1
|
Taboada RG, Cavalher FP, Rego JF, Riechelmann RP. Tyrosine kinase inhibitors in patients with neuroendocrine neoplasms: a systematic literature review. Ther Adv Med Oncol 2024; 16:17588359241286751. [PMID: 39421680 PMCID: PMC11483706 DOI: 10.1177/17588359241286751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Background Several tyrosine kinase receptors inhibitors (TKIs) have demonstrated antiproliferative effects in well-differentiated neuroendocrine tumors (NETs). We aimed to summarize and appraise the current evidence of the efficacy of TKIs in patients with different types of NETs. Methods We performed a systematic review of clinical trials of TKIs in patients with advanced gastroenteropancreatic or lung NETs (PROSPERO registration number: CRD42024507379). Population characteristics, efficacy, and safety results were summarized by type of NET. Results Twenty-eight studies were eligible, totaling 2284 patients. While sunitinib remains the only Food and Drug Administration-approved TKI in patients with NETs (for patients with pancreatic well-differentiated NETs), recent placebo-controlled randomized trials have demonstrated improved response rates and progression-free survival for patients with progressive and pre-treated well-differentiated pancreatic (cabozantinib or surufatinib) or gastrointestinal (GI) NETs (pazopanib, cabozantinib, or surufatinib). There is limited evidence to support the use of a TKI in patients with lung or grade 3 NETs. The toxicity associated with TKIs follows a class effect, with a significant proportion of patients experiencing fatigue, hypertension, and hand-foot skin reactions. Conclusion TKIs are effective therapies in patients with pancreatic or GI well-differentiated NETs and should be part of the therapeutical sequencing of these patients.
Collapse
Affiliation(s)
- Rodrigo G. Taboada
- Department of Clinical Oncology, A.C.Camargo Cancer Center, Sao Paulo, Brazil
| | - Felicia P. Cavalher
- Department of Clinical Oncology, A.C.Camargo Cancer Center, Sao Paulo, Brazil
| | | | - Rachel P. Riechelmann
- Department of Clinical Oncology, A.C.Camargo Cancer Center, Rua Antônio Prudente 211, São Paulo, SP 01509-010, Brazil
| |
Collapse
|
2
|
Chung HC, Saada-Bouzid E, Longo F, Yanez E, Im SA, Castanon E, Desautels DN, Graham DM, Garcia-Corbacho J, Lopez J, Dutcus C, Okpara CE, Ghori R, Jin F, Groisberg R, Korakis I. Lenvatinib plus pembrolizumab for patients with previously treated, advanced, triple-negative breast cancer: Results from the triple-negative breast cancer cohort of the phase 2 LEAP-005 Study. Cancer 2024; 130:3278-3288. [PMID: 39031824 DOI: 10.1002/cncr.35387] [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: 12/07/2023] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Novel treatments are needed for patients with advanced, triple-negative breast cancer (TNBC) that progresses or recurs after first-line treatment with chemotherapy. The authors report results from the TNBC cohort of the multicohort, open-label, single-arm, phase 2 LEAP-005 study of lenvatinib plus pembrolizumab in patients with advanced solid tumors (ClinicalTrials.gov identifier NCT03797326). METHODS Eligible patients had metastatic or unresectable TNBC with disease progression after one or two lines of therapy. Patients received lenvatinib (20 mg daily) plus pembrolizumab (200 mg every 3 weeks; up to 35 cycles). The primary end points were the objective response rate according to Response Evaluation Criteria in Solid Tumors, version 1.1, and safety (adverse events graded by the National Cancer Institute's Common Terminology Criteria for Adverse Events, version 4.0). Duration of response, progression-free survival, and overall survival were secondary end points. RESULTS Thirty-one patients were enrolled. The objective response rate by investigator assessment was 23% (95% confidence interval [CI], 10%-41%). Overall, the objective response rate by blinded independent central review (BICR) was 32% (95% CI, 17%-51%); and, in patients who had programmed cell death ligand 1 combined positive scores ≥10 (n = 8) and <10 (n = 22), the objective response rate was 50% (95% CI, 16%-84%) and 27% (95% CI, 11%-50%), respectively. The median duration of response by BICR was 12.1 months (range, from 3.0+ to 37.9+ months). The median progression-free survival by BICR was 5.1 months (95% CI, 1.9-11.8 months) and the median overall survival was 11.4 months (95% CI, 4.1-21.7 months). Treatment-related adverse events occurred in 94% of patients (grade 3, 52%; grade 4, 0%). One patient died due to a treatment-related adverse event of subarachnoid hemorrhage. CONCLUSIONS The combination of lenvatinib plus pembrolizumab demonstrated antitumor activity with a manageable safety profile in patients with previously treated, advanced TNBC.
Collapse
Affiliation(s)
- Hyun Cheol Chung
- Department of Medical Oncology, Yonsei Cancer Center, Yonsei Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Esma Saada-Bouzid
- Department of Medical Oncology, Cote d'Azur University, Centre Antoine Lacassagne, Nice, France
| | - Federico Longo
- Medical Oncology Department, Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria, Centro de Investigación Biomédica en Red Cáncer, Alcalá University, Madrid, Spain
| | - Eduardo Yanez
- Oncology-Hematology Unit, University of Frontera, Araucanía, Chile
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Eduardo Castanon
- Department of Oncology, Clínica Universidad de Navarra, Madrid, Spain
| | - Danielle N Desautels
- Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Donna M Graham
- Experimental Cancer Medicine Team, The Christie National Health Service Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | | | - Juanita Lopez
- Phase I Drug Development Unit, The Royal Marsden Hospital and The Institute of Cancer Research, Sutton, UK
| | | | | | - Razi Ghori
- Merck & Co., Inc., Rahway, New Jersey, USA
| | - Fan Jin
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | - Iphigenie Korakis
- Department of Medicine and Clinical Research Unit, Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse (IUCT-Oncopole), Toulouse, France
| |
Collapse
|
3
|
Marotta V, Rocco D, Crocco A, Deiana MG, Martinelli R, Di Gennaro F, Valeriani M, Valvano L, Caleo A, Pezzullo L, Faggiano A, Vitale M, Monti S. Survival Predictors of Radioiodine-refractory Differentiated Thyroid Cancer Treated With Lenvatinib in Real Life. J Clin Endocrinol Metab 2024; 109:2541-2552. [PMID: 38501238 DOI: 10.1210/clinem/dgae181] [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: 12/27/2023] [Revised: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
CONTEXT Lenvatinib is approved for the treatment of radioiodine-refractory differentiated thyroid cancer (RR-DTC). The definition of predictive factors of survival is incomplete. OBJECTIVE To identify pre- and posttreatment survival predictors in a real-life cohort of RR-DTC treated with lenvatinib. DESIGN Multicenter, retrospective, cohort study. SETTING 3 Italian thyroid cancer referral centers. PARTICIPANTS 55 RR-DTC treated with lenvatinib. MAIN OUTCOME MEASURES Progression-free survival (PFS) and overall survival (OS). RESULTS Lenvatinib was the first-line kinase-inhibitor in 96.4% of subjects. Median follow-up was 48 months. Median PFS and OS were 26 [95% confidence interval (CI) 19.06-32.93] and 70 months (95% CI 36-111.99), respectively. Pretreatment setting: Eastern Cooperative Oncology Group (ECOG) performance status was independently related to PFS [P < .001; hazard ratio (HR) 18.82; 95% CI 3.65-97.08: score 0-1 as reference] and OS (P = .001; HR 6.20; 95% CI 2.11-18.20; score 0-1 as reference); radioactive iodine (RAI) avidity was independently related to PFS (P = .047; HR 3.74; 95% CI 1.01-13.76; avid disease as reference). Patients with good ECOG status (0-1) and RAI-avid disease obtained objective response in 100% of cases and achieved a median PFS of 45 months without any death upon a median follow-up of 81 months. Posttreatment setting: the best radiological response independently predicted PFS (P = .001; HR 4.6; 95% CI 1.89-11.18; partial/complete response as reference) and OS (P = .013; HR 2.94; 95% CI 1.25-6.89; partial/complete response as reference). CONCLUSION RR-DTC with good performance status and RAI-avid disease obtains the highest clinical benefit from lenvatinib. After treatment initiation, objective response was the only independent survival predictor.
Collapse
Affiliation(s)
- Vincenzo Marotta
- UOC Clinica Endocrinologica e Diabetologica, AOU San Giovanni di Dio e Ruggi d'Aragona, 84131, Salerno, Italy
| | - Domenico Rocco
- Dipartimento di Medicina, Chirurgia e Odontoiatria, Università di Salerno, 84084, Salerno, Italy
| | - Anna Crocco
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori-Irccs-Fondazione G. Pascale, 80131, Napoli, Italy
| | - Maria Grazia Deiana
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00198, Rome, Italy
| | - Ruggero Martinelli
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00198, Rome, Italy
| | - Francesca Di Gennaro
- Struttura Complessa Medicina Nucleare e Terapia Radiometabolica-UOS Terapia Metabolica Sperimentale, Istituto Nazionale Tumori-Irccs-Fondazione G. Pascale, 80131, Napoli, Italy
| | - Mariafelicia Valeriani
- UOC Chirurgia Generale, AOU San Giovanni di Dio e Ruggi d'Aragona, 84131, Salerno, Italy
| | - Luca Valvano
- UOC Chirurgia Generale, AOU San Giovanni di Dio e Ruggi d'Aragona, 84131, Salerno, Italy
| | - Alessia Caleo
- UOC Anatomia Patologica, AOU San Giovanni di Dio e Ruggi d'Aragona, 84131, Salerno, Italy
| | - Luciano Pezzullo
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori-Irccs-Fondazione G. Pascale, 80131, Napoli, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00198, Rome, Italy
| | - Mario Vitale
- Dipartimento di Medicina, Chirurgia e Odontoiatria, Università di Salerno, 84084, Salerno, Italy
| | - Salvatore Monti
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00198, Rome, Italy
| |
Collapse
|
4
|
Varvarà P, Emanuele Drago S, Esposito E, Campolo M, Mauro N, Calabrese G, Conoci S, Morganti D, Fazio B, Giammona G, Pitarresi G. Biotinylated polyaminoacid-based nanoparticles for the targeted delivery of lenvatinib towards hepatocarcinoma. Int J Pharm 2024; 662:124537. [PMID: 39079592 DOI: 10.1016/j.ijpharm.2024.124537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/03/2024]
Abstract
In this work, we describe the development of targeted polymeric nanoparticles loaded with lenvatinib for the treatment of hepatocellular carcinoma (HCC). A synthetic brush copolymer (PHEA-g-BIB-pButMA-g-PEG-biotin) was synthesized from α-poly(N-2-hydroxyethyl)-D,L-aspartamide (PHEA) by a three-step reaction involving atom transfer radical polymerisation (ATRP) to graft hydrophobic polybutylmethacrylate pendant groups and further conjugation with biotinylated polyethylene glycol via carbonate ester. Subsequently, lenvatinib-loaded nanoparticles were obtained and characterized demonstrating colloidal size, negative zeta potential, biotin exposure on the surface and the ability to release lenvatinib in a sustained manner. Lenvatinib-loaded nanoparticles were tested in vitro on HCC cells to evaluate their anticancer efficacy compared to free drug. Furthermore, the enhanced in vivo efficacy of lenvatinib-loaded nanoparticles on nude mice HCC xenograft models was demonstrated by evaluating tumor burdens, apoptotic markers and histological scores after administration of lenvatinib-nanoparticles via intraperitoneal or oral route. Finally, in vivo biodistribution studies were performed, demonstrating the ability of the prepared drug delivery systems to significantly accumulate in the solid tumor by active targeting, due to the presence of biotin on the nanoparticle surface.
