995] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) associated with anaplastic lymphoma kinase (ALK) gene rearrangements (ALK-RCC) is currently considered an “emerging or provisional” tumor entity by the last World Health Organization classification published in 2016. Although several studies assessing ALK-RCC’s clinical and histological characteristics have been published in recent years, only a few publications have evaluated the activity of ALK inhibitors (ALK-i) in this subgroup of patients. Considering the well-recognized efficacy of this evolving class of targeted therapies in other ALK-positive tumors, we conducted a systematic review to evaluate the reported activity of ALK-i in the ALK-RCC subtype. MEDLINE was searched from its inception to 7 January 2022 for case reports and case series on adult metastatic ALK-RCC patients treated with ALK-i whose therapeutic outcomes were available. A virtual cohort of ALK-RCC patients was created. Our results showed a favorable activity of first- and second-generation ALK-i in pretreated ALK-RCC patients in terms of either radiological response or performance status improvement. We hope that the present work will prompt the creation of large, multi-institutional clinical trials to confirm these promising early data.
Collapse
|
45
|
Grela-Wojewoda A, Pacholczak-Madej R, Adamczyk A, Korman M, Püsküllüoğlu M. Cardiotoxicity Induced by Protein Kinase Inhibitors in Patients with Cancer. Int J Mol Sci 2022; 23:ijms23052815. [PMID: 35269958 PMCID: PMC8910876 DOI: 10.3390/ijms23052815] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Kinase inhibitors (KIs) represent a growing class of drugs directed at various protein kinases and used in the treatment of both solid tumors and hematologic malignancies. It is a heterogeneous group of compounds that are widely applied not only in different types of tumors but also in tumors that are positive for a specific predictive factor. This review summarizes common cardiotoxic effects of KIs, including hypertension, arrhythmias with bradycardia and QTc prolongation, and cardiomyopathy that can lead to heart failure, as well as less common effects such as fluid retention, ischemic heart disease, and elevated risk of thromboembolic events. The guidelines for cardiac monitoring and management of the most common cardiotoxic effects of protein KIs are discussed. Potential signaling pathways affected by KIs and likely contributing to cardiac damage are also described. Finally, the need for further research into the molecular mechanisms underlying the cardiovascular toxicity of these drugs is indicated.
Collapse
Affiliation(s)
- Aleksandra Grela-Wojewoda
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Correspondence: ; Tel.: +48-1263-48350
| | - Renata Pacholczak-Madej
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Department of Anatomy, Jagiellonian University Medical College, 31-008 Kraków, Poland
| | - Agnieszka Adamczyk
- Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland;
| | - Michał Korman
- Faculty of Medicine, Jagiellonian University Medical College, 31-008 Kraków, Poland;
| | - Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
| |
Collapse
|
46
|
Popescu VB, Kanhaiya K, Năstac DI, Czeizler E, Petre I. Network controllability solutions for computational drug repurposing using genetic algorithms. Sci Rep 2022; 12:1437. [PMID: 35082323 PMCID: PMC8791995 DOI: 10.1038/s41598-022-05335-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/29/2021] [Indexed: 12/22/2022] Open
Abstract
Control theory has seen recently impactful applications in network science, especially in connections with applications in network medicine. A key topic of research is that of finding minimal external interventions that offer control over the dynamics of a given network, a problem known as network controllability. We propose in this article a new solution for this problem based on genetic algorithms. We tailor our solution for applications in computational drug repurposing, seeking to maximize its use of FDA-approved drug targets in a given disease-specific protein-protein interaction network. We demonstrate our algorithm on several cancer networks and on several random networks with their edges distributed according to the Erdős-Rényi, the Scale-Free, and the Small World properties. Overall, we show that our new algorithm is more efficient in identifying relevant drug targets in a disease network, advancing the computational solutions needed for new therapeutic and drug repurposing approaches.
Collapse
Affiliation(s)
| | | | - Dumitru Iulian Năstac
- POLITEHNICA University of Bucharest, Faculty of Electronics, Telecommunications and Information Technology, 061071, Bucharest, Romania
| | - Eugen Czeizler
- Computer Science, Åbo Akademi University, 20500, Turku, Finland
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania
| | - Ion Petre
- Department of Mathematics and Statistics, University of Turku, 20014, Turku, Finland.
