1
|
Shelley CS, Galiègue-Zouitina S, Andritsos LA, Epperla N, Troussard X. The role of the JunD-RhoH axis in the pathogenesis of hairy cell leukemia and its ability to identify existing therapeutics that could be repurposed to treat relapsed or refractory disease. Leuk Lymphoma 2025; 66:637-655. [PMID: 39689307 DOI: 10.1080/10428194.2024.2438800] [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: 07/17/2024] [Revised: 11/19/2024] [Accepted: 12/01/2024] [Indexed: 12/19/2024]
Abstract
Hairy cell leukemia (HCL) is an indolent malignancy of mature B-lymphocytes. While existing front-line therapies achieve excellent initial results, a significant number of patients relapse and become increasingly treatment resistant. A major molecular driver of HCL is aberrant interlocking expression of the transcription factor JunD and the intracellular signaling molecule RhoH. Here we discuss the molecular basis of how the JunD-RhoH axis contributes to HCL pathogenesis. We also discuss how leveraging the JunD-RhoH axis identifies CD23, CD38, CD66a, CD115, CD269, integrin β7, and MET as new potential therapeutic targets. Critically, preclinical studies have already demonstrated that targeting CD38 with isatuximab effectively treats preexisiting HCL. Isatuximab and therapeutics directed against each of the other six new HCL targets are currently in clinical use to treat other disorders. Consequently, leveraging the JunD-RhoH axis has identified a battery of therapies that could be repurposed as new means of treating relapsed or refractory HCL.
Collapse
Affiliation(s)
| | | | - Leslie A Andritsos
- Division of Hematology Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Narendranath Epperla
- Division of Hematology, University of Utah Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Xavier Troussard
- Hematology CHU Caen Normandie, INSERM1245, MICAH, Normandie University of Caen and Rouen, UNIROUEN, UNICAEN, Hematology Institute, University Hospital Caen, Caen, France
| |
Collapse
|
2
|
Yang H, Zhang Y, Li Y, Peng S, An R, Du N, Cao J, Chu F, Min J. Bispecific c-Met/PD-1 CAR-T Cells Have Enhanced Therapeutic Effects on Solid Tumor. Technol Cancer Res Treat 2025; 24:15330338251336850. [PMID: 40261343 PMCID: PMC12035267 DOI: 10.1177/15330338251336850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/05/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
ObjectiveTo evaluate the killing effect of c-Met CAR-T on tumor cells with different degrees of c-Met expression. It was demonstrated that CAR-T autocrine PD-1 antibody could alleviate immune checkpoint inhibition and enhance the anti-tumor effect of T cells.MethodsThe specificity and clinical significance of c-Met and PD-L1 expression in various solid tumors were verified by bioinformatics analysis. c-Met specific CAR-T and c-Met specific CAR-T secreted by PD-L1 were synthesized, and c-Met CAR-T and c-Met/PD-1 CAR-T were prepared by constructing lentivirus. Flow cytometry was used to verify the positive rate and cell population of CAR-T, western blot was used to verify the secretion of PD-1 antibody, and cck-8 was used to detect the proliferation of CAR-T in tumor cells with different c-Met expression. LDH and ELISA further evaluated the antitumor effects of c-Met CAR-T and c-Met/PD-1 CAR-T in vitro.Resultsc-Met and PD-L1 were expressed in pancreatic cancer, ovarian cancer, esophageal cancer, bladder cancer, glioma and other tumors, and were associated with a variety of immune cell infiltration. Tumor cells with high expression of c-Met can strongly stimulate the proliferation of c-Met CAR-T, and c-Met CAR-T has strong cell lysis ability on tumor cells with high expression of c-Met. Autocrine PD-1 antibody can significantly improve the activity of c-Met CAR T cells, tumor lysis ability and cytokine secretion level.ConclusionThe antitumor activity of c-Met CAR-T is positively correlated with the expression of c-Met. c-Met CAR-T secreted by PD-1 showed enhanced antitumor function in solid tumor treatment.
Collapse
Affiliation(s)
- HuanRan Yang
- Department of Stomatology, BengBu Medical University, BengBu, China
| | - YanJun Zhang
- Department of Life Science, BengBu Medical University, BengBu, China
| | - YanQiu Li
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| | - Shang Peng
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| | - Ran An
- Department of Public foundation, BengBu Medical University, BengBu, China
| | - NaNa Du
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| | - JiaWei Cao
- Department of Clinical medicine, Bengbu Medical University, Bengbu, China
| | - Fei Chu
- The first affiliated Hospital, BengBu Medical University, BengBu, China
| | - JingTing Min
- Department of Basic Medicine, BengBu Medical University, BengBu, China
| |
Collapse
|
3
|
Lutimba S, Saleem B, Aleem E, Mansour MA. In Silico Analysis of Triamterene as a Potential Dual Inhibitor of VEGFR-2 and c-Met Receptors. J Xenobiot 2024; 14:1962-1987. [PMID: 39728413 DOI: 10.3390/jox14040105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
The vascular endothelial growth factor receptor 2 (VEGFR2) and the hepatocyte growth factor receptor (C-Met) are critical receptors for signaling pathways controlling crucial cellular processes such as cell growth, angiogenesis and tissue regeneration. However, dysregulation of these proteins has been reported in different diseases, particularly cancer, where these proteins promote tumour growth, invasiveness, metastasis and resistance to conventional therapies. The identification of dual inhibitors targeting both VEGFR-2 and c-Met has emerged as a strategic therapeutic approach to overcome the limitations and resistance mechanisms associated with single-target therapies in clinical settings. Through molecular dynamics simulations and comparative docking analysis, we tested the inhibitory potential of 2,016 Food and Drug Administration (FDA)-approved drugs targeting VEGFR-2 and/or c-Met receptors. The results revealed that entacapone and telmisartan are potent and selective inhibitors for c-Met and VEGFR-2, respectively. Interestingly, triamterene was identified as a promising dual inhibitor, demonstrating specific and significant binding affinity to both proteins. Molecular dynamics simulations revealed key interactions between the identified compounds and critical residues in the catalytic domains of both VEGFR-2 (e.g., Lys868, Asp1028, Asp1046) and c-Met (e.g., Asp1204, His1202, Asp1222), providing insights into their mechanism of action. These findings underscore the therapeutic potential of triamterene in targeting multiple signaling pathways involved in cancer progression, metastasis and poor prognosis in patients. Our study provides a foundational framework for the development of novel anticancer compounds able to target multiple pathways in cancer. Further preclinical and clinical investigations are needed to validate the efficacy of these compounds in clinical settings and to test their ability to overcome resistance and improve patient outcome.