Collapse
Affiliation(s)
- Paola Varvarà
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy; Fondazione Veronesi, Piazza Velasca 5, 20122 Milano, Italy
| | - Salvatore Emanuele Drago
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Emanuela Esposito
- Dipartimento Di Scienze Chimiche, Biologiche, farmaceutiche ed Ambientali (CHIBIOFARAM), via F. Stagno d'Alcontres 31, università degli Studi di Messina, Messina 98165, Italy
| | - Michela Campolo
- Dipartimento Di Scienze Chimiche, Biologiche, farmaceutiche ed Ambientali (CHIBIOFARAM), via F. Stagno d'Alcontres 31, università degli Studi di Messina, Messina 98165, Italy
| | - Nicolò Mauro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Giovanna Calabrese
- Dipartimento Di Scienze Chimiche, Biologiche, farmaceutiche ed Ambientali (CHIBIOFARAM), via F. Stagno d'Alcontres 31, università degli Studi di Messina, Messina 98165, Italy
| | - Sabrina Conoci
- Dipartimento Di Scienze Chimiche, Biologiche, farmaceutiche ed Ambientali (CHIBIOFARAM), via F. Stagno d'Alcontres 31, università degli Studi di Messina, Messina 98165, Italy; Dipartimento di Chimica "Giacomo Ciamician", Alma Mater Studiorum, Università di Bologna, Italy; LAB Sense Beyond Nano-URT Department of Sciences Physics and Technologies of Matter (DSFTM) CNR, Messina 98166, Italy
| | - Dario Morganti
- LAB Sense Beyond Nano-URT Department of Sciences Physics and Technologies of Matter (DSFTM) CNR, Messina 98166, Italy
| | - Barbara Fazio
- LAB Sense Beyond Nano-URT Department of Sciences Physics and Technologies of Matter (DSFTM) CNR, Messina 98166, Italy
| | - Gaetano Giammona
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Giovanna Pitarresi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy.
| |
Collapse
|
5
|
Mahboobnia K, Beveridge DJ, Yeoh GC, Kabir TD, Leedman PJ. MicroRNAs in Hepatocellular Carcinoma Pathogenesis: Insights into Mechanisms and Therapeutic Opportunities. Int J Mol Sci 2024; 25:9393. [PMID: 39273339 PMCID: PMC11395074 DOI: 10.3390/ijms25179393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Hepatocellular carcinoma (HCC) presents a significant global health burden, with alarming statistics revealing its rising incidence and high mortality rates. Despite advances in medical care, HCC treatment remains challenging due to late-stage diagnosis, limited effective therapeutic options, tumor heterogeneity, and drug resistance. MicroRNAs (miRNAs) have attracted substantial attention as key regulators of HCC pathogenesis. These small non-coding RNA molecules play pivotal roles in modulating gene expression, implicated in various cellular processes relevant to cancer development. Understanding the intricate network of miRNA-mediated molecular pathways in HCC is essential for unraveling the complex mechanisms underlying hepatocarcinogenesis and developing novel therapeutic approaches. This manuscript aims to provide a comprehensive review of recent experimental and clinical discoveries regarding the complex role of miRNAs in influencing the key hallmarks of HCC, as well as their promising clinical utility as potential therapeutic targets.
Collapse
Affiliation(s)
- Khadijeh Mahboobnia
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Dianne J Beveridge
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - George C Yeoh
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Tasnuva D Kabir
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Peter J Leedman
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
6
|
Gujarathi R, Franses JW, Pillai A, Liao CY. Targeted therapies in hepatocellular carcinoma: past, present, and future. Front Oncol 2024; 14:1432423. [PMID: 39267840 PMCID: PMC11390354 DOI: 10.3389/fonc.2024.1432423] [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: 05/14/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
Targeted therapies are the mainstay of systemic therapies for patients with advanced, unresectable, or metastatic hepatocellular carcinoma. Several therapeutic targets, such as c-Met, TGF-β, and FGFR, have been evaluated in the past, though results from these clinical studies failed to show clinical benefit. However, these remain important targets for the future with novel targeted agents and strategies. The Wnt/β-catenin signaling pathway, c-Myc oncogene, GPC3, PPT1 are exciting novel targets, among others, currently undergoing evaluation. Through this review, we aim to provide an overview of previously evaluated and potentially novel therapeutic targets and explore their continued relevance in ongoing and future studies for HCC.
Collapse
Affiliation(s)
- Rushabh Gujarathi
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Joseph W Franses
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Anjana Pillai
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Chicago, Chicago, IL, United States
| | - Chih-Yi Liao
- Section of Hematology and Oncology, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
7
|
Chen JH, Lin TH, Chien YC, Chen CY, Lin CT, Kuo WW, Chang WC. Aqueous Extracts of Ocimum gratissimum Sensitize Hepatocellular Carcinoma Cells to Cisplatin through BRCA1 Inhibition. Int J Mol Sci 2024; 25:8424. [PMID: 39125994 PMCID: PMC11313253 DOI: 10.3390/ijms25158424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Ocimum gratissimum (O. gratissimum), a medicinal herb with antifungal and antiviral activities, has been found to prevent liver injury and liver fibrosis and induce apoptosis in hepatocellular carcinoma (HCC) cells. In this study, we evaluated the effect of aqueous extracts of O. gratissimum (OGE) on improving the efficacy of chemotherapeutic drugs in HCC cells. Proteomic identification and functional assays were used to uncover the critical molecules responsible for OGE-induced sensitization mechanisms. The antitumor activity of OGE in combination with a chemotherapeutic drug was evaluated in a mouse orthotopic tumor model, and serum biochemical tests were further utilized to validate liver function. OGE sensitized HCC cells to the chemotherapeutic drug cisplatin. Proteomic analysis and Western blotting validation revealed the sensitization effect of OGE, likely achieved through the inhibition of breast cancer type 1 susceptibility protein (BRCA1). Mechanically, OGE treatment resulted in BRCA1 protein instability and increased proteasomal degradation, thereby synergistically increasing cisplatin-induced DNA damage. Moreover, OGE effectively inhibited cell migration and invasion, modulated epithelial-to-mesenchymal transition (EMT), and impaired stemness properties in HCC cells. The combinatorial use of OGE enhanced the efficacy of cisplatin and potentially restored liver function in a mouse orthotopic tumor model. Our findings may provide an alternate approach to improving chemotherapy efficacy in HCC.
Collapse
Affiliation(s)
- Jing-Huei Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (J.-H.C.); (Y.-C.C.)
| | - Tsai-Hui Lin
- Department of Chinese Medicine, China Medical University Hospital, Taichung 404327, Taiwan;
| | - Yu-Chuan Chien
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan; (J.-H.C.); (Y.-C.C.)
| | - Chung-Yu Chen
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan; (C.-Y.C.); (C.-T.L.)
| | - Chih-Tung Lin
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan; (C.-Y.C.); (C.-T.L.)
| | - Wei-Wen Kuo
- Program for Biotechnology Industry, China Medical University, Taichung 406040, Taiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung 406040, Taiwan
| |
Collapse
|
8
|
Sogo Y, Toyoda E, Nagai T, Takahashi T, Takizawa D, Watanabe M, Sato M. Disease-Modifying Effects of Lenvatinib, a Multiple Receptor Tyrosine Kinase Inhibitor, on Posttraumatic Osteoarthritis of the Knee. Int J Mol Sci 2024; 25:6514. [PMID: 38928219 PMCID: PMC11203559 DOI: 10.3390/ijms25126514] [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: 04/30/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Angiogenesis and vascular endothelial growth factor (VEGF) are involved in osteoarthritis (OA). We previously reported the inhibitory effect of bevacizumab in a rabbit model of OA. In the current study, we investigated the effects of lenvatinib, an angiogenesis inhibitor targeting the VEGF and fibroblast growth factor receptors, on synovitis, osteophyte formation, and cartilage degeneration in a rabbit OA model. Posttraumatic OA was induced by anterior cruciate ligament transection (ACLT) on one knee of each rabbit. Rabbits were placed into four groups according to the following lenvatinib doses: untreated control (n = 12), L0.3: 0.3 mg/kg/day (n = 15), L1.0: 1.0 mg/kg/day (n = 14), and L3.0: 3.0 mg/kg/day (n = 13) groups. We evaluated limb pain using the weight distribution ratio measured with an incapacitance tester, macroscopic osteophyte formation, and femoral condyle synovium and cartilage histology. For cartilage evaluation, the following distal sites of the femur were evaluated separately: femoral-tibial (FT), femoral-patellar (FP), and femoral corner (between FP and FT). The weight distribution ratio at 12 weeks after surgery was higher in the L0.3 and L1.0 groups than in the control group. Osteophyte formation and synovitis scores were significantly lower in the L0.3, L1.0, and L3.0 groups than in the control group. The Osteoarthritis Research Society International scores of the FT, corner, and FP sites in the L0.3 group were lower than in the control group. The cartilage thickness ratio at the FT and corner sites was significantly lower in the L0.3 group than in the control group. Krenn's grading system of cartilage synovitis showed that all lenvatinib-administered groups had significantly lower scores than the control group. MMP3 expression level in cartilage tissue was significantly lower in the L3.0 group compared with the other three groups. ADAMTS5 expression was lower in the L3.0 group compared with the control and L0.3 groups. Oral administration of lenvatinib inhibited synovitis, osteophyte formation, and cartilage degeneration and reduced pain in a rabbit ACLT model. Lenvatinib is an oral VEGF inhibitor that is easier to administer than other VEGF inhibitors and may have potential as a treatment of posttraumatic OA.
Collapse
Affiliation(s)
- Yasuyuki Sogo
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Eriko Toyoda
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Toshihiro Nagai
- Department of Orthopaedic Surgery, Tokai University Hachioji Hospital, 1838 Ishikawa-cho, Hachioji 192-0032, Kanagawa, Japan;
| | - Takumi Takahashi
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Daichi Takizawa
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| | - Masato Sato
- Department of Orthopaedic Surgery, Surgical Science, School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan; (Y.S.); (E.T.); (T.T.); (D.T.); (M.W.)
- Center for Musculoskeletal innovative Research and Advancement (C-MiRA), Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara 259-1193, Kanagawa, Japan
| |
Collapse
|
9
|
Côrte-Real L, Sergi B, Yildirim B, Colucas R, Starosta R, Fontrodona X, Romero I, André V, Acilan C, Correia I. Enhanced selectivity towards melanoma cells with zinc(II)-Schiff bases containing imidazole derivatives. Dalton Trans 2024; 53:9416-9432. [PMID: 38758025 DOI: 10.1039/d4dt00733f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Zinc(II)-complexes with the general formula [Zn(L)2] containing 8-hydroxyquinoline Schiff bases functionalized with 1-(3-aminopropyl)imidazole or 1-(3-aminopropyl)-2-methyl-1H-imidazole on 2-position and their respective ligands (HL1 or HL2) were synthesized and characterized by NMR, UV-Vis, FTIR and CD spectroscopies as well as ESI-MS spectrometry. Single crystals of HL2 and [Zn(L1)2]n were analysed by SC-XRD. [Zn(L1)2]n shows a 1D polymeric chain structure of alternating Zn(II) cations and bridging Schiff base ligands, in contrast to previously reported monomeric structures of analogous complexes. DFT calculations were performed to rationalize the polymeric X-ray structure of Zn(L1)2. Results showed that the ligands can bind as bi- or tridentate to Zn(II) and there is the possibility of a dynamic behavior for the complexes in solution. Both ligands and complexes present limited stability in aqueous media, however, in the presence of bovine serum albumin the complexes are stable. Molecular docking simulations and circular dichroism spectroscopic studies suggest binding to this protein in close proximity to the Trp213 residue. Biological studies on a panel of cancer cells revealed that the Zn(II)-complexes have a lower impact on cell viability than cisplatin, except for triple-negative breast cancer cells in which they were comparable. Notwithstanding, they display much higher selectivity towards cancer cells vs. normal cells, than cisplatin. They induce the generation of ROS and DNA double-strand breaks, primarily through apoptosis as the mode of cell death. Overall, the novel Zn(II)-complexes demonstrate improved induction of apoptosis and higher selectivity, particularly for melanoma cells, compared to previously reported analogues, making them promising candidates for clinical application.
Collapse
Affiliation(s)
- Leonor Côrte-Real
- Centro de Química Estrutural, Institute of Molecular Sciences, and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais 1, 1049-001 Lisboa, Portugal.
| | - Baris Sergi
- Koç University, Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Busra Yildirim
- Koç University, Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Raquel Colucas
- Centro de Química Estrutural, Institute of Molecular Sciences, and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais 1, 1049-001 Lisboa, Portugal.
| | - Radosław Starosta
- Centro de Química Estrutural, Institute of Molecular Sciences, and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais 1, 1049-001 Lisboa, Portugal.
- Faculty of Chemistry, University of Wrocław, ul. F. Joliot-Curie 14, 50-383 Wrocław, Poland
| | - Xavier Fontrodona
- Departament de Química and Serveis Técnicas de Recerca, Universitat de Girona, Spain
| | - Isabel Romero
- Departament de Química and Serveis Técnicas de Recerca, Universitat de Girona, Spain
| | - Vânia André
- Centro de Química Estrutural, Institute of Molecular Sciences, and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais 1, 1049-001 Lisboa, Portugal.
| | - Ceyda Acilan
- Koç University, School of Medicine, Sariyer, Istanbul, Turkey.