- National Institute for Research and Development in Biological Sciences, 060031, Bucharest, Romania.
| |
Collapse
|
47
|
Lu Y, Fan Z, Zhu S, Huang X, Zhuang Z, Li Y, Deng Z, Gao L, Hong X, Zhang T, Li L, Sun X, Huang W, Zhang J, Liu Y, Zhang B, Jiang J, Gui F, Wang Z, Li Q, Song S, Huang X, Wu Q, Chen L, Zhou D, Zhang J, Yun C, Chen L, Deng X. A new ALK inhibitor overcomes resistance to first- and second-generation inhibitors in NSCLC. EMBO Mol Med 2022; 14:e14296. [PMID: 34845836 PMCID: PMC8749467 DOI: 10.15252/emmm.202114296] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 11/15/2022] Open
Abstract
More than 60% of nonsmall cell lung cancer (NSCLC) patients show a positive response to the first ALK inhibitor, crizotinib, which has been used as the standard treatment for newly diagnosed patients with ALK rearrangement. However, most patients inevitably develop crizotinib resistance due to acquired secondary mutations in the ALK kinase domain, such as the gatekeeper mutation L1196M and the most refractory mutation, G1202R. Here, we develop XMU-MP-5 as a new-generation ALK inhibitor to overcome crizotinib resistance mutations, including L1196M and G1202R. XMU-MP-5 blocks ALK signaling pathways and inhibits the proliferation of cells harboring either wild-type or mutant EML4-ALK in vitro and suppresses tumor growth in xenograft mouse models in vivo. Structural analysis provides insights into the mode of action of XMU-MP-5. In addition, XMU-MP-5 induces significant regression of lung tumors in two genetically engineered mouse (GEM) models, further demonstrating its pharmacological efficacy and potential for clinical application. These preclinical data support XMU-MP-5 as a novel selective ALK inhibitor with high potency and selectivity. XMU-MP-5 holds great promise as a new therapeutic against clinically relevant secondary ALK mutations.
Collapse
Affiliation(s)
- Yue Lu
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Zhenzhen Fan
- Institute of Life and Health EngineeringJinan UniversityGuangzhouChina
| | - Su‐Jie Zhu
- Department of Biochemistry and BiophysicsInstitute of Systems BiomedicinePeking University Health Science CenterBeijingChina
- Beijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking University Health Science CenterBeijingChina
- Institute for Translational MedicineCollege of MedicineQingdao UniversityQingdaoChina
| | - Xiaoxing Huang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
- Present address:
Key Laboratory of Gastrointestinal Cancer (Fujian Medical University)Ministry of EducationFuzhouChina
| | - Zhongji Zhuang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Yunzhan Li
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Zhou Deng
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Lei Gao
- Institute of Life and Health EngineeringJinan UniversityGuangzhouChina
| | - Xuehui Hong
- Department of Gastrointestinal SurgeryAffiliated Zhongshan Hospital of Xiamen UniversityXiamenChina
| | - Ting Zhang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Li Li
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Xihuan Sun
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Wei Huang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Jingfang Zhang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Yan Liu
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Baoding Zhang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Jie Jiang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Fu Gui
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Zheng Wang
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Qiyuan Li
- National Institute for Data Science in Health and MedicineSchool of MedicineXiamen UniversityXiamenChina
| | - Siyang Song
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Xin Huang
- Division of Drug DiscoveryHongyun Biotech Co., Ltd.NanjingChina
| | - Qiao Wu
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Lanfen Chen
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Dawang Zhou
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| | - Jianming Zhang
- National Research Center for Translational MedicineRuijin HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Cai‐Hong Yun
- Department of Biochemistry and BiophysicsInstitute of Systems BiomedicinePeking University Health Science CenterBeijingChina
- Beijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Liang Chen
- Institute of Life and Health EngineeringJinan UniversityGuangzhouChina
| | - Xianming Deng
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamenChina
| |
Collapse
|
48
|
Sebai M, Tulasne D, Caputo SM, Verkarre V, Fernandes M, Reinhart F, Adams S, Maugard C, Caron O, Guillaud-Bataille M, Berthet P, Bignon YJ, Bressac-de Paillerets B, Burnichon N, Chiesa J, Giraud S, Lejeune S, Limacher JM, de Pauw A, Stoppa-Lyonnet D, Zattara-Cannoni H, Deveaux S, Lidereau R, Richard S, Rouleau E. Novel germline MET pathogenic variants in French patients with papillary renal cell carcinomas type I. Hum Mutat 2021; 43:316-327. [PMID: 34882875 DOI: 10.1002/humu.24313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 11/10/2022]