Collapse
Affiliation(s)
- Stuart Lutimba
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Baraya Saleem
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Eiman Aleem
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
| | - Mohammed A Mansour
- Cancer Biology and Therapy Laboratory, School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
4
|
Thu YM, Suzawa K, Tomida S, Ochi K, Tsudaka S, Takatsu F, Date K, Matsuda N, Iwata K, Nakata K, Shien K, Yamamoto H, Okazaki M, Sugimoto S, Toyooka S. PAI-1 mediates acquired resistance to MET-targeted therapy in non-small cell lung cancer. PLoS One 2024; 19:e0300644. [PMID: 38758826 PMCID: PMC11101109 DOI: 10.1371/journal.pone.0300644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/03/2024] [Indexed: 05/19/2024] Open
Abstract
Mechanisms underlying primary and acquired resistance to MET tyrosine kinase inhibitors (TKIs) in managing non-small cell lung cancer remain unclear. In this study, we investigated the possible mechanisms acquired for crizotinib in MET-amplified lung carcinoma cell lines. Two MET-amplified lung cancer cell lines, EBC-1 and H1993, were established for acquired resistance to MET-TKI crizotinib and were functionally elucidated. Genomic and transcriptomic data were used to assess the factors contributing to the resistance mechanism, and the alterations hypothesized to confer resistance were validated. Multiple mechanisms underlie acquired resistance to crizotinib in MET-amplified lung cancer cell lines. In EBC-1-derived resistant cells, the overexpression of SERPINE1, the gene encoding plasminogen activator inhibitor-1 (PAI-1), mediated the drug resistance mechanism. Crizotinib resistance was addressed by combination therapy with a PAI-1 inhibitor and PAI-1 knockdown. Another mechanism of resistance in different subline cells of EBC-1 was evaluated as epithelial-to-mesenchymal transition with the upregulation of antiapoptotic proteins. In H1993-derived resistant cells, MEK inhibitors could be a potential therapeutic strategy for overcoming resistance with downstream mitogen-activated protein kinase pathway activation. In this study, we revealed the different mechanisms of acquired resistance to the MET inhibitor crizotinib with potential therapeutic application in patients with MET-amplified lung carcinoma.
Collapse
Affiliation(s)
- Yin Min Thu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ken Suzawa
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuta Tomida
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Kosuke Ochi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shimpei Tsudaka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Fumiaki Takatsu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keiichi Date
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Naoki Matsuda
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuma Iwata
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kentaro Nakata
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Kazuhiko Shien
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromasa Yamamoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
5
|
Zeng M, Liu C, Gong H, Tang Z, Wen J, Wang S, Xiao S. Therapeutic potential of tyrosine-protein kinase MET in osteosarcoma. Front Mol Biosci 2024; 11:1367331. [PMID: 38596618 PMCID: PMC11002252 DOI: 10.3389/fmolb.2024.1367331] [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: 01/08/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Osteosarcoma, the most prevalent primary bone tumor in children and young adults, can often be successfully treated with standard chemotherapy and surgery when diagnosed at an early stage. However, patients presenting with metastases face significant challenges in achieving a cure. Despite advancements in classical therapies over the past few decades, clinical outcomes for osteosarcoma have not substantially improved. Recently, there has been increased understanding of the biology of osteosarcoma, leading to the identification of new therapeutic targets. One such target is MET, a tyrosine kinase receptor for Hepatocyte Growth Factor (HGF) encoded by the MET gene. In vitro and in vivo studies have demonstrated that the HGF/MET pathway plays a crucial role in cancer growth, invasion, metastasis, and drug resistance across various cancers. Clinical trials targeting this pathway are already underway for lung cancer and hepatocellular carcinoma. Moreover, MET has also been implicated in promoting osteosarcoma progression. This review summarizes 3 decades' worth of research on MET's involvement in osteosarcoma and further explores its potential as a therapeutic target for patients with this disease.