- Koç University, Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Isabel Correia
- Centro de Química Estrutural, Institute of Molecular Sciences, and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais 1, 1049-001 Lisboa, Portugal.
| |
Collapse
|
10
|
Sun JM, Adenis A, C Enzinger P, Shah MA, Kato K, Bennouna J, Doi T, Hawk NN, Yu L, Shah S, Bhagia P, Shen L. LEAP-014: first-line lenvatinib + pembrolizumab + chemotherapy in advanced/metastatic esophageal squamous cell carcinoma. Future Oncol 2024:1-13. [PMID: 38695479 DOI: 10.2217/fon-2022-1148] [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: 11/16/2022] [Accepted: 03/26/2024] [Indexed: 06/12/2024] Open
Abstract
Treatment options for patients with advanced or metastatic esophageal squamous cell carcinoma (ESCC) are improving. Current guidelines recommend first-line pembrolizumab plus chemotherapy for patients with unresectable or metastatic ESCC, which has led to improvements in survival outcomes. Antiangiogenic therapy combined with immune checkpoint inhibitors can act synergistically to convert the immunosuppressive tumor microenvironment to an immune supportive microenvironment, thus enhancing antitumor immune responses. In preclinical models, the antiangiogenic agent lenvatinib combined with an anti-PD-1 agent showed synergistic antitumor activity. We describe the design and rationale for the randomized, open-label, phase III LEAP-014 study of lenvatinib in combination with pembrolizumab plus chemotherapy in patients with advanced or metastatic ESCC. Overall survival and progression-free survival are the dual primary end points.Clinical Trial Registration: NCT04949256 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Jong-Mu Sun
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Antoine Adenis
- Institut du Cancer de Montpellier & IRCM, Université Montpellier, Montpellier, France
| | | | - Manish A Shah
- Weill Cornell Medical College, New York-Presbyterian Hospital, New York, NY, USA
| | - Ken Kato
- National Cancer Center Hospital, Tokyo, Japan
| | | | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Li Yu
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | - Lin Shen
- Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
11
|
Doostmohammadi A, Jooya H, Ghorbanian K, Gohari S, Dadashpour M. Potentials and future perspectives of multi-target drugs in cancer treatment: the next generation anti-cancer agents. Cell Commun Signal 2024; 22:228. [PMID: 38622735 PMCID: PMC11020265 DOI: 10.1186/s12964-024-01607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/05/2024] [Indexed: 04/17/2024] Open
Abstract
Cancer is a major public health problem worldwide with more than an estimated 19.3 million new cases in 2020. The occurrence rises dramatically with age, and the overall risk accumulation is combined with the tendency for cellular repair mechanisms to be less effective in older individuals. Conventional cancer treatments, such as radiotherapy, surgery, and chemotherapy, have been used for decades to combat cancer. However, the emergence of novel fields of cancer research has led to the exploration of innovative treatment approaches focused on immunotherapy, epigenetic therapy, targeted therapy, multi-omics, and also multi-target therapy. The hypothesis was based on that drugs designed to act against individual targets cannot usually battle multigenic diseases like cancer. Multi-target therapies, either in combination or sequential order, have been recommended to combat acquired and intrinsic resistance to anti-cancer treatments. Several studies focused on multi-targeting treatments due to their advantages include; overcoming clonal heterogeneity, lower risk of multi-drug resistance (MDR), decreased drug toxicity, and thereby lower side effects. In this study, we'll discuss about multi-target drugs, their benefits in improving cancer treatments, and recent advances in the field of multi-targeted drugs. Also, we will study the research that performed clinical trials using multi-target therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Ali Doostmohammadi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Jooya
- Biochemistry Group, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Kimia Ghorbanian
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Sargol Gohari
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
12
|
Xu Y, Xing Z, Abdalla Ibrahim Suliman R, Liu Z, Tang F. Ferroptosis in liver cancer: a key role of post-translational modifications. Front Immunol 2024; 15:1375589. [PMID: 38650929 PMCID: PMC11033738 DOI: 10.3389/fimmu.2024.1375589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
Ferroptosis is an emerging form of regulated cell death in an oxidative stress- and iron-dependent manner, primarily induced by the over-production of reactive oxygen species (ROS). Manipulation of ferroptosis has been considered a promising therapeutic approach to inhibit liver tumor growth. Nevertheless, the development of resistance to ferroptosis in liver cancer poses a significant challenge in cancer treatment. Post-translational modifications (PTMs) are crucial enzymatic catalytic reactions that covalently regulate protein conformation, stability and cellular activities. Additionally, PTMs play pivotal roles in various biological processes and divergent programmed cell death, including ferroptosis. Importantly, key PTMs regulators involved in ferroptosis have been identified as potential targets for cancer therapy. PTMs function of two proteins, SLC7A11, GPX4 involved in ferroptosis resistance have been extensively investigated in recent years. This review will summarize the roles of PTMs in ferroptosis-related proteins in hepatocellular carcinoma (HCC) treatment.
Collapse
Affiliation(s)
- Ying Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Zhiyao Xing
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | | | - Zichuan Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
| | - Fengyuan Tang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
- Thinking Biomed (Beijing) Co., Ltd, Beijing Economic and Technological Development Zone, Beijing, China
| |
Collapse
|
13
|
Kim HD, Yeh CY, Chang YC, Kim CH. Dawn era for revisited cancer therapy by innate immune system and immune checkpoint inhibitors. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167019. [PMID: 38211726 DOI: 10.1016/j.bbadis.2024.167019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
Immunotherapy is a promising therapeutic strategy for cancer. However, it shows limited efficacy against certain tumor types. The activation of innate immunity can suppress tumors by mitigating inflammatory and malignant behaviors through immune surveillance. The tumor microenvironment, which is composed of immune cells and cancer cells, plays a crucial role in determining the outcomes of immunotherapy. Relying solely on immune checkpoint inhibitors is not an optimal approach. Instead, there is a need to consider the use of a combination of immune checkpoint inhibitors with other modulators of the innate immune system to improve the tumor microenvironment. This can be achieved through methods such as immune cell antigen presentation and recognition. In this review, we delve into the significance of innate immune cells in tumor regression, as well as the role of the interaction of tumor cells with innate immune cells in evading host immune surveillance. These findings pave the way for the next chapter in the field of immunotherapy.
Collapse
Affiliation(s)
- Hee-Do Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do 16419, Republic of Korea
| | - Chia-Ying Yeh
- Department of Biomedicine Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Chan Chang
- Department of Biomedicine Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do 16419, Republic of Korea; Samsung Advanced Institute of Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea.
| |
Collapse
|
14
|
Nguyen CB, Oh E, Bahar P, Vaishampayan UN, Else T, Alva AS. Novel Approaches with HIF-2α Targeted Therapies in Metastatic Renal Cell Carcinoma. Cancers (Basel) 2024; 16:601. [PMID: 38339352 PMCID: PMC10854987 DOI: 10.3390/cancers16030601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Germline inactivation of the Von Hippel-Lindau (VHL) tumor suppressor is the defining hallmark in hereditary VHL disease and VHL-associated renal cell carcinoma (RCC). However, somatic VHL mutations are also observed in patients with sporadic RCC. Loss of function VHL mutations result in constitutive activation of hypoxia-inducible factor-2 alpha (HIF-2α), which leads to increased expression of HIF target genes that promote angiogenesis and tumor growth. As of 2023, belzutifan is currently the only approved HIF-2α inhibitor for both VHL-associated and sporadic metastatic RCC (mRCC). However, there is potential for resistance with HIF-2α inhibitors which warrants novel HIF-2α-targeting strategies. In this review, we discuss the potential resistance mechanisms with belzutifan and current clinical trials evaluating novel combinations of belzutifan with other targeted therapies and immune checkpoint inhibitors which may enhance the efficacy of HIF-2α targeting. Lastly, we also discuss newer generation HIF-2α inhibitors that are currently under early investigation and outline future directions and challenges with HIF-2α inhibitors for mRCC.
Collapse
Affiliation(s)
- Charles B. Nguyen
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eugene Oh
- University of Michigan Medical School, Ann Arbor, MI 48109, USA; (E.O.); (P.B.)
| | - Piroz Bahar
- University of Michigan Medical School, Ann Arbor, MI 48109, USA; (E.O.); (P.B.)
| | - Ulka N. Vaishampayan
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tobias Else
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ajjai S. Alva
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (U.N.V.); (T.E.); (A.S.A.)
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
15
|
Becht R, Kiełbowski K, Wasilewicz MP. New Opportunities in the Systemic Treatment of Hepatocellular Carcinoma-Today and Tomorrow. Int J Mol Sci 2024; 25:1456. [PMID: 38338736 PMCID: PMC10855889 DOI: 10.3390/ijms25031456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Liver cirrhosis, hepatitis B, hepatitis C, and non-alcoholic fatty liver disease represent major risk factors of HCC. Multiple different treatment options are available, depending on the Barcelona Clinic Liver Cancer (BCLC) algorithm. Systemic treatment is reserved for certain patients in stages B and C, who will not benefit from regional treatment methods. In the last fifteen years, the arsenal of available therapeutics has largely expanded, which improved treatment outcomes. Nevertheless, not all patients respond to these agents and novel combinations and drugs are needed. In this review, we aim to summarize the pathway of trials investigating the safety and efficacy of targeted therapeutics and immunotherapies since the introduction of sorafenib. Furthermore, we discuss the current evidence regarding resistance mechanisms and potential novel targets in the treatment of advanced HCC.
Collapse
Affiliation(s)
- Rafał Becht
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland; (R.B.); (K.K.)
| | - Kajetan Kiełbowski
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland; (R.B.); (K.K.)
| | - Michał P. Wasilewicz
- Liver Unit, Department of Gastroenterology, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| |
Collapse
|
16
|
Makker V, Taylor MH, Aghajanian C, Cohn AL, Brose MS, Simone CD, Cao ZA, Suttner L, Loboda A, Cristescu R, Jelinic P, Orlowski R, Dutta L, Matsui J, Dutcus CE, Minoshima Y, Messing MJ. Evaluation of potential biomarkers for lenvatinib plus pembrolizumab among patients with advanced endometrial cancer: results from Study 111/KEYNOTE-146. J Immunother Cancer 2024; 12:e007929. [PMID: 38242717 PMCID: PMC10806562 DOI: 10.1136/jitc-2023-007929] [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] [Accepted: 12/04/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Lenvatinib plus pembrolizumab demonstrated clinically meaningful benefit in patients with previously treated advanced endometrial carcinoma in Study 111/KEYNOTE-146 (NCT02501096). In these exploratory analyses from this study, we evaluated the associations between clinical outcomes and gene expression signature scores and descriptively summarized response in biomarker subpopulations defined by tumor mutational burden (TMB) and DNA variants for individual genes of interest. METHODS Patients with histologically confirmed metastatic endometrial carcinoma received oral lenvatinib 20 mg once daily plus intravenous pembrolizumab 200 mg every 3 weeks for 35 cycles. Archived formalin-fixed paraffin-embedded tissue was obtained from all patients. T-cell-inflamed gene expression profile (TcellinfGEP) and 11 other gene signatures were evaluated by RNA sequencing. TMB, hotspot mutations in PIK3CA (oncogene), and deleterious mutations in PTEN and TP53 (tumor suppressor genes) were evaluated by whole-exome sequencing (WES). RESULTS 93 and 79 patients were included in the RNA-sequencing-evaluable and WES-evaluable populations, respectively. No statistically significant associations were observed between any of the RNA-sequencing signature scores and objective response rate or progression-free survival. Area under the receiver operating characteristic curve values for response ranged from 0.39 to 0.54; all 95% CIs included 0.50. Responses were seen regardless of TMB (≥175 or <175 mutations/exome) and mutation status. There were no correlations between TcellinfGEP and TMB, TcellinfGEP and microvessel density (MVD), or MVD and TMB. CONCLUSIONS This analysis demonstrated efficacy for lenvatinib plus pembrolizumab regardless of biomarker status. Results from this study do not support clinical utility of the evaluated biomarkers. Further investigation of biomarkers for this regimen is warranted. TRIAL REGISTRATION NUMBER NCT02501096.
