1
|
Conlon NT, Roche S, Mahdi AF, Browne A, Breen L, Gaubatz J, Meiller J, O'Neill F, O'Driscoll L, Cremona M, Hennessy BT, Eli LD, Crown J, Collins DM. Neratinib plus dasatinib is highly synergistic in HER2-positive breast cancer in vitro and in vivo. Transl Oncol 2024; 49:102073. [PMID: 39191139 PMCID: PMC11396364 DOI: 10.1016/j.tranon.2024.102073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND HER2-targeted therapies have revolutionised the treatment of HER2-positive breast cancer. However, de novo resistance or the emergence of acquired resistance is a persistent clinical problem. Here we report that neratinib, an irreversible pan-HER inhibitor, in combination with the multi-kinase inhibitor dasatinib, currently used to treat certain leukemias, has strong anti-proliferative effects against models of HER2-positive breast cancer that are innately resistant to trastuzumab or have acquired resistance to neratinib. METHODS Neratinib plus dasatinib was examined in a panel of 20 breast cancer cell lines, including HER2-positive, estrogen-receptor-positive, triple negative, and acquired HER2-targeted therapy resistant models. Drug effects on migration and apoptosis induction was evaluated and signaling alterations were determined by reverse phase protein array (RPPA). In vivo efficacy was examined using orthotopically-implanted HCC1954 cells. RESULTS Synergy was observed in cell lines innately resistant to trastuzumab, models with acquired resistance to neratinib, and in triple negative breast cancer cell lines. Further investigation showed that neratinib plus dasatinib induced apoptosis and inhibited cell migration to a greater degree than either drug alone. RPPA revealed that the combination caused suppression of key survival signaling through EGFR, Akt, and MAPK inhibition. In vivo, neratinib plus dasatinib was well tolerated and had a prolonged anti-tumor effect against HCC1954 xenografts. CONCLUSIONS This study provides a strong pre-clinical rationale for the clinical investigation neratinib and dasatinib in HER2+ breast cancer.
Collapse
Affiliation(s)
- Neil T Conlon
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Sandra Roche
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Amira F Mahdi
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Alacoque Browne
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Laura Breen
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Johanna Gaubatz
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Justine Meiller
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Fiona O'Neill
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Science & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mattia Cremona
- Molecular Medicine - Laboratory of Molecular Oncology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bryan T Hennessy
- Molecular Medicine - Laboratory of Molecular Oncology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Lisa D Eli
- Puma Biotechnology, Inc., 10880 Wilshire Boulevard, Suite 2150, Los Angeles, CA, 90024, USA
| | - John Crown
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland; Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Denis M Collins
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland
| |
Collapse
|
2
|
Jiang M, Li Q, Xu B. Spotlight on ideal target antigens and resistance in antibody-drug conjugates: Strategies for competitive advancement. Drug Resist Updat 2024; 75:101086. [PMID: 38677200 DOI: 10.1016/j.drup.2024.101086] [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: 10/24/2023] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a novel and promising approach in targeted therapy, uniting the specificity of antibodies that recognize specific antigens with payloads, all connected by the stable linker. These conjugates combine the best targeted and cytotoxic therapies, offering the killing effect of precisely targeting specific antigens and the potent cell-killing power of small molecule drugs. The targeted approach minimizes the off-target toxicities associated with the payloads and broadens the therapeutic window, enhancing the efficacy and safety profile of cancer treatments. Within precision oncology, ADCs have garnered significant attention as a cutting-edge research area and have been approved to treat a range of malignant tumors. Correspondingly, the issue of resistance to ADCs has gradually come to the fore. Any dysfunction in the steps leading to the ADCs' action within tumor cells can lead to the development of resistance. A deeper understanding of resistance mechanisms may be crucial for developing novel ADCs and exploring combination therapy strategies, which could further enhance the clinical efficacy of ADCs in cancer treatment. This review outlines the brief historical development and mechanism of ADCs and discusses the impact of their key components on the activity of ADCs. Furthermore, it provides a detailed account of the application of ADCs with various target antigens in cancer therapy, the categorization of potential resistance mechanisms, and the current state of combination therapies. Looking forward, breakthroughs in overcoming technical barriers, selecting differentiated target antigens, and enhancing resistance management and combination therapy strategies will broaden the therapeutic indications for ADCs. These progresses are anticipated to advance cancer treatment and yield benefits for patients.