Collapse
Affiliation(s)
- Ming Zeng
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Can Liu
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Haoli Gong
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zhongwen Tang
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Sisi Wang
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Sheng Xiao
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
6
|
Luo NY, Minne RL, Gallant JP, Gunaratne GS, West JL, Javeri S, Robertson AJ, Lake EW, Engle JW, Mixdorf JC, Aluicio-Sarduy E, Nickel KP, Hernandez R, Kimple RJ, Baschnagel AM, LeBeau AM. Development of an Engineered Single-Domain Antibody for Targeting MET in Non-Small Cell Lung Cancer. Bioconjug Chem 2024; 35:389-399. [PMID: 38470611 PMCID: PMC12060584 DOI: 10.1021/acs.bioconjchem.4c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
The Mesenchymal Epithelial Transition (MET) receptor tyrosine kinase is upregulated or mutated in 5% of non-small-cell lung cancer (NSCLC) patients and overexpressed in multiple other cancers. We sought to develop a novel single-domain camelid antibody with high affinity for MET that could be used to deliver conjugated payloads to MET expressing cancers. From a naïve camelid variable-heavy-heavy (VHH) domain phage display library, we identified a VHH clone termed 1E7 that displayed high affinity for human MET and was cross-reactive with MET across multiple species. When expressed as a bivalent human Fc fusion protein, 1E7-Fc was found to selectively bind to EBC-1 (MET amplified) and UW-Lung 21 (MET exon 14 mutated) cell lines by flow cytometry and immunofluorescence imaging. Next, we investigated the ability of [89Zr]Zr-1E7-Fc to detect MET expression in vivo by PET/CT imaging. [89Zr]Zr-1E7-Fc demonstrated rapid localization and high tumor uptake in both xenografts with a %ID/g of 6.4 and 5.8 for EBC-1 and UW-Lung 21 at 24 h, respectively. At the 24 h time point, clearance from secondary and nontarget tissues was also observed. Altogether, our data suggest that 1E7-Fc represents a platform technology that can be employed to potentially both image and treat MET-altered NSCLC.
Collapse
Affiliation(s)
- Natalie Y. Luo
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Rachel L. Minne
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Joseph P. Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Gihan S Gunaratne
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Jayden L. West
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Saahil Javeri
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Austin J. Robertson
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Molecular and Cellular Pharmacology Program, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Eric W. Lake
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Jonathan W. Engle
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Jason C. Mixdorf
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Eduardo Aluicio-Sarduy
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Kwang P. Nickel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Reinier Hernandez
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Randall J. Kimple
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Andrew M. Baschnagel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| | - Aaron M. LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, United States
| |
Collapse
|
7
|
Evers A, Krah S, Demir D, Gaa R, Elter D, Schroeter C, Zielonka S, Rasche N, Dotterweich J, Knuehl C, Doerner A. Engineering hydrophobicity and manufacturability for optimized biparatopic antibody-drug conjugates targeting c-MET. MAbs 2024; 16:2302386. [PMID: 38214660 PMCID: PMC10793681 DOI: 10.1080/19420862.2024.2302386] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024] Open
Abstract
Optimal combinations of paratopes assembled into a biparatopic antibody have the capacity to mediate high-grade target cross-linking on cell membranes, leading to degradation of the target, as well as antibody and payload delivery in the case of an antibody-drug conjugate (ADC). In the work presented here, molecular docking suggested a suitable paratope combination targeting c-MET, but hydrophobic patches in essential binding regions of one moiety necessitated engineering. In addition to rational design of HCDR2 and HCDR3 mutations, site-specific spiking libraries were generated and screened in yeast and mammalian surface display approaches. Comparative analyses revealed similar positions amendable for hydrophobicity reduction, with a broad combinatorial diversity obtained from library outputs. Optimized variants showed high stability, strongly reduced hydrophobicity, retained affinities supporting the desired functionality and enhanced producibility. The resulting biparatopic anti-c-MET ADCs were comparably active on c-MET expressing tumor cell lines as REGN5093 exatecan DAR6 ADC. Structural molecular modeling of paratope combinations for preferential inter-target binding combined with protein engineering for manufacturability yielded deep insights into the capabilities of rational and library approaches. The methodologies of in silico hydrophobicity identification and sequence optimization could serve as a blueprint for rapid development of optimal biparatopic ADCs targeting further tumor-associated antigens in the future.
Collapse
Affiliation(s)
- Andreas Evers
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Deniz Demir
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Ramona Gaa
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Desislava Elter
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Stefan Zielonka
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Nicolas Rasche
- ADC and Targeted Therapeutics, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Christine Knuehl
- Research Unit Oncology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Achim Doerner
- Antibody Discovery and Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
8
|
Araghi M, Mannani R, Heidarnejad maleki A, Hamidi A, Rostami S, Safa SH, Faramarzi F, Khorasani S, Alimohammadi M, Tahmasebi S, Akhavan-Sigari R. Recent advances in non-small cell lung cancer targeted therapy; an update review. Cancer Cell Int 2023; 23:162. [PMID: 37568193 PMCID: PMC10416536 DOI: 10.1186/s12935-023-02990-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 07/12/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer continues to be the leading cause of cancer-related death worldwide. In the last decade, significant advancements in the diagnosis and treatment of lung cancer, particularly NSCLC, have been achieved with the help of molecular translational research. Among the hopeful breakthroughs in therapeutic approaches, advances in targeted therapy have brought the most successful outcomes in NSCLC treatment. In targeted therapy, antagonists target the specific genes, proteins, or the microenvironment of tumors supporting cancer growth and survival. Indeed, cancer can be managed by blocking the target genes related to tumor cell progression without causing noticeable damage to normal cells. Currently, efforts have been focused on improving the targeted therapy aspects regarding the encouraging outcomes in cancer treatment and the quality of life of patients. Treatment with targeted therapy for NSCLC is changing rapidly due to the pace of scientific research. Accordingly, this updated study aimed to discuss the tumor target antigens comprehensively and targeted therapy-related agents in NSCLC. The current study also summarized the available clinical trial studies for NSCLC patients.