Collapse
Affiliation(s)
- Vicky Makker
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Matthew H Taylor
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | | | - Allen L Cohn
- Rocky Mountain Cancer Center, Denver, Colorado, USA
| | - Marcia S Brose
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Buttell A, Qiu W. The action and resistance mechanisms of Lenvatinib in liver cancer. Mol Carcinog 2023; 62:1918-1934. [PMID: 37671815 PMCID: PMC10840925 DOI: 10.1002/mc.23625] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/07/2023]
Abstract
Lenvatinib is a tyrosine kinase inhibitor that prevents the formation of new blood vessels namely by inhibiting tyrosine kinase enzymes as the name suggests. Specifically, Lenvatinib acts on vascular endothelial growth factor receptors 1-3 (VEGFR1-3), fibroblast growth factor receptors 1-4 (FGFR1-4), platelet-derived growth factor receptor-alpha (PDGFRα), tyrosine-kinase receptor (KIT), and rearranged during transfection receptor (RET). Inhibition of these receptors works to inhibit tumor proliferation. It is through these inhibition mechanisms that Lenvatinib was tested to be noninferior to Sorafenib. However, resistance to Lenvatinib is common, making the positive effects of Lenvatinib on a patient's survival null after resistance is acquired. Therefore, it is crucial to understand mechanisms related to Lenvatinib resistance. This review aims to piece together various mechanisms involved in Lenvatinib resistance and summarizes the research done so far investigating it.
Collapse
Affiliation(s)
- Anna Buttell
- Departments of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
- Departments of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Wei Qiu
- Departments of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
- Departments of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| |
Collapse
|
18
|
Li J, Gu A, Nong XM, Zhai S, Yue ZY, Li MY, Liu Y. Six-Membered Aromatic Nitrogen Heterocyclic Anti-Tumor Agents: Synthesis and Applications. CHEM REC 2023; 23:e202300293. [PMID: 38010365 DOI: 10.1002/tcr.202300293] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Indexed: 11/29/2023]
Abstract
Cancer stands as a serious malady, posing substantial risks to human well-being and survival. This underscores the paramount necessity to explore and investigate novel antitumor medications. Nitrogen-containing compounds, especially those derived from natural sources, form a highly significant category of antitumor agents. Among these, antitumor agents with six-membered aromatic nitrogen heterocycles have consistently attracted the attention of chemists and pharmacologists. Accordingly, we present a comprehensive summary of synthetic strategies and clinical implications of these compounds in this review. This entails an in-depth analysis of synthesis pathways for pyridine, quinoline, pyrimidine, and quinazoline. Additionally, we explore the historical progression, targets, mechanisms of action, and clinical effectiveness of small molecule inhibitors possessing these structural features.
Collapse
Affiliation(s)
- Jiatong Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ao Gu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Xiao-Mei Nong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Shuyang Zhai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Zhu-Ying Yue
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Meng-Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
19
|
Mortensen ACL, Berglund H, Hariri M, Papalanis E, Malmberg C, Spiegelberg D. Combination therapy of tyrosine kinase inhibitor sorafenib with the HSP90 inhibitor onalespib as a novel treatment regimen for thyroid cancer. Sci Rep 2023; 13:16844. [PMID: 37803074 PMCID: PMC10558458 DOI: 10.1038/s41598-023-43486-z] [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: 07/14/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023] Open
Abstract
Thyroid cancer is the most common endocrine malignancy, affecting nearly 600,000 new patients worldwide. Treatment with the BRAF inhibitor sorafenib partially prolongs progression-free survival in thyroid cancer patients, but fails to improve overall survival. This study examines enhancing sorafenib efficacy by combination therapy with the novel HSP90 inhibitor onalespib. In vitro efficacy of sorafenib and onalespib monotherapy as well as in combination was assessed in papillary (PTC) and anaplastic (ATC) thyroid cancer cells using cell viability and colony formation assays. Migration potential was studied in wound healing assays. The in vivo efficacy of sorafenib and onalespib therapy was evaluated in mice bearing BHT-101 xenografts. Sorafenib in combination with onalespib significantly inhibited PTC and ATC cell proliferation, decreased metabolic activity and cancer cell migration. In addition, the drug combination approach significantly inhibited tumor growth in the xenograft model and prolonged the median survival. Our results suggest that combination therapy with sorafenib and onalespib could be used as a new therapeutic approach in the treatment of thyroid cancer, significantly improving the results obtained with sorafenib as monotherapy. This approach has the potential to reduce treatment adaptation while at the same time providing therapeutic anti-cancer benefits such as reducing tumor growth and metastatic potential.
Collapse
Affiliation(s)
- Anja Charlotte Lundgren Mortensen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Berglund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mehran Hariri
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Eleftherios Papalanis
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Diana Spiegelberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
20
|
Rimel BJ, Crane EK, Hou J, Nakayama J, MacDonald J, Lutz K, Makker V, O'Cearbhaill RE. Tyrosine kinase inhibitor toxicities: A society of gynecologic oncology review and recommendations. Gynecol Oncol 2023; 174:148-156. [PMID: 37207499 DOI: 10.1016/j.ygyno.2023.05.007] [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: 04/05/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/21/2023]
Abstract
OBJECTIVE Oral tyrosine kinase inhibitors (TKIs) have new indications for treatment in gynecologic malignancies. These targeted drugs have both unique and overlapping toxicities, which require careful attention and management. New combination therapies with immune-oncology agents have demonstrated promise in endometrial cancer. This review examines common adverse events associated with TKIs and provides readers with an evidence-based review on current uses and strategies for the management of these medications. METHODS A comprehensive review of the medical literature on TKI use in gynecologic cancer was undertaken by a committee approach. Details of each drug, its molecular target, and relevant data on both clinical efficacy and side effects were compiled and organized for clinical use. Information on drug-related secondary effects and management strategies for specific toxicities, including dose reduction and concomitant medications, were gathered. RESULTS TKIs can potentially offer improved response rates and durable responses for a group of patients who were previously without an effective standard second-line therapy. The combination of lenvatinib and pembrolizumab represents a more targeted approach to the drivers of endometrial cancer; however, there remains significant drug-related toxicity, and thus dose reduction and dose delay are frequently required. Toxicity management requires frequent check-ins and management strategies to help patients find the highest tolerable dose. TKIs are expensive and patient financial toxicity is as critical a measure of a drug's utility as any drug side effect. Many of these drugs have patient assistance programs, which should be fully utilized to minimize cost. CONCLUSIONS Future studies are needed to expand the role of TKIs into new molecularly driven groups. Attention to cost, durability of response, and long-term toxicity management is needed to ensure all eligible patients have access to treatment.
Collapse
Affiliation(s)
- Bobbie J Rimel
- Cedars-Sinai Medical Center, Los Angeles, CA, United States of America.
| | - Erin K Crane
- Levine Cancer Institute, Charlotte, NC, United States of America
| | - June Hou
- Columbia University Irvings Medical Center, New York, NY, United States of America
| | - John Nakayama
- Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Jennifer MacDonald
- Medical University of South Carolina, Charleston, SC, United States of America
| | - Kathleen Lutz
- New York University Langone Health Perlmutter Cancer Center, New York, NY, United States of America
| | - Vicky Makker
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America
| | - Roisin E O'Cearbhaill
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, United States of America
| |
Collapse
|
21
|
Wang L, Yang Q, Zhou Q, Fang F, Lei K, Liu Z, Zheng G, Zhu L, Huo J, Li X, Peng S, Kuang M, Lin S, Huang M, Xu L. METTL3-m 6A-EGFR-axis drives lenvatinib resistance in hepatocellular carcinoma. Cancer Lett 2023; 559:216122. [PMID: 36898427 DOI: 10.1016/j.canlet.2023.216122] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
Lenvatinib is emerging as the first-line therapeutic option for advanced hepatocellular carcinoma (HCC), but drug resistance remains a major hurdle for its long-term therapy efficiency in clinic. N6-methyladenosine (m6A) is the most abundant mRNA modification. Here, we aimed to investigate the modulatory effects and underlying mechanisms of m6A in lenvatinib resistance in HCC. Our data revealed that m6A mRNA modification was significantly upregulated in the HCC lenvatinib resistance (HCC-LR) cells compared to parental cells. Methyltransferase-like 3 (METTL3) was the most significantly upregulated protein among the m6A regulators. Either genetic or pharmacological inhibition of m6A methylation through METTL3 deactivation in primary resistant cell line MHCC97H and acquired resistant Huh7-LR cells decreased cell proliferation and increased cell apoptosis upon lenvatinib treatment in vitro and in vivo. In addition, the specific METTL3 inhibitor STM2457 improved tumor response to lenvatinib in multiple mouse HCC models, including subcutaneous, orthotopic and hydrodynamic models. The MeRIP-seq results showed that epidermal growth factor receptor (EGFR) was a downstream target of METTL3. EGFR overexpression abrogated the METTL3 knocked down-induced cell growth arrest upon lenvatinib treatment in HCC-LR cells. Thus, we concluded that targeting METTL3 using specific inhibitor STM2457 improved the sensitivity to lenvatinib in vitro and in vivo, indicating that METTL3 may be a potential therapeutic target to overcome lenvatinib resistance in HCC.
Collapse
Affiliation(s)
- Lina Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingxia Yang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianying Zhou
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fei Fang
- Clinical Trial Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Lei
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ziqin Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gaomin Zheng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lefan Zhu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jihui Huo
- Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxing Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sui Peng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Clinical Trial Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Kuang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuibin Lin
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Manling Huang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Lixia Xu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
22
|
Candido MF, Medeiros M, Veronez LC, Bastos D, Oliveira KL, Pezuk JA, Valera ET, Brassesco MS. Drugging Hijacked Kinase Pathways in Pediatric Oncology: Opportunities and Current Scenario. Pharmaceutics 2023; 15:pharmaceutics15020664. [PMID: 36839989 PMCID: PMC9966033 DOI: 10.3390/pharmaceutics15020664] [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: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Childhood cancer is considered rare, corresponding to ~3% of all malignant neoplasms in the human population. The World Health Organization (WHO) reports a universal occurrence of more than 15 cases per 100,000 inhabitants around the globe, and despite improvements in diagnosis, treatment and supportive care, one child dies of cancer every 3 min. Consequently, more efficient, selective and affordable therapeutics are still needed in order to improve outcomes and avoid long-term sequelae. Alterations in kinases' functionality is a trademark of cancer and the concept of exploiting them as drug targets has burgeoned in academia and in the pharmaceutical industry of the 21st century. Consequently, an increasing plethora of inhibitors has emerged. In the present study, the expression patterns of a selected group of kinases (including tyrosine receptors, members of the PI3K/AKT/mTOR and MAPK pathways, coordinators of cell cycle progression, and chromosome segregation) and their correlation with clinical outcomes in pediatric solid tumors were accessed through the R2: Genomics Analysis and Visualization Platform and by a thorough search of published literature. To further illustrate the importance of kinase dysregulation in the pathophysiology of pediatric cancer, we analyzed the vulnerability of different cancer cell lines against their inhibition through the Cancer Dependency Map portal, and performed a search for kinase-targeted compounds with approval and clinical applicability through the CanSAR knowledgebase. Finally, we provide a detailed literature review of a considerable set of small molecules that mitigate kinase activity under experimental testing and clinical trials for the treatment of pediatric tumors, while discuss critical challenges that must be overcome before translation into clinical options, including the absence of compounds designed specifically for childhood tumors which often show differential mutational burdens, intrinsic and acquired resistance, lack of selectivity and adverse effects on a growing organism.
Collapse
Affiliation(s)
- Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Mariana Medeiros
- Regional Blood Center, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Luciana Chain Veronez
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - David Bastos
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Karla Laissa Oliveira
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Julia Alejandra Pezuk
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - María Sol Brassesco
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
- Correspondence: ; Tel.: +55-16-3315-9144; Fax: +55-16-3315-4886
| |
Collapse
|
23
|
Peng C, Rabold K, Netea MG, Jaeger M, Netea-Maier RT. Influence of Lenvatinib on the Functional Reprogramming of Peripheral Myeloid Cells in the Context of Non-Medullary Thyroid Carcinoma. Pharmaceutics 2023; 15:pharmaceutics15020412. [PMID: 36839733 PMCID: PMC9960916 DOI: 10.3390/pharmaceutics15020412] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Lenvatinib is a multitarget tyrosine kinase inhibitor (TKI) approved for the treatment of several types of cancers, including metastatic differentiated thyroid cancer (DTC). The intended targets include VEGFR 1-3, FGFR 1-4, PDGFRα, RET, and KIT signaling pathways, but drug resistance inevitably develops and a complete cure is very rare. Recent data has revealed that most of the TKIs have additional 'off-target' immunological effects, which might contribute to a protective antitumor immune response; however, human cellular data are lacking regarding Lenvatinib-mediated immunomodulation in DTC. Here, we investigated in ex vivo models the impact of Lenvatinib on the function of immune cells in healthy volunteers. We found that monocytes and macrophages were particularly susceptible to Lenvatinib, while neutrophiles and lymphocytes were less affected. In tumor-immune cell co-culture experiments, Lenvatinib exerted a broad inhibitory effect on the proinflammatory response in TC-induced macrophages. Interestingly, Lenvatinib-treated cells had decreased cellular M2 membrane markers, whereas they secreted a significantly higher level of the anti-inflammatory cytokine IL-10 upon LPS stimulation. In addition, prolonged exposure to Lenvatinib impaired macrophages survival and phenotypical differentiation, which was accompanied by remarkable morphological changes and suppressed cellular metabolic activity. These effects were mediated by myeloid cell-intrinsic mechanisms which are independent of Lenvatinib's on-target activity. Finally, using specific inhibitors, we argue that dual effects on p38 MAPK and Syk pathways are likely the underlying mechanism of the off-target immunological effects we observed in this study. Collectively, our data show the immunomodulatory properties of Lenvatinib on human monocytes. These insights could be harnessed for the future design of novel treatment strategies involving a combination of Lenvatinib with other immunotherapeutic agents.