Collapse
Affiliation(s)
- Mingxia Jiang
- 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, China
| | - Qiao Li
- 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, China.
| | - Binghe Xu
- Department of Medical Oncology, State Key Laboratory of Mocelular Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Yang GZ, Wang L, Gao K, Zhu X, Lou LG, Yue JM. Design and Synthesis of Cyclolipopeptide Mimics of Dysoxylactam A and Evaluation of the Reversing Potencies against P-Glycoprotein-Mediated Multidrug Resistance. J Med Chem 2024. [PMID: 38502936 DOI: 10.1021/acs.jmedchem.3c01920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Inspired by the structure of dysoxylactam A (DLA) that has been demonstrated to reverse P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) effectively, 61 structurally simplified cyclolipopeptides were thus designed and synthesized via an effective method, and their reversing P-gp-mediated MDR potentials were evaluated, which provided a series of more potent analogues and allowed us to explore their structure-activity relationship (SAR). Among them, a well-simplified compound, 56, with only two chiral centers that all derived from amino acids dramatically reversed drug resistance in KBV200 cells at 10 μM in combination with vinorelbine (VNR), paclitaxel (PTX), and adriamycin (ADR), respectively, which is more promising than DLA. The mechanism study showed that 56 reversed the MDR of tumor cells by inhibiting the transport function of P-gp rather than reducing its expression. Notably, compound 56 effectively restored the sensitivity of MDR tumors to VNR in vivo at a dosage without obvious toxicity.
Collapse
Affiliation(s)
- Guan-Zhou Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Lei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Kun Gao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xi Zhu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Li-Guang Lou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Jian-Min Yue
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
- Research Units of Discovery of New Drug Lead Molecules, Chinese Academy of Medical Sciences, Shanghai 201203, People's Republic of China
| |
Collapse
|
4
|
Li Z, Metzger Filho O, Viale G, dell'Orto P, Russo L, Goyette MA, Kamat A, Yardley DA, Gupta Abramson V, Arteaga CL, Spring LM, Chiotti K, Halsey C, Waks AG, King TA, Lester SC, Bellon JR, Winer EP, Spellman PT, Krop IE, Polyak K. HER2 heterogeneity and treatment response-associated profiles in HER2-positive breast cancer in the NCT02326974 clinical trial. J Clin Invest 2024; 134:e176454. [PMID: 38300710 PMCID: PMC10977978 DOI: 10.1172/jci176454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/30/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUNDHER2-targeting therapies have great efficacy in HER2-positive breast cancer, but resistance, in part due to HER2 heterogeneity (HET), is a significant clinical challenge. We previously described that in a phase II neoadjuvant trastuzumab emtansine (T-DM1) and pertuzumab (P) clinical trial in early-stage HER2-positive breast cancer, none of the patients with HER2-HET tumors had pathologic complete response (pCR).METHODSTo investigate cellular and molecular differences among tumors according to HER2 heterogeneity and pCR, we performed RNA sequencing and ERBB2 FISH of 285 pretreatment and posttreatment tumors from 129 patients in this T-DM1+P neoadjuvant trial. A subset of cases was also subject to NanoString spatial digital profiling.RESULTSPretreatment tumors from patients with pCR had the highest level of ERBB2 mRNA and ERBB signaling. HER2 heterogeneity was associated with no pCR, basal-like features, and low ERBB2 expression yet high ERBB signaling sustained by activation of downstream pathway components. Residual tumors showed decreased HER2 protein levels and ERBB2 copy number heterogeneity and increased PI3K pathway enrichment and luminal features. HET tumors showed minimal treatment-induced transcriptomic changes compared with non-HET tumors. Immune infiltration correlated with pCR and HER2-HET status.CONCLUSIONResistance mechanisms in HET and non-HET tumors are distinct. HER2-targeting antibodies have limited efficacy in HET tumors. Our results support the stratification of patients based on HET status and the use of agents that target downstream components of the ERBB signaling pathway in patients with HET tumors.TRIAL REGISTRATIONClinicalTrials.gov NCT02326974.FUNDINGThis study was funded by Roche and the National Cancer Institute.