Collapse
Affiliation(s)
- Mahmood Araghi
- Department of Pathology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Mannani
- Vascular Surgeon, Department of Surgery, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Adel Hamidi
- Razi Vaccine and Serum Research Institute, Arak Branch, karaj, Iran
| | - Samaneh Rostami
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Fatemeh Faramarzi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sahar Khorasani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warsaw, Poland
| |
Collapse
|
9
|
Karpinski P, Rosales I, Laczmanski L, Kowalik A, Wenson S, Hoang MP. Expression of Genes Associated With Epithelial-Mesenchymal Transition in Merkel Cell Polyomavirus-Negative Merkel Cell Carcinoma. J Transl Med 2023; 103:100177. [PMID: 37207705 DOI: 10.1016/j.labinv.2023.100177] [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: 12/28/2022] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/21/2023] Open
Abstract
Two accepted possible pathways for Merkel cell carcinoma (MCC) pathogenesis include the clonal integration of the Merkel cell polyomavirus (MCPyV) into the neoplastic cells and by UV irradiation. We hypothesize that, in UV etiology, the expression of genes associated with epithelial-mesenchymal transition (EMT) would be higher in MCPyV-negative MCCs. We compared RNA expression in 16 MCPyV-negative with that in 14 MCPyV-positive MCCs in 30 patients using NanoString panel of 760 gene targets as an exploratory method. Subsequently, we confirmed the findings with a publicly available RNA sequencing data set. The NanoString method showed that 29 of 760 genes exhibited significant deregulation. Ten genes (CD44, COL6A3, COL11A1, CXCL8, INHBA, MMP1, NID2, SPP1, THBS1, and THY1) were part of the EMT pathway. The expression of CDH1/E-cadherin, a key EMT gene, and TWIST1, regulator gene of EMT, was higher in MCPyV-negative tumors. To further investigate the expression of EMT genes in MCPyV-negative MCCs, we analyzed publicly available RNA sequencing data of 111 primary MCCs. Differential expression and gene set enrichment analysis of 35 MCPyV-negative versus 76 MCPyV-positive MCCs demonstrated significantly higher expression of EMT-related genes and associated pathways such as Notch signaling, TGF-β signaling, and Hedgehog signaling, and UV response pathway in MCPyV-negative MCCs. The significance of the EMT pathway in MCPyV-negative MCCs was confirmed independently by a coexpression module analysis. One of the modules (M3) was specifically activated in MCPyV-negative MCCs and showed significant enrichment for genes involved in EMT. A network analysis of module M3 revealed that CDH1/E-cadherin was among the most connected genes (hubs). E-cadherin and LEF1 immunostains demonstrated significantly more frequent expression in MCPvV-negative versus MCPyV-positive tumors (P < .0001). In summary, our study showed that the expression of EMT-associated genes is higher in MCPyV-negative MCC. Because EMT-related proteins can be targeted, the identification of EMT pathways in MCPyV-negative MCCs is of potential therapeutic relevance.
Collapse
Affiliation(s)
- Pawel Karpinski
- Department of Genetics, Wroclaw Medical University, Wroclaw, Poland; Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Laboratory of Genomics and Bioinformatics, Wroclaw, Poland
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lukasz Laczmanski
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Laboratory of Genomics and Bioinformatics, Wroclaw, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland; Division of Medical Biology, Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Scott Wenson
- Department of Pathology, Newton-Wellesley Hospital, Boston, Massachusetts
| | - Mai P Hoang
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
10
|
Li D, Wang J, Liu C, Luo Y, Xu H, Wang Y, Sun N, He J. Making the Best Use of Available Weapons for the Inevitable Rivalry-Resistance to EGFR-TKIs. Biomedicines 2023; 11:biomedicines11041141. [PMID: 37189759 DOI: 10.3390/biomedicines11041141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/19/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023] Open
Abstract
The emergence of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) revolutionized the treatment of advanced-stage non-small cell lung cancer (NSCLC). Detected in more than 50% of late-stage lung adenocarcinoma in Asian patients, the EGFR mutation was regarded as a golden mutation for Asians. However, resistance to TKIs seems inevitable and severely hinders patients from getting further benefits from treatment. Even though resistance caused by EGFR T790M could be effectively managed by third-generation EGFR-TKIs currently, resistance to third-generation EGFR-TKIs is still a troublesome issue faced by both clinicians and patients. Various efforts have been made to maximize the benefits of patients from EGFR-TKIs therapy. Thus, new requirements and challenges have been posed to clinicians of this era. In this review, we summarized the clinical evidence on the efficacy of third-generation EGFR-TKIs in patients with EGFR-mutated NSCLC. Then, we discussed advancements in sequential treatment aiming to delay the onset of resistance. Moreover, the resistance mechanisms and features were depicted to help us better understand our enemies. Lastly, we put forward future strategies, including recent approaches involving the utilization of antibody drug conjugates against resistance and research directions about shaping the evolution of NSCLC as a core idea in the management of NSCLC.
Collapse
Affiliation(s)
- Dongyu Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- 4 + 4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Jingnan Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chengming Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuejun Luo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Haiyan Xu
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
11
|
Therapeutic strategies for non-small cell lung cancer: Experimental models and emerging biomarkers to monitor drug efficacies. Pharmacol Ther 2023; 242:108347. [PMID: 36642389 DOI: 10.1016/j.pharmthera.2023.108347] [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: 09/15/2022] [Revised: 12/15/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
While new targeted therapies have considerably changed the treatment and prognosis of non-small cell lung cancer (NSCLC), they are frequently unsuccessful due to primary or acquired resistances. Chemoresistance is a complex process that combines cancer cell intrinsic mechanisms including molecular and genetic abnormalities, aberrant interactions within the tumor microenvironment, and the pharmacokinetic characteristics of each molecule. From a pharmacological point of view, two levers could improve the response to treatment: (i) developing tools to predict the response to chemo- and targeted therapies and (ii) gaining a better understanding of the influence of the tumor microenvironment. Both personalized medicine approaches require the identification of relevant experimental models and biomarkers to understand and fight against chemoresistance mechanisms. After describing the main therapies in NSCLC, the scope of this review will be to identify and to discuss relevant in vitro and ex vivo experimental models that are able to mimic tumors. In addition, the interests of these models in the predictive responses to proposed therapies will be discussed. Finally, this review will evaluate the involvement of novel secreted biomarkers such as tumor DNA or micro RNA in predicting responses to anti-tumor therapies.