Collapse
Affiliation(s)
- Chunying Peng
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Katrin Rabold
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Romana T. Netea-Maier
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Correspondence: ; Tel.: +31-24-3614599
| |
Collapse
|
24
|
Basu D, Pal R, Sarkar M, Barma S, Halder S, Roy H, Nandi S, Samadder A. To Investigate Growth Factor Receptor Targets and Generate Cancer Targeting Inhibitors. Curr Top Med Chem 2023; 23:2877-2972. [PMID: 38164722 DOI: 10.2174/0115680266261150231110053650] [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: 05/26/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024]
Abstract
Receptor tyrosine kinase (RTK) regulates multiple pathways, including Mitogenactivated protein kinases (MAPKs), PI3/AKT, JAK/STAT pathway, etc. which has a significant role in the progression and metastasis of tumor. As RTK activation regulates numerous essential bodily processes, including cell proliferation and division, RTK dysregulation has been identified in many types of cancers. Targeting RTK is a significant challenge in cancer due to the abnormal upregulation and downregulation of RTK receptors subfamily EGFR, FGFR, PDGFR, VEGFR, and HGFR in the progression of cancer, which is governed by multiple RTK receptor signalling pathways and impacts treatment response and disease progression. In this review, an extensive focus has been carried out on the normal and abnormal signalling pathways of EGFR, FGFR, PDGFR, VEGFR, and HGFR and their association with cancer initiation and progression. These are explored as potential therapeutic cancer targets and therefore, the inhibitors were evaluated alone and merged with additional therapies in clinical trials aimed at combating global cancer.
Collapse
Affiliation(s)
- Debroop Basu
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Riya Pal
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, IndiaIndia
| | - Maitrayee Sarkar
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Soubhik Barma
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Sumit Halder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Harekrishna Roy
- Nirmala College of Pharmacy, Vijayawada, Guntur, Andhra Pradesh, India
| | - Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur, 244713, India
| | - Asmita Samadder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| |
Collapse
|
25
|
Overcoming cancer chemotherapy resistance by the induction of ferroptosis. Drug Resist Updat 2023; 66:100916. [PMID: 36610291 DOI: 10.1016/j.drup.2022.100916] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Development of resistance to chemotherapy in cancer continues to be a major challenge in cancer management. Ferroptosis, a unique type of cell death, is mechanistically and morphologically different from other forms of cell death. Ferroptosis plays a pivotal role in inhibiting tumour growth and has presented new opportunities for treatment of chemotherapy-insensitive tumours in recent years. Emerging studies have suggested that ferroptosis can regulate the therapeutic responses of tumours. Accumulating evidence supports ferroptosis as a potential target for chemotherapy resistance. Pharmacological induction of ferroptosis could reverse drug resistance in tumours. In this review article, we first discuss the key principles of chemotherapeutic resistance in cancer. We then provide a brief overview of the core mechanisms of ferroptosis in cancer chemotherapeutic drug resistance. Finally, we summarise the emerging data that supports the fact that chemotherapy resistance in different types of cancers could be subdued by pharmacologically inducing ferroptosis. This review article suggests that pharmacological induction of ferroptosis by bioactive compounds (ferroptosis inducers) could overcome chemotherapeutic drug resistance. This article also highlights some promising therapeutic avenues that could be used to overcome chemotherapeutic drug resistance in cancer.
Collapse
|
26
|
Cao J, Chow L, Dow S. Strategies to overcome myeloid cell induced immune suppression in the tumor microenvironment. Front Oncol 2023; 13:1116016. [PMID: 37114134 PMCID: PMC10126309 DOI: 10.3389/fonc.2023.1116016] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer progression and metastasis due to tumor immune evasion and drug resistance is strongly associated with immune suppressive cellular responses, particularly in the case of metastatic tumors. The myeloid cell component plays a key role within the tumor microenvironment (TME) and disrupts both adaptive and innate immune cell responses leading to loss of tumor control. Therefore, strategies to eliminate or modulate the myeloid cell compartment of the TME are increasingly attractive to non-specifically increase anti-tumoral immunity and enhance existing immunotherapies. This review covers current strategies targeting myeloid suppressor cells in the TME to enhance anti-tumoral immunity, including strategies that target chemokine receptors to deplete selected immune suppressive myeloid cells and relieve the inhibition imposed on the effector arms of adaptive immunity. Remodeling the TME can in turn improve the activity of other immunotherapies such as checkpoint blockade and adoptive T cell therapies in immunologically "cold" tumors. When possible, in this review, we have provided evidence and outcomes from recent or current clinical trials evaluating the effectiveness of the specific strategies used to target myeloid cells in the TME. The review seeks to provide a broad overview of how myeloid cell targeting can become a key foundational approach to an overall strategy for improving tumor responses to immunotherapy.
Collapse
Affiliation(s)
- Jennifer Cao
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lyndah Chow
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Steven Dow
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- *Correspondence: Steven Dow,
| |
Collapse
|
27
|
Saeed RF, Awan UA, Saeed S, Mumtaz S, Akhtar N, Aslam S. Targeted Therapy and Personalized Medicine. Cancer Treat Res 2023; 185:177-205. [PMID: 37306910 DOI: 10.1007/978-3-031-27156-4_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Targeted therapy and personalized medicine are novel emerging disciplines of cancer research intended for treatment and prevention. One of the most significant advancements in modern oncology is the shift from an organ-centric strategy to a personalized strategy guided by deep molecular analysis. This shift in view, which focuses on the tumour's precise molecular changes, has paved the way for individualized treatment. Researchers and clinicians are using targeted therapies to select the best treatment available based on the molecular characterization of malignant cancer. In the treatment of a cancer, personalized medicine entails the use of genetic, immunological, and proteomic profiling to provide therapeutic alternatives as well as prognostic information about cancer. In this book, targeted therapies and personalized medicine have been covered for specific malignancies, including latest FDA-approved targeted therapies and it also sheds light on effective anti-cancer regimens and drug resistance. This will help to enhance our ability to conduct individualized health planning, make early diagnoses, and choose optimal medications for each cancer patient with predictable side effects and outcomes in a quickly evolving era. Various applications and tools' capacity have been improved for early diagnosis of cancer and the growing number of clinical trials that choose specific molecular targets reflects this predicament. Nevertheless, there are several limitations that must need to be addressed. Hence, in this chapter, we will discuss recent advancements, challenges, and opportunities in personalized medicine for various cancers, with a specific emphasis on target therapies in diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rida Fatima Saeed
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
| | - Uzma Azeem Awan
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | | | - Sara Mumtaz
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Nosheen Akhtar
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Shaista Aslam
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| |
Collapse
|
28
|
Motzer RJ, Schmidinger M, Eto M, Suarez C, Figlin R, Liu Y, Perini R, Zhang Y, Heng DY. LITESPARK-011: belzutifan plus lenvatinib vs cabozantinib in advanced renal cell carcinoma after anti-PD-1/PD-L1 therapy. Future Oncol 2023; 19:113-121. [PMID: 36752726 DOI: 10.2217/fon-2022-0802] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
The first-in-class, small molecule HIF-2α inhibitor, belzutifan, has demonstrated promising antitumor activity in previously treated patients with clear cell renal cell carcinoma (RCC). HIF-2α also regulates VEGF expression and is involved in resistance to anti-VEGF therapy. This study describes the rationale and design for a randomized, phase III study evaluating efficacy and safety of belzutifan plus the tyrosine kinase inhibitor (TKI) lenvatinib versus the TKI cabozantinib in patients with advanced RCC progressing after anti-PD-1/PD-L1 therapy in the first- or second-line setting or as adjuvant therapy. Considering the unmet need for effective and tolerable treatment of advanced RCC following immune checkpoint inhibitors, belzutifan plus lenvatinib may have a positive benefit/risk profile. Clinical Trial Registration: NCT04586231 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Robert J Motzer
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Masatoshi Eto
- Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Cristina Suarez
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Barcelona, 08035, Spain
| | - Robert Figlin
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|
29
|
Delcuratolo MD, Tucci M, Turco F, Di Stefano RF, Ungaro A, Audisio M, Samuelly A, Brusa F, Audisio A, Di Maio M, Scagliotti GV, Buttigliero C. Therapeutic sequencing in advanced renal cell carcinoma: How to choose considering clinical and biological factors. Crit Rev Oncol Hematol 2023; 181:103881. [PMID: 36427772 DOI: 10.1016/j.critrevonc.2022.103881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/26/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022] Open
Abstract
In the last fifteen years a better understanding of the biological processes promoting tumour growth and progression led to an impressive revolution in metastatic renal cell carcinoma (mRCC) treatment landscape. Angiogenesis plays a critical role in the pathogenesis of RCC. These biological evidences led to targeted therapies interfering with vascular endothelial growth factor and mammalian target of rapamycin pathway. Another big step in the RCC therapeutic landscape was recently made because of the understanding of the interplay between angiogenesis and immune cells. Dual immune checkpoint inhibitors (ICIs) and ICIs plus tyrosine kinase inhibitors (TKI) combinations have been approved considering overall survival benefit compared to targeted therapies as first line treatment. We summarize the activity and the biological rationale of ICIs combinations as mRCC first line therapy. Additionally, we review the clinical and biological criteria useful to guide clinicians in the choice of treatment sequencing focusing on ICIs combinations resistance mechanisms.
Collapse
Affiliation(s)
- Marco Donatello Delcuratolo
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Marcello Tucci
- Medical Oncology Department, Cardinal Massaia Hospital, Asti 14100, Italy.
| | - Fabio Turco
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Rosario Francesco Di Stefano
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Antonio Ungaro
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Marco Audisio
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Alessandro Samuelly
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Federica Brusa
- Medical Oncology Department, Cardinal Massaia Hospital, Asti 14100, Italy
| | - Alessandro Audisio
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, at Division of Medical Oncology, Ordine Mauriziano Hospital, Via Magellano 1, Turin 10028, Italy
| | - Giorgio Vittorio Scagliotti
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| | - Consuelo Buttigliero
- Department of Oncology, University of Turin, at Division of Medical Oncology, San Luigi Gonzaga Hospital, Regione Gonzole 10, Orbassano, Turin 10043, Italy
| |
Collapse
|
30
|
Wu Z, Bian Y, Chu T, Wang Y, Man S, Song Y, Wang Z. The role of angiogenesis in melanoma: Clinical treatments and future expectations. Front Pharmacol 2022; 13:1028647. [PMID: 36588679 PMCID: PMC9797529 DOI: 10.3389/fphar.2022.1028647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
The incidence of melanoma has increased rapidly over the past few decades, with mortality accounting for more than 75% of all skin cancers. The high metastatic potential of Melanoma is an essential factor in its high mortality. Vascular angiogenic system has been proved to be crucial for the metastasis of melanoma. An in-depth understanding of angiogenesis will be of great benefit to melanoma treatment and may promote the development of melanoma therapies. This review summarizes the recent advances and challenges of anti-angiogenic agents, including monoclonal antibodies, tyrosine kinase inhibitors, human recombinant Endostatin, and traditional Chinese herbal medicine. We hope to provide a better understanding of the mechanisms, clinical research progress, and future research directions of melanoma.