Collapse
Affiliation(s)
- Zheqi Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Otto Metzger Filho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Giuseppe Viale
- Division of Pathology, European Institute of Oncology, IRCCS, Milan, Italy
- University of Milan, School of Medicine, Milan, Italy
| | - Patrizia dell'Orto
- Division of Pathology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Leila Russo
- Division of Pathology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Marie-Anne Goyette
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Avni Kamat
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard University, Cambridge, Massachusetts, USA
| | - Denise A Yardley
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville, Tennessee, USA
| | | | - Carlos L Arteaga
- University of Texas Southwestern, Simmons Comprehensive Cancer Center, Dallas, Texas, USA
| | - Laura M Spring
- Department of Medical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kami Chiotti
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Carol Halsey
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Adrienne G Waks
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Tari A King
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan C Lester
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer R Bellon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric P Winer
- Department of Internal Medicine, Yale Cancer Center, New Haven, Connecticut, USA
| | - Paul T Spellman
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Ian E Krop
- Department of Internal Medicine, Yale Cancer Center, New Haven, Connecticut, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Kook E, Chun KS, Kim DH. Emerging Roles of YES1 in Cancer: The Putative Target in Drug Resistance. Int J Mol Sci 2024; 25:1450. [PMID: 38338729 PMCID: PMC10855972 DOI: 10.3390/ijms25031450] [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: 12/29/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Src family kinases (SFKs) are non-receptor tyrosine kinases that are recognized as proto-oncogenic products. Among SFKs, YES1 is frequently amplified and overexpressed in a variety of human tumors, including lung, breast, ovarian, and skin cancers. YES1 plays a pivotal role in promoting cell proliferation, survival, and invasiveness during tumor development. Recent findings indicate that YES1 expression and activation are associated with resistance to chemotherapeutic drugs and tyrosine kinase inhibitors in human malignancies. YES1 undergoes post-translational modifications, such as lipidation and nitrosylation, which can modulate its catalytic activity, subcellular localization, and binding affinity for substrate proteins. Therefore, we investigated the diverse mechanisms governing YES1 activation and its impact on critical intracellular signal transduction pathways. We emphasized the function of YES1 as a potential mechanism contributing to the anticancer drug resistance emergence.
Collapse
Affiliation(s)
- Eunjin Kook
- Department of Chemistry, Kyonggi University, Suwon 16227, Republic of Korea;
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42691, Republic of Korea;
| | - Do-Hee Kim
- Department of Chemistry, Kyonggi University, Suwon 16227, Republic of Korea;
| |
Collapse
|
6
|
Lapouge M, Meloche S. A renaissance for YES in cancer. Oncogene 2023; 42:3385-3393. [PMID: 37848624 DOI: 10.1038/s41388-023-02860-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
Most of our understanding regarding the involvement of SRC-family tyrosine kinases in cancer has stemmed from studies focused on the prototypical SRC oncogene. However, emerging research has shed light on the important role of YES signaling in oncogenic transformation, tumor growth, metastatic progression, and resistance to various cancer therapies. Clinical evidence indicates that dysregulated expression or activity of YES is a frequent occurrence in human cancers and is associated with unfavorable outcomes. These findings provide a compelling rationale for specifically targeting YES in certain cancer subtypes. Here, we review the crucial role of YES in cancer and discuss the challenges associated with translating preclinical observations into effective YES-targeted therapies.