Collapse
|
12
|
Tang W, Zhang Y, Zhang H, Zhang Y. Vascular Niche Facilitates Acquired Drug Resistance to c-Met Inhibitor in Originally Sensitive Osteosarcoma Cells. Cancers (Basel) 2022; 14:cancers14246201. [PMID: 36551686 PMCID: PMC9776923 DOI: 10.3390/cancers14246201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents characterized by drug resistance and poor prognosis. As one of the key oncogenes, c-Met is recognized as a promising therapeutic target for OS. In this report, we show that c-Met inhibitor PF02341066 specifically killed OS cells with highly phosphorylated c-Met in vitro. However, the inhibitory effect of PF02341066 was abrogated in vivo due to interference from the vascular niche. OS cells adjacent to microvessels or forming vascular mimicry suppressed c-Met expression and phosphorylation. Moreover, VEGFR2 was activated in OS cells and associated with acquired drug resistance. Dual targeting of c-Met and VEGFR2 could effectively shrink the tumor size in a xenograft model. c-Met-targeted therapy combined with VEGFR2 inhibition might be beneficial to achieve an ideal therapeutic effect in OS patients. Together, our results confirm the pivotal role of tumor heterogeneity and the microenvironment in drug response and reveal the molecular mechanism underlying acquired drug resistance to c-Met-targeted therapy.
Collapse
Affiliation(s)
| | | | | | - Yan Zhang
- Correspondence: ; Tel.: +86-20-3933-2955
| |
Collapse
|
13
|
Clough W, Al Jaberi M, Dalia S. Non-small cell lung cancer with MET exon 14 skip mutation: case report. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1287. [PMID: 36618809 PMCID: PMC9816822 DOI: 10.21037/atm-2022-53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/19/2022] [Indexed: 11/21/2022]
Abstract
Background Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, making up 80-85% of all lung malignancies. It can be further subdivided into different types. The three main subtypes are adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. Traditionally, NSCLCs have been treated with a combination of chemotherapy, surgery, and/or radiation therapy. However, with the advent of genotype subtype analysis and targeted therapy it has become possible to have individualized treatment options for patients with NSCLC. Case Description We present a case report of a 68-year-old female with NSCLC. Patient initially only received radiation therapy due to her not being a surgical candidate. While initial treatment was responsive, later imaging showed metastasis of disease. Subsequent genotype analysis of the patient's tumor indicated a MET exon 14 skipping mutation which qualified her for treatment with Capmatinib (Tabrecta). Conclusions Patients on Capmatinib have minimal side effects and better efficacy than traditional chemotherapy. Patients with MET exon 14 skipping mutations should be considered for Capmatinib therapy.
Collapse
Affiliation(s)
| | | | - Samir Dalia
- Department of Medical Oncology, Mercy Hospital, Joplin, MO, USA
| |
Collapse
|
14
|
Michaels E, Bestvina CM. Meeting an un-MET need: Targeting MET in non-small cell lung cancer. Front Oncol 2022; 12:1004198. [PMID: 36338701 PMCID: PMC9634070 DOI: 10.3389/fonc.2022.1004198] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
The MET pathway can be activated by MET exon 14 skipping mutations, gene amplification, or overexpression. Mutations within this pathway carry a poor prognosis for patients with non-small cell lung cancer (NSCLC). MET exon 14 skipping mutations occur in 3-4% of patients with NSCLC, while MET amplifications are found in 1-6% of patients. The most effective method for detection of MET amplification is fluorescent in situ hybridization (FISH) and of MET exon 14 skipping mutations is RNA-based next generation sequencing (NGS). Immunohistochemistry (IHC) is an alternative method of diagnosis but is not as reliable. Early studies of MET tyrosine kinase inhibitors (TKIs) demonstrated limited clinical benefit. However, newer selective MET TKIs, such as capmatinib and tepotinib, have improved efficacy. Both drugs have an acceptable safety profile with the most common treatment-related adverse event being peripheral edema. One of the most frequent resistance mechanisms to EGFR inhibition with osimertinib is MET amplification. There is interest in combining EGFR inhibition plus MET inhibition in an attempt to target this resistance mechanism. Additional ways of targeting MET alterations are currently under investigation, including the bi-specific antibody amivantamab. Additional research is needed to further understand resistance mechanisms to MET inhibition. There is limited research into the efficacy of immune checkpoint inhibition for MET-altered NSCLC, though some data suggests decreased efficacy compared with wild-type patients and increased toxicity associated with the combination of immunotherapy and MET TKIs. Future directions for research will include combination clinical trials and understanding rational combinations for MET alterations.