Abstract
Hereditary papillary renal cell carcinoma (HPRC) is a rare inherited renal cancer syndrome characterized by bilateral and multifocal papillary type 1 renal tumors (PRCC1). Activating germline pathogenic variants of MET gene were identified in HPRC families. We reviewed the medical and molecular records of a large French series of 158 patients screened for MET oncogenic variants. MET pathogenic and likely pathogenic variants rate was 12.4% with 40.6% among patients with familial PRCC1 and 5% among patients with sporadic PRCC1. The phenotype in cases with MET pathogenic and likely pathogenic variants was characteristic: PRCC1 tumors were mainly bilateral (84.3%) and multifocal (87.5%). Histologically, six out of seven patients with MET pathogenic variant harboured biphasic squamoid alveolar PRCC. Genetic screening identified one novel pathogenic variant MET c.3389T>C, p.(Leu1130Ser) and three novel likely pathogenic variants: MET c.3257A>T, p.(His1086Leu); MET c.3305T>C, p.(Ile1102Thr) and MET c.3373T>G, p.(Cys1125Gly). Functional assay confirmed their oncogenic effect as they induced an abnormal focus formation. The genotype-phenotype correlation between MET pathogenic variants and PRCC1 presentation should encourage to widen the screening, especially toward non-familial PRCC1. This precise phenotype also constitutes a strong argument for the classification of novel missense variants within the tyrosine kinase domain when functional assays aren't accessible. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Molka Sebai
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy, 94800, Villejuif, France
| | - David Tulasne
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Sandrine M Caputo
- Department of Genetics, Institut Curie, 75005, Paris, France.,Paris Sciences Lettres Research University, 75005, Paris, France
| | - Virginie Verkarre
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique Hôpitaux de Paris, 75015, Paris, France.,French National Network for Rare Cancers in Adults PREDIR labelled by INCa, AP-HP, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Marie Fernandes
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Fanny Reinhart
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique Hôpitaux de Paris, 75015, Paris, France
| | - Séverine Adams
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy, 94800, Villejuif, France
| | - Christine Maugard
- Department of molecular oncogenetics, Hôpitaux Universitaires de Strasbourg, 67091, Strasbourg, France
| | - Olivier Caron
- Department of Medical Oncogenetics, Gustave Roussy, 94800, Villejuif, France
| | - Marine Guillaud-Bataille
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy, 94800, Villejuif, France
| | - Pascaline Berthet
- French National Network for Rare Cancers in Adults PREDIR labelled by INCa, AP-HP, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France.,Oncogenetics Department, Centre François Baclesse, 14000, Caen, France
| | - Yves-Jean Bignon
- French National Network for Rare Cancers in Adults PREDIR labelled by INCa, AP-HP, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France.,Oncogenetics Department, Centre Jean-Perrin, BP 392, 63011, Clermont-Ferrand, France
| | | | - Nelly Burnichon
- Université de Paris, AP-HP, Hôpital Européen Georges Pompidou, Genetics department, Paris, France
| | - Jean Chiesa
- Department of Cytogenetics, Nimes University Hospital, 30029, Nîmes, France
| | - Sophie Giraud
- French National Network for Rare Cancers in Adults PREDIR labelled by INCa, AP-HP, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France.,Genetics Department, Hospices Civils de LYON (HCL), 69002, Lyon, France
| | - Sophie Lejeune
- Department of genetics, CHRU Lille, 59000, Lille, France
| | | | - Antoine de Pauw
- Department of Genetics, Institut Curie, 75005, Paris, France.,Paris Sciences Lettres Research University, 75005, Paris, France
| | - Dominique Stoppa-Lyonnet
- Department of Genetics, Institut Curie, 75005, Paris, France.,INSERM U830, Institut Curie Paris, 75505, Paris, France.,Paris-University, 75006, Paris, France
| | | | - Sophie Deveaux
- French National Network for Rare Cancers in Adults PREDIR labelled by INCa, AP-HP, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | | | - Stéphane Richard
- French National Network for Rare Cancers in Adults PREDIR labelled by INCa, AP-HP, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France.,EPHE, PSL University, UMR 9019 CNRS, Paris-Saclay University, Gustave Roussy, 94800, Villejuif, France
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy, 94800, Villejuif, France