Collapse
Affiliation(s)
- Marjorie Lapouge
- Institute for Research in Immunology and Cancer, Montreal, QC, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, QC, Canada.
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada.
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
7
|
Leyton JV. The endosomal-lysosomal system in ADC design and cancer therapy. Expert Opin Biol Ther 2023; 23:1067-1076. [PMID: 37978880 DOI: 10.1080/14712598.2023.2285996] [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: 09/05/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION This discourse delves into the intricate connections between the endosomal-lysosomal system and antibody-drug conjugates (ADCs), shedding light on an essential yet less understood dimension of targeted therapy. While ADCs have revolutionized cancer treatment, resistance remains a formidable challenge, often involving diverse and overlapping mechanisms. AREAS COVERED This discourse highlights the roles of various components within the endosomal machinery, including Rab proteins, in ADC resistance development. It also explores how the transferrin-transferrin receptor and epidermal growth factor-epidermal growth factor receptor complexes, known for their roles in recycling and degradation process, respectively, can offer valuable insights for ADC design. Selected strategies to enhance lysosomal targeting are discussed, and potentially offer solutions to improve ADC efficacy. EXPERT OPINION By harnessing these different insights that connect ADCs with the endosomal-lysosomal system, the field may benefit to shape the next-generation of ADC design for increased efficacy and improved patient outcomes.
Collapse
Affiliation(s)
- Jeffrey V Leyton
- School of Pharmaceutical Sciences and Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
Awah CU, Glemaud Y, Levine F, Yang K, Ansary A, Dong F, Ash L, Zhang J, Ogunwobi OO. Destabilized 3'UTR elements therapeutically degrade ERBB2 mRNA in drug-resistant ERBB2+ cancer models. Front Genet 2023; 14:1184600. [PMID: 37359373 PMCID: PMC10287955 DOI: 10.3389/fgene.2023.1184600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Breast, lung, and colorectal cancer resistance to molecular targeted therapy is a major challenge that unfavorably impacts clinical outcomes leading to hundreds of thousands of deaths annually. In ERBB2+ cancers regardless of the tissue of origin, many ERBB2+ cancers are resistant to ERBB2-targeted therapy. We discovered that ERBB2+ cancer cells are enriched with poly U sequences on their 3'UTR which are mRNA-stabilizing sequences. We developed a novel technology, in which we engineered these ERBB2 mRNA-stabilizing sequences to unstable forms that successfully overwrote and outcompeted the endogenous ERBB2 mRNA-encoded message and degraded ERBB2 transcripts which led to the loss of the protein across multiple cancer cell types both in the wildtype and drug-resistance settings in vitro and in vivo, offering a unique safe novel modality to control ERBB2 mRNA and other pervasive oncogenic signals where current targeted therapies fail.
Collapse
Affiliation(s)
- Chidiebere U. Awah
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Yana Glemaud
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
| | - Fayola Levine
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Kiseok Yang
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
| | - Afrin Ansary
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
| | - Fu Dong
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
| | - Leonard Ash
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
| | - Junfei Zhang
- Department of Pathology and Cell Biology, Department of System Biology, Columbia University Medical Center, New York, NY, United States
| | - Olorunseun O. Ogunwobi
- Department of Biological Sciences, Hunter College of The City University of New York, New York City, NY, United States
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
9
|
Zhou H, Sun D, Tao J, Xu M, Zhang X, Hou H. Role of YES1 signaling in tumor therapy resistance. CANCER INNOVATION 2023; 2:210-218. [PMID: 38089407 PMCID: PMC10686156 DOI: 10.1002/cai2.51] [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: 09/21/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 10/15/2024]
Abstract
YES proto-oncogene 1 (YES1) is an SRC family kinase (SFK) that plays a key role in cancer cell proliferation, adhesion, invasion, survival, and angiogenesis during tumorigenesis and tumor development. Reports suggest that YES1 amplification is associated with resistance to chemotherapeutic drugs and tyrosine kinase inhibitors (TKIs) in human malignancies. However, the mechanisms of drug resistance have not been fully elucidated. In this article, we review the literature on YES1 and discuss the implications of YES1 signaling for targeted therapy and chemotherapy resistance in malignancies. Moreover, recent advances in targeted therapy for YES1-amplified malignancies are summarized. Finally, we conclude that targeting YES1 may reverse drug resistance and serve as a valuable tumor treatment strategy.