Collapse
Affiliation(s)
- Elena Michaels
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Christine M. Bestvina
- Department of Medicine, University of Chicago Comprehensive Cancer Center, Chicago, IL, United States
| |
Collapse
|
15
|
Mach T, Qi A, Bouganim N, Trinh E. Targeted Cancer Therapies Causing Elevations in Serum Creatinine Through Tubular Secretion Inhibition: A Case Report and Review of the Literature. Can J Kidney Health Dis 2022; 9:20543581221106246. [PMID: 35756332 PMCID: PMC9218431 DOI: 10.1177/20543581221106246] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/17/2022] [Indexed: 02/06/2023] Open
Abstract
Rationale Targeted cancer therapies have revolutionized the field of oncology by selecting for specific molecular pathways, thus improving overall clinical prognosis. However, many of these targeted treatments have been reported to have adverse kidney effects, including acute kidney injury, interstitial nephritis, and glomerular disease. Furthermore, some of these targeted therapies have also been found to cause an asymptomatic rise in serum creatinine levels through inhibition of active tubular secretion. Presenting concerns A 79-year-old woman was being followed for stage 4 A2 chronic kidney disease secondary to type 2 diabetes and longstanding hypertension. She was diagnosed with invasive mammary carcinoma and was initiated on letrozole, an aromatase inhibitor, and palbociclib, a selective cyclin-dependent kinase inhibitor, was subsequently added. Prior to the initiation of her treatments, her baseline estimated glomerular filtration rate (eGFR) fluctuated between 25 and 28 mL/min/1.73 m2 over the previous year. After initiating palbociclib, her serum creatinine progressively increased, despite having well-controlled blood pressure and diabetes. In addition, there was no history of pre-renal events nor any sonographic evidence of obstruction. Within 7 months, her eGFR based on serum creatinine had decreased down to 12 mL/min/1.73 m2. Interventions Given that there were no clinical or other biochemical changes suggestive of worsening renal function, a serum cystatin C was measured using an immunoturbidimetric assay, which was 1.71 mg/L and correlated with an eGFR of 33 mL/min/1.73 m2 based on the chronic kidney disease epidemiology collaboration (CKD-EPI) cystatin C equation (2012). This value was consistent with her previous baseline. Based on these findings, the significant decrease in eGFR measured by serum creatinine was attributed to the inhibitory effects of palbociclib on tubular creatinine secretion, rather than representing true kidney damage. Thus, a kidney biopsy was not performed in this context. Outcomes Seven months later, a repeat serum cystatin C was repeated to assess for any worsening of the patient's kidney function and revealed an eGFR of 35 mL/min/1.73 m2 based on the CKD-EPI cystatin C equation (2012), thus revealing stable kidney function and reinforcing the inhibitory effects of palbociclib on tubular creatinine secretion through its direct effects on kidney transporters. Teaching points This case report and literature review acknowledges the importance of using alternative methods of assessing kidney function when patients are undergoing targeted cancer therapies known to affect tubular creatinine secretion, which include cyclin-dependent kinase 4/6 inhibitors, poly(adenosine diphosphate-ribose) polymerase inhibitors, tyrosine kinase inhibitors, and mesenchymal-epithelial transition inhibitors. The use of non-creatinine-based markers of glomerular filtration rate (GFR), such as cystatin C and nuclear renal scans, will allow for more accurate estimation of kidney function in the appropriate setting, thus avoiding invasive diagnostic tests and unnecessary adjustments of treatment plans. However, certain targeted cancer therapies have also been proven to cause true kidney injury; therefore, physicians must still maintain a high degree of suspicion and consider invasive investigations and/or cessation or reduction of treatments when alternative measurements of kidney function do not suggest an underestimation of GFR via serum creatinine.
Collapse
Affiliation(s)
| | - Amy Qi
- McGill University, Montreal, QC, Canada
| | - Nathaniel Bouganim
- Division of Oncology, Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Emilie Trinh
- Division of Nephrology, Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
16
|
Yang X, Liao HY, Zhang HH. Roles of MET in human cancer. Clin Chim Acta 2021; 525:69-83. [PMID: 34951962 DOI: 10.1016/j.cca.2021.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/18/2023]
Abstract
The MET proto-oncogene was first identified in osteosarcoma cells exposed to carcinogens. Although expressed in many normal cells, MET is overexpressed in many human cancers. MET is involved in the initiation and development of various human cancers and mediates proliferation, migration and invasion. Accordingly, MET has been successfully used as a biomarker for diagnosis and prognosis, survival, post-operative recurrence, risk assessment and pathologic grading, as well as a therapeutic target. In addition, recent work indicates that inhibition of MET expression and function has potential clinical benefit. This review summarizes the role, mechanism, and clinical significance of MET in the formation and development of human cancer.
Collapse
Affiliation(s)
- Xin Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
17
|
Sugiyama MG, Cui H, Redka DS, Karimzadeh M, Rujas E, Maan H, Hayat S, Cheung K, Misra R, McPhee JB, Viirre RD, Haller A, Botelho RJ, Karshafian R, Sabatinos SA, Fairn GD, Madani Tonekaboni SA, Windemuth A, Julien JP, Shahani V, MacKinnon SS, Wang B, Antonescu CN. Multiscale interactome analysis coupled with off-target drug predictions reveals drug repurposing candidates for human coronavirus disease. Sci Rep 2021; 11:23315. [PMID: 34857794 PMCID: PMC8640055 DOI: 10.1038/s41598-021-02432-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/03/2021] [Indexed: 12/20/2022] Open
Abstract
The COVID-19 pandemic has highlighted the urgent need for the identification of new antiviral drug therapies for a variety of diseases. COVID-19 is caused by infection with the human coronavirus SARS-CoV-2, while other related human coronaviruses cause diseases ranging from severe respiratory infections to the common cold. We developed a computational approach to identify new antiviral drug targets and repurpose clinically-relevant drug compounds for the treatment of a range of human coronavirus diseases. Our approach is based on graph convolutional networks (GCN) and involves multiscale host-virus interactome analysis coupled to off-target drug predictions. Cell-based experimental assessment reveals several clinically-relevant drug repurposing candidates predicted by the in silico analyses to have antiviral activity against human coronavirus infection. In particular, we identify the MET inhibitor capmatinib as having potent and broad antiviral activity against several coronaviruses in a MET-independent manner, as well as novel roles for host cell proteins such as IRAK1/4 in supporting human coronavirus infection, which can inform further drug discovery studies.