Collapse
Affiliation(s)
- Zhuzhu Wu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yifei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuman Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuai Man
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Yongmei Song
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Zhenguo Wang
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| |
Collapse
|
31
|
Stępniak J, Krawczyk-Lipiec J, Lewiński A, Karbownik-Lewińska M. Sorafenib versus Lenvatinib Causes Stronger Oxidative Damage to Membrane Lipids in Noncancerous Tissues of the Thyroid, Liver, and Kidney: Effective Protection by Melatonin and Indole-3-Propionic Acid. Biomedicines 2022; 10:biomedicines10112890. [PMID: 36428458 PMCID: PMC9687109 DOI: 10.3390/biomedicines10112890] [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: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
Abstract
Sorafenib and lenvatinib are multi-targeted tyrosine kinase inhibitors which are currently approved to treat advanced hepatocellular carcinoma, renal cell carcinoma and radioiodine-refractory differentiated thyroid carcinoma. However this treatment is often limited due to common adverse events which may occur via oxidative stress. The study aims to compare sorafenib- and lenvatinib-induced oxidative damage to membrane lipids (lipid peroxidation, LPO) in homogenates of porcine noncancerous tissues of the thyroid, the liver, and the kidney and to check if it can be prevented by antioxidants melatonin and indole-3-propionic acid (IPA). Homogenates of individual tissues were incubated in the presence of sorafenib or lenvatinib (1 mM, 100 µM, 10 µM, 1 µM, 100 nM, 10 nM, 1 nM, 100 pM) together with/without melatonin (5.0 mM) or IPA (5.0 mM). The concentration of malondialdehyde + 4-hydroxyalkenals, as the LPO index, was measured spectrophotometrically. The incubation of tissue homogenates with sorafenib resulted in a concentration-dependent increase in LPO (statistically significant for concentrations of 1mM and 100 µM in the thyroid and the liver, and of 1 mM, 100 µM, and 10 µM in the kidney). The incubation of thyroid homogenates with lenvatinib did not change LPO level. In case of the liver and the kidney, lenvatinib increased LPO but only in its highest concentration of 1 mM. Melatonin and IPA reduced completely (to the level of control) sorafenib- and lenvatinib-induced LPO in all examined tissues regardless of the drug concentration. In conclusion, sorafenib comparing to lenvatinib is a stronger damaging agent of membrane lipids in noncancerous tissues of the thyroid, the liver, and the kidney. The antioxidants melatonin and IPA can be considered to be used in co-treatment with sorafenib and lenvatinib to prevent their undesirable toxicity occurring via oxidative stress.
Collapse
Affiliation(s)
- Jan Stępniak
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Joanna Krawczyk-Lipiec
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Andrzej Lewiński
- Polish Mother’s Memorial Hospital—Research Institute, 93-338 Lodz, Poland
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
| | - Małgorzata Karbownik-Lewińska
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752 Lodz, Poland
- Polish Mother’s Memorial Hospital—Research Institute, 93-338 Lodz, Poland
- Correspondence:
| |
Collapse
|
32
|
Wang Y, Tan K, Hu W, Hou Y, Yang G. LncRNA AC026401.3 interacts with OCT1 to intensify sorafenib and lenvatinib resistance by activating E2F2 signaling in hepatocellular carcinoma. Exp Cell Res 2022; 420:113335. [PMID: 36084669 DOI: 10.1016/j.yexcr.2022.113335] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 12/24/2022]
Abstract
Multitargeted kinase inhibitors (MKIs) including sorafenib and lenvatinib, are applied for first-line treatment for inoperable hepatocellular carcinoma (HCC) patients, but the therapeutic effect is limited because of drug resistance. Therefore, we sought potential biomarkers to indicate sorafenib and lenvatinib resistance in HCC. In this article, we report a novel long non-coding RNA (lncRNA), AC026401.3, in promoting sorafenib and lenvatinib resistance of HCC cells. AC026401.3 is upregulated in HCC tissues and is positively relevant to HCC patients with large tumor size, cancer recurrence, advanced TNM stage, and poor prognosis. AC026401.3 knockdown or knockout enhances the sensitivity of HCC cells to sorafenib and lenvatinib, respectively. Moreover, AC026401.3 upregulates the expression of the transcription factor E2F2. Mechanistically, AC026401.3 interacts with OCT1 and promotes the recruitment of OCT1 to the promoter region of E2F2, intensifying sorafenib and lenvatinib resistance in HCC by activating the transcription of E2F2. In conclusion, our results reveal that lncRNA AC026401.3 is a risk factor for HCC patients by enhancing sorafenib and lenvatinib resistance of HCC cells, and targeting the AC026401.3-OCT1-E2F2 signaling axis would be a promising strategy for HCC therapeutics.
Collapse
Affiliation(s)
- Yun Wang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710038, China
| | - Kai Tan
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710038, China
| | - Wen Hu
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710038, China
| | - Yan Hou
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710038, China
| | - Guang Yang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, 710038, China.
| |
Collapse
|
33
|
Sambo M. Use of multikinase inhibitors/lenvatinib in patients with synchronous/metachronous cancers coinciding with radioactive-resistant differentiated thyroid cancer. Cancer Med 2022; 11 Suppl 1:26-32. [PMID: 36202607 PMCID: PMC9537058 DOI: 10.1002/cam4.5107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 11/13/2022] Open
Abstract
This review focuses on patients with differentiated thyroid carcinoma (DTC) associated with multiple primary malignant neoplasm (MPMN) treated by multikinase inhibitors (MKIs) as systemic treatment for advanced disease. Despite the increasing frequency of MPMNs (many at an advanced stage) and the usefulness of MKIs for multiple metastatic cancers, published data on the management of MPMN and MKI therapies in this scenario are scarce. There are infrequent descriptions of patients with advanced MPMN treated with MKIs, but only a few have described advanced DTC. The management of MPMNs, including DTC and its particular circumstances, is reviewed, focusing on the evidence for MKI therapies. Some considerations for MPMN patients with advanced DTC are discussed, with the intention of helping physicians make decisions in these challenging situations and improving treatment and patient outcomes.
Collapse
Affiliation(s)
- Marcel Sambo
- Endocrinology and Nutrition DepartmentGeneral University Hospital Gregorio MaranonMadridSpain
| |
Collapse
|
34
|
Hashemi G, Dight J, Khosrotehrani K, Sormani L. Melanoma Tumour Vascularization and Tissue-Resident Endothelial Progenitor Cells. Cancers (Basel) 2022; 14:4216. [PMID: 36077754 PMCID: PMC9454996 DOI: 10.3390/cancers14174216] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
The aggressiveness of solid cancers, such as melanoma, relies on their metastatic potential. It has become evident that this key cause of mortality is largely conferred by the tumour-associated stromal cells, especially endothelial cells. In addition to their essential role in the formation of the tumour vasculature, endothelial cells significantly contribute to the establishment of the tumour microenvironment, thus enabling the dissemination of cancer cells. Melanoma tumour vascularization occurs through diverse biological processes. Vasculogenesis is the formation of de novo blood vessels from endothelial progenitor cells (EPCs), and recent research has shown the role of EPCs in melanoma tumour vascularization. A more detailed understanding of the complex role of EPCs and how they contribute to the abnormal vessel structures in tumours is of importance. Moreover, anti-angiogenic drugs have a limited effect on melanoma tumour vascularization, and the role of these drugs on EPCs remains to be clarified. Overall, targeting cancer vasculature remains a challenge, and the role of anti-angiogenic drugs and combination therapies in melanoma, a focus of this review, is an area of extensive exploration.
Collapse
Affiliation(s)
| | | | - Kiarash Khosrotehrani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Laura Sormani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
35
|
Concept of Hybrid Drugs and Recent Advancements in Anticancer Hybrids. Pharmaceuticals (Basel) 2022; 15:ph15091071. [PMID: 36145292 PMCID: PMC9500727 DOI: 10.3390/ph15091071] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a complex disease, and its treatment is a big challenge, with variable efficacy of conventional anticancer drugs. A two-drug cocktail hybrid approach is a potential strategy in recent drug discovery that involves the combination of two drug pharmacophores into a single molecule. The hybrid molecule acts through distinct modes of action on several targets at a given time with more efficacy and less susceptibility to resistance. Thus, there is a huge scope for using hybrid compounds to tackle the present difficulties in cancer medicine. Recent work has applied this technique to uncover some interesting molecules with substantial anticancer properties. In this study, we report data on numerous promising hybrid anti-proliferative/anti-tumor agents developed over the previous 10 years (2011–2021). It includes quinazoline, indole, carbazole, pyrimidine, quinoline, quinone, imidazole, selenium, platinum, hydroxamic acid, ferrocene, curcumin, triazole, benzimidazole, isatin, pyrrolo benzodiazepine (PBD), chalcone, coumarin, nitrogen mustard, pyrazole, and pyridine-based anticancer hybrids produced via molecular hybridization techniques. Overall, this review offers a clear indication of the potential benefits of merging pharmacophoric subunits from multiple different known chemical prototypes to produce more potent and precise hybrid compounds. This provides valuable knowledge for researchers working on complex diseases such as cancer.
Collapse
|
36
|
Targeting NOX4 disrupts the resistance of papillary thyroid carcinoma to chemotherapeutic drugs and lenvatinib. Cell Death Dis 2022; 8:177. [PMID: 35396551 PMCID: PMC8990679 DOI: 10.1038/s41420-022-00994-7] [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: 10/21/2021] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 12/02/2022]
Abstract
Advanced differentiated thyroid cancer cells are subjected to extreme nutritional starvation which contributes to develop resistance to treatments; however, the underlying mechanism remains unclear. Cells were subjected to serum deprivation by culture in medium containing 0.5% fetal bovine serum. A CCK8 assay, cell death Detection ELISAPLUS kit, and PI staining were conducted to determine cell viability, cell apoptosis, and cell cycle, respectively. NADPH oxidase 4 (NOX4) knockdown–stable cell lines were generated by lentivirus-mediated shRNA knockdown in BCPAP cells and TPC-1 cells. Etoposide and doxorubicin, two chemotherapeutic drugs, as well as lenvatinib were utilized to determine the effect of NOX4 on drug resistance. Lenvatinib-resistant BCPAP cells (LRBCs) were established to confirm this effect. The underlining mechanisms of NOX4 under starvation were explored using western blot. Finally, GLX351322, an inhibitor targeting NOX4, was used to inhibit NOX4-derived ROS in vitro and detect its effect on drug resistance of tumor cells in vivo. NOX4 is overexpressed under serum deprivation in BCPAP or TPC-1 cells. NOX4 knockdown impairs cell viability, increases cell apoptosis, extends G1 phase during cell cycle and modulates the level of energy-associated metabolites in starved cells. When the starved cells or LRBCs are treated with chemotherapeutic drugs or Lenvatinib, NOX4 knockdown inhibits cell viability and aggravates cell apoptosis depending on NOX4-derived ROS production. Mechanistically, starvation activates TGFβ1/SMAD3 signal, which mediates NOX4 upregulation. The upregulated NOX4 then triggers ERKs and PI3K/AKT pathway to influence cell apoptosis. GLX351322, a NOX4-derived ROS inhibitor, has an inhibitory effect on cell growth in vitro and the growth of BCPAP-derived even LRBCs-derived xenografts in vivo. These findings highlight NOX4 and NOX4-derived ROS as a potential therapeutic target in resistance to PTC.
Collapse
|
37
|
Pham B, Kwon SM, Castillo DR, Majeed Y, Ahmad S, Hou J, Ganesan L, Mohammad S, Cao H. Late renal toxicity in patient with radioiodine-refractory differentiated thyroid cancer treated with lenvatinib: A case report and literature review. J Oncol Pharm Pract 2022; 28:1930-1935. [DOI: 10.1177/10781552221092329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction Thyroid carcinoma is the most common endocrine neoplasm. Multimodal therapy including surgery, radioactive iodine (RAI) therapy, and indefinite suppression of thyroid-stimulating hormone has led to an 85% cure rate in differentiated thyroid tumors (DTT). Approximately 5–10% of patients will have recurrence or metastases that have the potential to become resistant to RAI treatment. 1 10-year overall survival rates are reported to be 10% in these patients versus 56% in patients with RAI avid disease. 2 Lenvatinib, a multi-tyrosine-kinase inhibitor (TKI), was shown to have a 65% overall response rate in addition to a significant improvement in progression-free survival (PFS), approved to treat RAI-resistant DTTs. 3 , 4 Case Report We are reporting a very rare case of late renal toxicity in a 68-year-old woman with a history of type 2 diabetes and metastatic RAI-resistant follicular thyroid carcinoma (Hurthle cell variant) who developed thrombotic microangiopathy 21 months after initiation of treatment. Management & Outcome It was determined that LEN should be held, due to worsening renal function secondary to TKI-induced kidney injury. Although the patient's renal function eventually improved and returned to her baseline after discontinuation of LEN, there was marked disease progression after drug cessation. Discussion Renal toxicity is a rare adverse event (AE) that tends to occur typically within three weeks of initiation of treatment. The utilization of TKIs can lead to glomerulosclerosis, and careful considerations and precautions should be taken by clinicians who intend to initiate TKI therapy in patients with pre-existing diabetes to prevent renal toxicity.