Collapse
Affiliation(s)
- Hai Zhou
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Dantong Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junyan Tao
- Qingdao Sixth People's HospitalQingdaoShandongChina
| | - Mingjin Xu
- Department of Radiation OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Xiaochun Zhang
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Helei Hou
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
10
|
Chen Y, Xu Y, Shao Z, Yu K. Resistance to antibody-drug conjugates in breast cancer: mechanisms and solutions. Cancer Commun (Lond) 2023; 43:297-337. [PMID: 36357174 PMCID: PMC10009672 DOI: 10.1002/cac2.12387] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/13/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a rapidly developing therapeutic approach in cancer treatment that has shown remarkable activity in breast cancer. Currently, there are two ADCs approved for the treatment of human epidermal growth factor receptor 2-positive breast cancer, one for triple-negative breast cancer, and multiple investigational ADCs in clinical trials. However, drug resistance has been noticed in clinical use, especially in trastuzumab emtansine. Here, the mechanisms of ADC resistance are summarized into four categories: antibody-mediated resistance, impaired drug trafficking, disrupted lysosomal function, and payload-related resistance. To overcome or prevent resistance to ADCs, innovative development strategies and combination therapy options are being investigated. Analyzing predictive biomarkers for optimal therapy selection may also help to prevent drug resistance.
Collapse
Affiliation(s)
- Yu‐Fei Chen
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Ying‐ying Xu
- Department of Breast SurgeryFirst Affiliated Hospital of China Medical UniversityShenyangLiaoning110001P. R. China
| | - Zhi‐Ming Shao
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Ke‐Da Yu
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Shanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| |
Collapse
|
11
|
Garmendia I, Redin E, Montuenga LM, Calvo A. YES1: a novel therapeutic target and biomarker in cancer. Mol Cancer Ther 2022; 21:1371-1380. [PMID: 35732509 DOI: 10.1158/1535-7163.mct-21-0958] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022]
Abstract
YES1 is a non-receptor tyrosine kinase that belongs to the SRC family of kinases (SFKs) and controls multiple cancer signaling pathways. YES1 is amplified and overexpressed in many tumor types, where it promotes cell proliferation, survival and invasiveness. Therefore, YES1 has been proposed as an emerging target in solid tumors. In addition, studies have shown that YES1 is a prognostic biomarker and a predictor of dasatinib activity. Several SFKs-targeting drugs have been developed and some of them have reached clinical trials. However, these drugs have encountered challenges to their utilization in the clinical practice in unselected patients due to toxicity and lack of efficacy. In the case of YES1, novel specific inhibitors have been developed and tested in preclinical models, with impressive antitumor effects. In this review, we summarize the structure and activation of YES1 and describe its role in cancer as a target and prognostic and companion biomarker. We also address the efficacy of SFKs inhibitors that are currently in clinical trials, highlighting the main hindrances for their clinical use. Current available information strongly suggests that inhibiting YES1 in tumors with high expression of this protein is a promising strategy against cancer.