Collapse
Affiliation(s)
- Michael G Sugiyama
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Haotian Cui
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
| | | | | | - Edurne Rujas
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Biofisika Institute (CSIC, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Hassaan Maan
- Vector Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Centre, Toronto, ON, Canada
| | - Sikander Hayat
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA, USA
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Kyle Cheung
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
| | - Rahul Misra
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Joseph B McPhee
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
| | - Russell D Viirre
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
| | - Andrew Haller
- Phoenox Pharma, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Roberto J Botelho
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
| | - Raffi Karshafian
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between Ryerson University and St. Michael's Hospital, Toronto, ON, Canada
- Department of Physics, Ryerson University, Toronto, ON, Canada
| | - Sarah A Sabatinos
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | | | | | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Immunology, Toronto, ON, Canada
| | | | | | - Bo Wang
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.
- Vector Institute, Toronto, ON, Canada.
- Peter Munk Cardiac Centre, University Health Centre, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada.
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada.
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.
| |
Collapse
|
18
|
Li X, Yao Y, Qian J, Jin G, Zeng G, Zhao H. Overexpression and diagnostic significance of INTS7 in lung adenocarcinoma and its effects on tumor microenvironment. Int Immunopharmacol 2021; 101:108346. [PMID: 34781123 DOI: 10.1016/j.intimp.2021.108346] [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] [Received: 07/25/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Lung cancer is the leading cause of death worldwide, and lung adenocarcinoma (LUAD) is the most common histological subtype. INTS7, one of the subunits of the integrator complex, is upregulated in several tumors. Thus, we aimed to investigate the expression profile and clinical significance of INTS7 in LUAD. METHODS The expression profile of INTS7 was tested in TCGA database and clinical specimens. ROC curve was used to detect the diagnostic value of INTS7, CEA and INTS7 combined with CEA. Kaplan-Meier analysis was used to analyze the prognostic value of INTS7. Differentially expressed genes (DEGs) related to INTS7 were analyzed, and functional enrichment analysis was used to explore the potential mechanisms related to DEGs. The correlations between INTS7 and tumor-infiltrating immune cells, immune scores, stromal scores, and immune checkpoints were explored. Finally, the relationship between INTS7 expression and sensitivity to molecular-targeted therapy was examined. RESULTS Data from TCGA database showed that INTS7 mRNA expression was substantially upregulated in LUAD, the AUC values of INTS7 for diagnosing LUAD were >0.8, combined detection of INTS7 and CEA could improve the diagnostic efficiency and early stage patients with high expression of INTS7 showed shorter overall survival. IHC analysis of clinical samples further verified the overexpression of INTS7 protein and confirmed the diagnostic value of INTS7 in LUAD, especially for patients at advanced stages with the AUC >0.8. A total of 192 DEGs were identified and DEGs were primarily involved in cell cycle, inflammatory response, and immune response. Moreover, INTS7 expression was negatively correlated with memory B cells, regulatory T cells (Treg), monocytes, resting myeloid dentritic cells and activated mast cells infiltration, and positively correlated with naive B cells, T follicular helper cells (Tfh), activated myeloid dentritic cells and neutrophils infiltration. In addition, patients with high expression of INTS7 showed less expression of immune checkpoints and exhibited less sensitivity to molecular-targeted drugs. CONCLUSION INTS7 is a potential diagnostic biomarker for LUAD. And its expression level may correlate with tumor microenvireoment, immunotherapy responsiveness, and molecular-targeted therapy responsiveness in LUAD.
Collapse
Affiliation(s)
- Xiang Li
- Department of Respiratory and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu 215008, PR China
| | - Yiyong Yao
- Department of Respiratory and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu 215008, PR China
| | - Jinxian Qian
- Department of Respiratory and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu 215008, PR China
| | - Guomin Jin
- Department of Internal Medicine, Guli Hospital of Changshu, 166 Tieqin North Street, Guli Town, Changshu County, Suzhou, Jiangsu 215500, PR China
| | - Gang Zeng
- Department of Respiratory and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu 215008, PR China.
| | - Hongmei Zhao
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing 100000, PR China.
| |
Collapse
|
19
|
Drusbosky LM, Dawar R, Rodriguez E, Ikpeazu CV. Therapeutic strategies in METex14 skipping mutated non-small cell lung cancer. J Hematol Oncol 2021; 14:129. [PMID: 34425853 PMCID: PMC8381548 DOI: 10.1186/s13045-021-01138-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
METex14 skipping mutations occur in about 3–4% of lung adenocarcinoma patients and 1–2% of patients with other lung cancer histology. The MET receptor tyrosine kinase and its ligand hepatocyte growth factor (HGF) are established oncogenic drivers of NSCLC. A mutation that results in loss of exon 14 in the MET gene leads to dysregulation and inappropriate signaling that is associated with increased responsiveness to MET TKIs. Results from GEOMETRY mono-1 and VISION Phase I/II clinical trials demonstrated significant clinical activity in patients treated with the MET Exon 14 skipping mutation inhibitors capmatinib and tepotinib with tolerable toxicity profile. In the GEOMETRY mono-1 trial, capmatinib was especially active in treatment-naïve patients supporting the upfront testing of this oncogenic driver. Tepotinib demonstrated superior activity in the pretreated patients in the VISION trial. Savolitinib is another MET TKI that has shown efficacy in the first- and second-line settings, including patients with aggressive pulmonary sarcomatoid carcinoma. These studies have demonstrated that these TKIs can cross the blood brain barrier and demonstrated some activity toward CNS metastases. MET Exon 14 skipping mutation is detected by NGS-based testing of liquid or tissue biopsies, with preference for RNA-based NGS. The activity of capmatinib and tepotinib is limited by the development of acquired resistance. Current research is focused on strategies to overcome resistance and improve the effectiveness of these agents. Our aim is to review the current status of MET Exon 14 skipping mutation as it pertains NSCLC.