| |
Collapse
|
49
|
Knockdown of microRNA-214-3p Promotes Tumor Growth and Epithelial-Mesenchymal Transition in Prostate Cancer. Cancers (Basel) 2021; 13:cancers13235875. [PMID: 34884984 PMCID: PMC8656576 DOI: 10.3390/cancers13235875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Prostate Cancer is the second leading cause of cancer-related deaths in the United States. In this study, we analyzed a molecule known as a microRNA, which regulates the expression of genes. microRNAs are involved in processes related to cancer onset and progression. Abnormal expression of microRNAs can promote prostate cancer. This study showed that knockdown of microRNA miR-214-3p enhanced the progression and of prostate cancer. In addition, miR-214 regulated the expression of many genes. These results are useful to better understand the function of miR-214-3p in prostate cancer and can be a useful target in the treatment of the disease. Abstract Abnormal expression of microRNA miR-214-3p (miR-214) is associated with multiple cancers. In this study, we assessed the effects of CRISPR/Cas9 mediated miR-214 depletion in prostate cancer (PCa) cells and the underlying mechanisms. Knockdown of miR-214 promoted PCa cell proliferation, invasion, migration, epithelial-mesenchymal transition (EMT), and increased resistance to anoikis, a key feature of PCa cells that undergo metastasis. The reintroduction of miR-214 in miR-214 knockdown cells reversed these effects and significantly suppressed cell proliferation, migration, and invasion. These in vitro studies are consistent with the role of miR-214 as a tumor suppressor. Moreover, miR-214 knockout increased tumor growth in PCa xenografts in nude mice supporting its anti-oncogenic role in PCa. Knockdown of miR-214 increased the expression of its target protein, Protein Tyrosine Kinase 6 (PTK6), a kinase shown to promote oncogenic signaling and tumorigenesis in PCa. In addition, miR-214 modulated EMT as exhibited by differential regulation of E-Cadherin, N-Cadherin, and Vimentin both in vitro and in vivo. RNA-seq analysis of miR-214 knockdown cells revealed altered gene expression related to PCa tumor growth pathways, including EMT and metastasis. Collectively, our findings reveal that miR-214 is a key regulator of PCa oncogenesis and is a potential novel therapeutic target for the treatment of the disease.
Collapse
|
50
|
Aboualizadeh F, Yao Z, Guan J, Drecun L, Pathmanathan S, Snider J, Umapathy G, Kotlyar M, Jurisica I, Palmer R, Stagljar I. Mapping the Phospho-dependent ALK Interactome to Identify Novel Components in ALK Signaling. J Mol Biol 2021; 433:167283. [PMID: 34606829 DOI: 10.1016/j.jmb.2021.167283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 10/25/2022]
Abstract
Protein-protein interactions (PPIs) play essential roles in Anaplastic Lymphoma Kinase (ALK) signaling. Systematic characterization of ALK interactors helps elucidate novel ALK signaling mechanisms and may aid in the identification of novel therapeutics targeting related diseases. In this study, we used the Mammalian Membrane Two-Hybrid (MaMTH) system to map the phospho-dependent ALK interactome. By screening a library of 86 SH2 domain-containing full length proteins, 30 novel ALK interactors were identified. Many of their interactions are correlated to ALK phosphorylation activity: oncogenic ALK mutations potentiate the interactions and ALK inhibitors attenuate the interactions. Among the novel interactors, NCK2 was further verified in neuroblastoma cells using co-immunoprecipitation. Modulation of ALK activity by addition of inhibitors lead to concomitant changes in the tyrosine phosphorylation status of NCK2 in neuroblastoma cells, strongly supporting the functionality of the ALK/NCK2 interaction. Our study provides a resource list of potential novel ALK signaling components for further study.
Collapse
Affiliation(s)
| | - Zhong Yao
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Jikui Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-40530, Sweden
| | - Luka Drecun
- Donnelly Centre, University of Toronto, Ontario, Canada
| | | | - Jamie Snider
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-40530, Sweden
| | - Max Kotlyar
- Krembil Research Institute, University Health Network, Ontario, Canada
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Ontario, Canada; Department of Computer Science, University of Toronto, Ontario, Canada; Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Ruth Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-40530, Sweden
| | - Igor Stagljar
- Donnelly Centre, University of Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Ontario, Canada; Mediterranean Institute for Life Sciences, Meštrovićevo Šetalište 45, Split, Croatia; School of Medicine, University of Split, Split, Croatia. https://twitter.com/stagljar
| |
Collapse
|