Collapse
Affiliation(s)
- Bryan Pham
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, California, United States
| | - Sue Min Kwon
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, California, United States
| | - Dan Ran Castillo
- Hematology/Oncology Department, Loma Linda University Medical Center, Loma Linda, California, United States
| | - Yasamin Majeed
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, California, United States
| | - Sarmad Ahmad
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, California, United States
| | - Jean Hou
- Department of Pathology, Cedars Sinai Medical Center, Los Angeles, California, United States
| | - Lakshmi Ganesan
- Department of Internal Medicine, Loma Linda University Medical Center, Loma Linda, California, United States
| | - Sharif Mohammad
- Department of Nephrology, Loma Linda University Medical Center, Loma Linda, California, United States
| | - Huynh Cao
- Hematology/Oncology Department, Loma Linda University Medical Center, Loma Linda, California, United States
| |
Collapse
|
38
|
Motzer RJ, Taylor MH, Evans TRJ, Okusaka T, Glen H, Lubiniecki GM, Dutcus C, Smith AD, Okpara CE, Hussein Z, Hayato S, Tamai T, Makker V. Lenvatinib dose, efficacy, and safety in the treatment of multiple malignancies. Expert Rev Anticancer Ther 2022; 22:383-400. [PMID: 35260027 PMCID: PMC9484451 DOI: 10.1080/14737140.2022.2039123] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/03/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Lenvatinib is an oral multitargeted tyrosine kinase inhibitor that has shown efficacy and manageable safety across multiple cancer types. The recommended starting doses for lenvatinib differ across cancer types and indications based on whether it is used as monotherapy or as combination therapy. AREAS COVERED This review covers clinical trials that established the dosing paradigm and efficacy of lenvatinib and defined its adverse-event profile as a monotherapy; or in combination with the mTOR inhibitor, everolimus; or the anti-PD-1 antibody, pembrolizumab; and/or chemotherapy. EXPERT OPINION Lenvatinib has been established as standard-of-care either as a monotherapy or in combination with other anticancer agents for the treatment of radioiodine-refractory differentiated thyroid carcinoma, hepatocellular carcinoma, renal cell carcinoma, and endometrial carcinoma, and is being investigated further across several other tumor types. The dosing and adverse-event management strategies for lenvatinib have been developed through extensive clinical trial experience. Collectively, the data provide the rationale to start lenvatinib at the recommended doses and then interrupt or dose reduce as necessary to achieve required dose intensity for maximized patient benefit. The adverse-event profile of lenvatinib is consistent with that of other tyrosine kinase inhibitors, and clinicians are encouraged to review and adopt relevant symptom-management strategies.
Collapse
Affiliation(s)
- Robert J. Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY, USA
| | - Matthew H. Taylor
- Division of Hematology and Oncology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Thomas R. Jeffry Evans
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Medical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hilary Glen
- Medical Oncology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Gregory M. Lubiniecki
- Global Clinical Development, Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | | | - Ziad Hussein
- Clinical Pharmacology Science, Eisai Europe Ltd., Hatfield, UK
| | - Seiichi Hayato
- Clinical Pharmacology Science, Eisai Co., Ltd., Tokyo, Japan
| | | | - Vicky Makker
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY, USA
| |
Collapse
|
39
|
Kasherman L, Liu S(L, Karakasis K, Lheureux S. Angiogenesis: A Pivotal Therapeutic Target in the Drug Development of Gynecologic Cancers. Cancers (Basel) 2022; 14:1122. [PMID: 35267430 PMCID: PMC8908988 DOI: 10.3390/cancers14051122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/27/2023] Open
Abstract
Since the discovery of angiogenesis and its relevance to the tumorigenesis of gynecologic malignancies, a number of therapeutic agents have been developed over the last decade, some of which have become standard treatments in combination with other therapies. Limited clinical activity has been demonstrated with anti-angiogenic monotherapies, and ongoing trials are focused on combination strategies with cytotoxic agents, immunotherapies and other targeted treatments. This article reviews the science behind angiogenesis within the context of gynecologic cancers, the evidence supporting the targeting of these pathways and future directions in clinical trials.
Collapse
Affiliation(s)
- Lawrence Kasherman
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia;
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2052, Australia
- Illawarra Cancer Care Centre, Department of Medical Oncology, Wollongong, NSW 2500, Australia
| | | | | | - Stephanie Lheureux
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
40
|
Adashek JJ, Breunig JJ, Posadas E, Bhowmick NA, Ellis L, Freedland SJ, Kim H, Figlin R, Gong J. First-line Immune Checkpoint Inhibitor Combinations in Metastatic Renal Cell Carcinoma: Where Are We Going, Where Have We Been? Drugs 2022; 82:439-453. [PMID: 35175588 DOI: 10.1007/s40265-022-01683-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2022] [Indexed: 01/03/2023]
Abstract
The combination of targeted therapy and immunotherapy in the treatment of metastatic renal cell carcinoma (mRCC) has significantly improved outcomes for many patients. There are multiple FDA-approved regimens for the frontline setting based on numerous randomized Phase III trials. Despite these efforts, there remains a conundrum of identifying a biomarker-driven approach for these patients and it is unclear how to predict which patients are most likely to respond to these agents. This is due, in part, to an incomplete understanding of how these drug combinations work. The use of tyrosine kinase inhibitors that have multiple 'off-target' effects may lend themselves to the benefits observed when given in combination with immunotherapy. Further, targeting multiple clones within a patient's heterogenic tumor that are responsive to targeted therapy and others that are responsive to immunotherapy may also explain some level of improved response rates to the combination approaches compared to monotherapies. This review highlights the 5 FDA-approved regimens for mRCC in the frontline setting and offers insights into potential mechanisms for improved outcomes seen in these combination approaches.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Internal Medicine, University of South Florida, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Joshua J Breunig
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Edwin Posadas
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Neil A Bhowmick
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Leigh Ellis
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Stephen J Freedland
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA.,Division of Urology, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Section of Urology, Durham VA Medical Center, Durham, NC, USA
| | - Hyung Kim
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA.,Division of Urology, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert Figlin
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA
| | - Jun Gong
- Division of Hematology and Oncology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA, 90048, USA.
| |
Collapse
|
41
|
Hsieh SL, Li JH, Dong CD, Chen CW, Wu CC. Carnosine suppresses human colorectal cancer cell proliferation by inducing necroptosis and autophagy and reducing angiogenesis. Oncol Lett 2022; 23:44. [PMID: 34976156 PMCID: PMC8674876 DOI: 10.3892/ol.2021.13162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/06/2021] [Indexed: 12/24/2022] Open
Abstract
Carnosine (β-alanyl-L-histidine) is found in beef and fish. The present study aimed to investigate the effects of carnosine on the cell proliferation of human colorectal cancer cells. After human colorectal cancer HCT-116 cells were treated carnosine for 72 or 96 h, the cell proliferation, apoptosis, autophagy, necroptosis, angiogenesis and the expression of related regulatory molecules were detected using MTT assays, fluorescence image analysis and RT-qPCR in this study. Treatment of HCT-116 cells with 5, 10 or 15 mM carnosine for 72 or 96 h significantly decreased cell viability (P<0.05). The mRNA expression of β-catenin and transcription factor 4 (Tcf-4) was significantly reduced by 15–23% and 11–80%, respectively (P<0.05). When HCT-116 cells were treated with 15 mM carnosine, the mRNA levels of 1A/1B-light chain 3 and phosphatidylinositol 3-kinase were significantly increased by 235% and 249%, respectively (P<0.05). The mRNA level of Beclin-1 and autophagy levels were significantly increased by 137–141% in HCT-116 cells treated with 5, 10 or 15 mM carnosine (P<0.05). Carnosine (15 mM) also increased reactive oxygen species levels and mixed lineage kinase domain-like protein mRNA expression and depleted ATP levels (P<0.05). The angiogenesis-regulating molecules vascular endothelial growth factor, epidermal growth factor receptor and hypoxia-inducible factor 1-α were all significantly decreased by 10 or 15 mM carnosine treatment. These results showed that carnosine could suppress human colorectal cell proliferation by reducing β-catenin/Tcf-4 signaling, inducing autophagy and necroptosis and inhibiting angiogenesis. It was demonstrated that carnosine is a potential compound from dietary food for the future clinical treatment and/or prevention of colorectal cancer.
Collapse
Affiliation(s)
- Shu-Ling Hsieh
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan, R.O.C
| | - Jia-Huei Li
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan, R.O.C
| | - Cheng-Di Dong
- Marine Environmental Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan, R.O.C
| | - Chiu-Wen Chen
- Marine Environmental Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan, R.O.C
| | - Chih-Chung Wu
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan, R.O.C
| |
Collapse
|
42
|
Kneppers J, Bergman AM, Zwart W. Prostate Cancer Epigenetic Plasticity and Enhancer Heterogeneity: Molecular Causes, Consequences and Clinical Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:255-275. [DOI: 10.1007/978-3-031-11836-4_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
|
43
|
Sansar B, Singh N, Singh A. Lenvatinib: A potent oral tyrosine kinase inhibitor across multiple histologies with differing dosages. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_31_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
44
|
Guo J, Zhu P, Ye Z, Wang M, Yang H, Huang S, Shu Y, Zhang W, Zhou H, Li Q. YRDC Mediates the Resistance of Lenvatinib in Hepatocarcinoma Cells via Modulating the Translation of KRAS. Front Pharmacol 2021; 12:744578. [PMID: 34658879 PMCID: PMC8517968 DOI: 10.3389/fphar.2021.744578] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Lenvatinib is the latest and promising agent that has demonstrated a significant improvement of progression-free survival in advanced hepatocellular carcinoma (HCC). However, resistance emerges soon after initial treatment, limiting the clinical benefits of lenvatinib. Therefore, understanding the mechanism of resistance is necessary for improving lenvatinib efficacy. YRDC promotes the proliferation of hepatocarcinoma cells via regulating the activity of the RAS/RAF/MEK/ERK pathway, which was the primary pathway of the anticancer effect of lenvatinib. The purpose of this study is to investigate whether YRDC modulates the sensitivity of lenvatinib in hepatocarcinoma cells. Using the CCK-8 cell viability assay, wound-healing assay and clone formation assay in cell models, and xenograft assay in null mouse, we demonstrated that Huh7 cells with YRDC knockdown showed decreased susceptibility to lenvatinib than their control cells. Furthermore, we found that lenvatinib inhibited the expression of YRDC in a time-dependent manner. This effect may aggravate resistance to lenvatinib in hepatocarcinoma cells and may be an underlying cause of resistance, which emerges soon after lenvatinib initial treatment. To investigate how YRDC modulates the sensitivity of lenvatinib, we assessed the effect of tRNA with different t6A levels on the translation of the KRAS gene by in vitro rabbit reticulocyte translation system and measured the expression levels of the KRAS gene by western blot together with qPCR. We found that YRDC regulates the protein translation of KRAS in cell models, and the tRNA with low t6A modification level reduces the translation of the KRAS in the in vitro translation system. These results suggested that YRDC mediates the resistance of lenvatinib in hepatocarcinoma cells via modulating the translation of the KRAS. In this study, YRDC was confirmed to be a potential novel predictive biomarker of lenvatinib sensitivity in HCC.