Collapse
Affiliation(s)
| | - Richa Dawar
- Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, 1475 NW 12th Avenue, Miami, FL, 33136, USA
| | - Estelamari Rodriguez
- Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, 1475 NW 12th Avenue, Miami, FL, 33136, USA
| | - Chukwuemeka V Ikpeazu
- University of Miami Sylvester Comprehensive Cancer Center, 8100 SW 10th Street, Ste 3310F, Plantation, FL, 33324, USA. .,Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, 1475 NW 12th Avenue, Miami, FL, 33136, USA.
| |
Collapse
|
20
|
Rebuzzi SE, Zullo L, Rossi G, Grassi M, Murianni V, Tagliamento M, Prelaj A, Coco S, Longo L, Dal Bello MG, Alama A, Dellepiane C, Bennicelli E, Malapelle U, Genova C. Novel Emerging Molecular Targets in Non-Small Cell Lung Cancer. Int J Mol Sci 2021; 22:ijms22052625. [PMID: 33807876 PMCID: PMC7961376 DOI: 10.3390/ijms22052625] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
In the scenario of systemic treatment for advanced non-small cell lung cancer (NSCLC) patients, one of the most relevant breakthroughs is represented by targeted therapies. Throughout the last years, inhibitors of the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-Ros oncogene 1 (ROS1), and V-raf murine sarcoma viral oncogene homolog B (BRAF) have been approved and are currently used in clinical practice. However, other promising molecular drivers are rapidly emerging as therapeutic targets. This review aims to cover the molecular alterations with a potential clinical impact in NSCLC, including amplifications or mutations of the mesenchymal–epithelial transition factor (MET), fusions of rearranged during transfection (RET), rearrangements of the neurotrophic tyrosine kinase (NTRK) genes, mutations of the Kirsten rat sarcoma viral oncogene (KRAS) and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA), as well as amplifications or mutations of human epidermal growth factor receptor 2 (HER2). Additionally, we summarized the current status of targeted agents under investigation for such alterations. This revision of the current literature on emerging molecular targets is needed as the evolving knowledge on novel actionable oncogenic drivers and targeted agents is expected to increase the proportion of patients who will benefit from tailored therapeutic approaches.
Collapse
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.G.); (V.M.)
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy; (M.T.); (C.G.)
- Correspondence:
| | - Lodovica Zullo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Giovanni Rossi
- Medical Oncology Department, Ospedale Padre Antero Micone, 16153 Genoa, Italy;
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via Roma 151, 07100 Sassari, Italy
| | - Massimiliano Grassi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.G.); (V.M.)
| | - Veronica Murianni
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.G.); (V.M.)
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy; (M.T.); (C.G.)
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Arsela Prelaj
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
- Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Luca Longo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Maria Giovanna Dal Bello
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Angela Alama
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Chiara Dellepiane
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Elisa Bennicelli
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.Z.); (S.C.); (L.L.); (M.G.D.B.); (A.A.); (C.D.); (E.B.)
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80138 Naples, Italy;
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy; (M.T.); (C.G.)
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
21
|
de Mello RA, Neves NM, Tadokoro H, Amaral GA, Castelo-Branco P, Zia VADA. New Target Therapies in Advanced Non-Small Cell Lung Cancer: A Review of the Literature and Future Perspectives. J Clin Med 2020; 9:3543. [PMID: 33153004 PMCID: PMC7693900 DOI: 10.3390/jcm9113543] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Lung cancer (LC) is the most common neoplasm worldwide, and 85% of these tumors are classified as non-small cell lung cancer (NSCLC). LC treatment was initially restricted to cytotoxic chemotherapy-platinum compounds associated with 3rd generation cytotoxic agents (paclitaxel, gemcitabine, pemetrexed) and, more recently, with monoclonal antibodies (bevacizumab, ramucirumab). Advancements in treatment are correlated with prolonged overall survival (OS). Current advances are focused on target therapies. Target agents: Anti-epidermal growth factor receptor (EGFR) therapy consists of 1st and 2nd generation tyrosine kinase inhibitors (TKIs such as erlotinib, afatinib). In 60% of cases, resistance to these TKIs occurs due to T790M mutation in EGFR, which is overcome 3rd generation drugs (osimertinib). Anaplastic lymphoma kinase (ALK) is the target for drugs such as crizotinib, alectinib, ceritinib. Programmed death 1 (PD-1) and its ligand serve as targets for immunotherapy agents such as pembrolizumab, nivolumab, atezolizumab. DISCUSSION Challenges in NSCLC treatment include resistance to 3rd generation TKIs, the high cost of ALK inhibitors, and the need for further research on new drugs.
Collapse
Affiliation(s)
- Ramon Andrade de Mello
- Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve (DCBM UALG), 8005-139 Faro, Portugal; (H.T.); (P.C.-B.)
- Division of Medical Oncology, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo 04037-004, SP, Brazil; (N.M.N.); (G.A.A.); (V.A.d.A.Z.)
- Precision Oncology and Health Economics Group (ONCOPRECH), Post-Graduation Program in Medicine, Nine of July University (UNINOVE), São Paulo 04037-004, SP, Brazil
- Division of Oncology, Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal
| | - Nathália Moisés Neves
- Division of Medical Oncology, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo 04037-004, SP, Brazil; (N.M.N.); (G.A.A.); (V.A.d.A.Z.)
| | - Hakaru Tadokoro
- Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve (DCBM UALG), 8005-139 Faro, Portugal; (H.T.); (P.C.-B.)
| | - Giovanna Araújo Amaral
- Division of Medical Oncology, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo 04037-004, SP, Brazil; (N.M.N.); (G.A.A.); (V.A.d.A.Z.)
| | - Pedro Castelo-Branco
- Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve (DCBM UALG), 8005-139 Faro, Portugal; (H.T.); (P.C.-B.)
| | - Victor André de Almeida Zia
- Division of Medical Oncology, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo 04037-004, SP, Brazil; (N.M.N.); (G.A.A.); (V.A.d.A.Z.)
| |
Collapse
|