Collapse
Affiliation(s)
- Jun Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Mengke Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Shiqiong Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Yan Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, MD, United States
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| |
Collapse
|
45
|
Bedrose S, Miller KC, Altameemi L, Ali MS, Nassar S, Garg N, Daher M, Eaton KD, Yorio JT, Daniel DB, Campbell M, Bible KC, Ryder M, Chintakuntlawar AV, Habra MA. Combined lenvatinib and pembrolizumab as salvage therapy in advanced adrenal cortical carcinoma. J Immunother Cancer 2021; 8:jitc-2020-001009. [PMID: 32737143 PMCID: PMC7394183 DOI: 10.1136/jitc-2020-001009] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Background There is no effective systemic therapy for metastatic adrenal cortical carcinoma (ACC) after failure of platinum-based chemotherapy. The efficacies of single-agent oral multikinase inhibitors (MKIs) or salvage immune checkpoint inhibitors (CPIs) have been very limited. It is unknown whether combining CPIs, such as pembrolizumab (PEM), with other therapies, such as MKIs, could yield higher response rates in ACC, yet this combination has shown promise in other cancers. Herein, we describe the first case series using PEM in combination with the MKI lenvatinib (LEN) in patients with progressive, metastatic ACC. Methods A retrospective case series describing the use of LEN/PEM as salvage therapy in patients with progressive/metastatic ACC. Results Eight patients were treated with the LEN/PEM combination therapy. Half were female, and the median age at time of diagnosis was 38 years (range 21–49). Three (37.5%) patients had hormonally active ACC. The median number of prior lines of systemic therapy was 4 (range 2–9). Six (75%) patients had had disease progression on prior CPIs and five (62.5%) patients had progressed on prior MKI therapy. The median progression-free survival was 5.5 months (95% CI 1.8–not reached) and median duration of therapy was 8.5 months (range 2–22). Two (25%) patients had a partial response, one (12.5%) patient had stable disease, and five (62.5%) patients had progressive disease. None of the eight patients stopped therapy because of adverse events. Conclusions In our small cohort of heavily pretreated patients with ACC, the combination of LEN/PEM was associated with objective responses in a subset of patients without significant toxicity. This combination should be formally investigated in phase II clinical trial with robust correlative studies to identify predictors for response.
Collapse
Affiliation(s)
- Sara Bedrose
- Department of Endocrine Neoplasia and Hormonal Disorders, Unit 1461, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Section of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, Texas, USA
| | | | - Lina Altameemi
- Department of Endocrine Neoplasia and Hormonal Disorders, Unit 1461, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mohamed S Ali
- Section of Endocrinology, Diabetes and Metabolism, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sameh Nassar
- Department of Abdominal Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naveen Garg
- Department of Abdominal Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marilyne Daher
- Department of Endocrine Neoplasia and Hormonal Disorders, Unit 1461, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keith D Eaton
- Department of Medical Oncology, University of Washington, Seattle, Washington, USA
| | | | | | - Matthew Campbell
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keith C Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mabel Ryder
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mouhammed Amir Habra
- Department of Endocrine Neoplasia and Hormonal Disorders, Unit 1461, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
46
|
Innovations and Patent Trends in the Development of USFDA Approved Protein Kinase Inhibitors in the Last Two Decades. Pharmaceuticals (Basel) 2021; 14:ph14080710. [PMID: 34451807 PMCID: PMC8400070 DOI: 10.3390/ph14080710] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Protein kinase inhibitors (PKIs) are important therapeutic agents. As of 31 May 2021, the United States Food and Drug Administration (USFDA) has approved 70 PKIs. Most of the PKIs are employed to treat cancer and inflammatory diseases. Imatinib was the first PKI approved by USFDA in 2001. This review summarizes the compound patents and the essential polymorph patents of the PKIs approved by the USFDA from 2001 to 31 May 2021. The dates on the generic drug availability of the PKIs in the USA market have also been forecasted. It is expected that 19 and 48 PKIs will be genericized by 2025 and 2030, respectively, due to their compound patent expiry. This may reduce the financial toxicity associated with the existing PKIs. There are nearly 535 reported PKs. However, the USFDA approved PKIs target only about 10-15% of the total said PKs. As a result, there are still a large number of unexplored PKs. As the field advances during the next 20 years, one can anticipate that PKIs with many scaffolds, chemotypes, and pharmacophores will be developed.
Collapse
|
47
|
Shalata W, Iraqi M, Bhattacharya B, Fuchs V, Roisman LC, Cohen AY, Massalha I, Yakobson A, Prasad M, Elkabets M, Porgador A, Peled N. Rapid Response to the Combination of Lenvatinib and Pembrolizumab in Patients with Advanced Carcinomas (Lung Adenocarcinoma and Malignant Pleural Mesothelioma). Cancers (Basel) 2021; 13:3630. [PMID: 34298855 PMCID: PMC8307809 DOI: 10.3390/cancers13143630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/15/2021] [Indexed: 12/29/2022] Open
Abstract
The new era of cancer treatments has made immune checkpoint inhibitors (ICIs) and emerging multikinase inhibitors (TKIs) the standards of care, thus drastically improving patient prognoses. Pembrolizumab is an anti-programmed cell death-1 antibody drug, and lenvatinib is a TKI with preferential antiangiogenic activity. We present, to our knowledge, the first reported series of cases consisting of patients with metastatic non-small cell lung cancer and malignant pleural mesothelioma who were treated with several types of chemotherapy combinations and ICIs followed by disease progression. They were subsequently treated with combined immunotherapy and TKI treatment, resulting in a near complete response within a very short time. Clinical responses were supported by in vitro testing of each patient's lymphocytic response to pembrolizumab after pre-exposure of target cancer cells to lenvatinib.
Collapse
Affiliation(s)
- Walid Shalata
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (W.S.); (A.Y.C.); (I.M.); (A.Y.)
| | - Muhammed Iraqi
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.I.); (B.B.); (V.F.); (M.P.); (M.E.)
| | - Baisali Bhattacharya
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.I.); (B.B.); (V.F.); (M.P.); (M.E.)
| | - Vered Fuchs
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.I.); (B.B.); (V.F.); (M.P.); (M.E.)
| | - Laila C. Roisman
- Shaare Zedek Medical Center, Oncology Division and Cancer Institute, Jerusalem 9103102, Israel;
| | - Ahron Yehonatan Cohen
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (W.S.); (A.Y.C.); (I.M.); (A.Y.)
| | - Ismaell Massalha
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (W.S.); (A.Y.C.); (I.M.); (A.Y.)
| | - Alexander Yakobson
- The Legacy Heritage Center & Dr. Larry Norton Institute, Soroka Medical Center, Beer Sheva 84105, Israel; (W.S.); (A.Y.C.); (I.M.); (A.Y.)
| | - Manu Prasad
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.I.); (B.B.); (V.F.); (M.P.); (M.E.)
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.I.); (B.B.); (V.F.); (M.P.); (M.E.)
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel; (M.I.); (B.B.); (V.F.); (M.P.); (M.E.)
| | - Nir Peled
- Shaare Zedek Medical Center, Oncology Division and Cancer Institute, Jerusalem 9103102, Israel;
| |
Collapse
|
48
|
Qureshi A, Michel M, Lerner J, Dasanu CA. Evolving therapeutic strategies for advanced hepatocellular carcinoma. Expert Opin Pharmacother 2021; 22:2495-2506. [PMID: 34252328 DOI: 10.1080/14656566.2021.1953473] [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: 10/20/2022]
Abstract
Introduction: While sorafenib dominated the therapeutic arena in advanced hepatocellular carcinoma (HCC) for almost a decade, newer agents and combinations have been changing the therapeutic landscape in the last years.Areas covered: The authors outline the etiopathogenesis and evaluate the diagnostics in HCC, followed by a comprehensive review of the currently approved and experimental treatment approaches, with a focus on various systemic agents and combinations of agents. The manuscript was subdivided into relevant categories, thus making it applicable for both clinical practice and research endeavors.Expert opinion: Recently, combination therapies including immune checkpoint inhibitors with anti-VEGF/R agents have shown superior clinical efficacy in HCC. The Atezolizumab-bevacizumab combination is currently the preferred first-line therapy. Single-agents cabozantinib and regorafenib as well as nivolumab-ipilimumab combination are favored as second-line therapies. Further research is needed to identify the predictors of response to various treatments and establish the distinct patient profiles that will derive most benefit. Tumor mutation analysis and germline mutation testing could identify rational therapeutic targets in HCC in the near future. As the skyline for new therapeutic agents and combinations in HCC continues to expand, the outlook as of today is cautiously optimistic in this still difficult-to-treat malignant neoplastic disease.
Collapse
Affiliation(s)
- Ammar Qureshi
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Miguel Michel
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Jaren Lerner
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Constantin A Dasanu
- Department of Hematology-Oncology, Eisenhower Lucy Curci Cancer Center, Rancho Mirage, CA, USA.,Department of Hematology-Oncology, UC San Diego Health, San Diego, CA, USA
| |
Collapse
|
49
|
Pohl MO, Busnadiego I, Marrafino F, Wiedmer L, Hunziker A, Fernbach S, Glas I, Moroz-Omori EV, Hale BG, Caflisch A, Stertz S. Combined computational and cellular screening identifies synergistic inhibition of SARS-CoV-2 by lenvatinib and remdesivir. J Gen Virol 2021; 102:001625. [PMID: 34319869 PMCID: PMC8491895 DOI: 10.1099/jgv.0.001625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/09/2021] [Indexed: 01/10/2023] Open
Abstract
Rapid repurposing of existing drugs as new therapeutics for COVID-19 has been an important strategy in the management of disease severity during the ongoing SARS-CoV-2 pandemic. Here, we used high-throughput docking to screen 6000 compounds within the DrugBank library for their potential to bind and inhibit the SARS-CoV-2 3 CL main protease, a chymotrypsin-like enzyme that is essential for viral replication. For 19 candidate hits, parallel in vitro fluorescence-based protease-inhibition assays and Vero-CCL81 cell-based SARS-CoV-2 replication-inhibition assays were performed. One hit, diclazuril (an investigational anti-protozoal compound), was validated as a SARS-CoV-2 3 CL main protease inhibitor in vitro (IC50 value of 29 µM) and modestly inhibited SARS-CoV-2 replication in Vero-CCL81 cells. Another hit, lenvatinib (approved for use in humans as an anti-cancer treatment), could not be validated as a SARS-CoV-2 3 CL main protease inhibitor in vitro, but serendipitously exhibited a striking functional synergy with the approved nucleoside analogue remdesivir to inhibit SARS-CoV-2 replication, albeit this was specific to Vero-CCL81 cells. Lenvatinib is a broadly-acting host receptor tyrosine kinase (RTK) inhibitor, but the synergistic effect with remdesivir was not observed with other approved RTK inhibitors (such as pazopanib or sunitinib), suggesting that the mechanism-of-action is independent of host RTKs. Furthermore, time-of-addition studies revealed that lenvatinib/remdesivir synergy probably targets SARS-CoV-2 replication subsequent to host-cell entry. Our work shows that combining computational and cellular screening is a means to identify existing drugs with repurposing potential as antiviral compounds. Future studies could be aimed at understanding and optimizing the lenvatinib/remdesivir synergistic mechanism as a therapeutic option.
Collapse
Affiliation(s)
- Marie O. Pohl
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Idoia Busnadiego
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Francesco Marrafino
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Department of Pharmacy, University of Salerno, Fisciano SA, 84084, Italy
| | - Lars Wiedmer
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Annika Hunziker
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Sonja Fernbach
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Irina Glas
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Elena V. Moroz-Omori
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Benjamin G. Hale
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Silke Stertz
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
50
|
Chau V, Madan RA, Aragon-Ching JB. Protein kinase inhibitors for the treatment of prostate cancer. Expert Opin Pharmacother 2021; 22:1889-1899. [PMID: 33989112 DOI: 10.1080/14656566.2021.1925250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Protein kinases have emerged as targetable pathways used in metastatic prostate cancer given their role in prostatic tumor growth, proliferation and metastases. Protein kinase inhibitors are small molecules that target varying pathways including the breakpoint cluster region (BCR)-Abelson tyrosine kinase (ABL), colony stimulating factor-1 receptor (CSF1R), vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) and phosphoinositide 3-kinase (PI3K) pathways and have been studied in prostate cancer trials with variable results. In particular, cabozantinib when used in combination trials and ipatasertib, when used with abiraterone in patients who harbor phosphatase and tensin homologue (PTEN) loss, have been promising. AREAS COVERED This article reviews the key early and late phase clinical trials currently investigating the use of protein kinase inhibitors in prostate cancer. EXPERT OPINION While multiple kinase inhibitors show promising results in prostate cancer, none have yet garnered Food and Drug Administration (FDA) approval. Studies are ongoing with the best candidate drugs discussed herein. However, multiple drugs have failed primary endpoints in prostate cancer. Therefore, further understanding of the potential mechanisms of resistance, combination and trial design of combination therapy may help pave the way for targeting kinase inhibition in prostate cancer.
Collapse
Affiliation(s)
- Vincent Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeanny B Aragon-Ching
- Genitourinary Cancers, Inova Medical Group, Inova Schar Cancer Institute, Fairfax, VA, USA.,Department of Internal Medicine, University of Virginia University School of Medicine, Charlottesville, VA, USA
| |
Collapse
|