Collapse
Affiliation(s)
- Irati Garmendia
- INSERM UMRS1138. Centre de Recherche des Cordeliers, Paris, France
| | | | - Luis M Montuenga
- CIMA and Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Alfonso Calvo
- Center for Applied Medical Research (CIMA), Pamplona, Spain
| |
Collapse
|
12
|
Fujihara M, Shien T, Shien K, Suzawa K, Takeda T, Zhu Y, Mamori T, Otani Y, Yoshioka R, Uno M, Suzuki Y, Abe Y, Hatono M, Tsukioki T, Takahashi Y, Kochi M, Iwamoto T, Taira N, Doihara H, Toyooka S. YES1 as a Therapeutic Target for HER2-Positive Breast Cancer after Trastuzumab and Trastuzumab-Emtansine (T-DM1) Resistance Development. Int J Mol Sci 2021; 22:ijms222312809. [PMID: 34884609 PMCID: PMC8657782 DOI: 10.3390/ijms222312809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 01/16/2023] Open
Abstract
Trastuzumab-emtansine (T-DM1) is a therapeutic agent molecularly targeting human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC), and it is especially effective for MBC with resistance to trastuzumab. Although several reports have described T-DM1 resistance, few have examined the mechanism underlying T-DM1 resistance after the development of acquired resistance to trastuzumab. We previously reported that YES1, a member of the Src family, plays an important role in acquired resistance to trastuzumab in HER2-amplified breast cancer cells. We newly established a trastuzumab/T-DM1-dual-resistant cell line and analyzed the resistance mechanisms in this cell line. At first, the T-DM1 effectively inhibited the YES1-amplified trastuzumab-resistant cell line, but resistance to T-DM1 gradually developed. YES1 amplification was further enhanced after acquired resistance to T-DM1 became apparent, and the knockdown of the YES1 or the administration of the Src inhibitor dasatinib restored sensitivity to T-DM1. Our results indicate that YES1 is also strongly associated with T-DM1 resistance after the development of acquired resistance to trastuzumab, and the continuous inhibition of YES1 is important for overcoming resistance to T-DM1.
Collapse
Affiliation(s)
- Miwa Fujihara
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Tadahiko Shien
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
- Correspondence: ; Tel.: +81-86-235-7265
| | - Kazuhiko Shien
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Ken Suzawa
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Tatsuaki Takeda
- Departments of Pharmacy, Okayama University Hospital, Okayama 700-8558, Japan;
| | - Yidan Zhu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Tomoka Mamori
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Yusuke Otani
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Ryo Yoshioka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Maya Uno
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Yoko Suzuki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Yuko Abe
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Minami Hatono
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Takahiro Tsukioki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Yuko Takahashi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Mariko Kochi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Takayuki Iwamoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Naruto Taira
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Hiroyoshi Doihara
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (M.F.); (K.S.); (K.S.); (Y.Z.); (T.M.); (Y.O.); (R.Y.); (M.U.); (Y.S.); (Y.A.); (M.H.); (T.T.); (Y.T.); (M.K.); (T.I.); (N.T.); (H.D.); (S.T.)
| |
Collapse
|
13
|
Marin BM, Porath KA, Jain S, Kim M, Conage-Pough JE, Oh JH, Miller CL, Talele S, Kitange GJ, Tian S, Burgenske DM, Mladek AC, Gupta SK, Decker PA, McMinn MH, Stopka SA, Regan MS, He L, Carlson BL, Bakken K, Burns TC, Parney IF, Giannini C, Agar NYR, Eckel-Passow JE, Cochran JR, Elmquist WF, Vaubel RA, White FM, Sarkaria JN. Heterogeneous delivery across the blood-brain barrier limits the efficacy of an EGFR-targeting antibody drug conjugate in glioblastoma. Neuro Oncol 2021; 23:2042-2053. [PMID: 34050676 PMCID: PMC8643472 DOI: 10.1093/neuonc/noab133] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Antibody drug conjugates (ADCs) targeting the epidermal growth factor receptor (EGFR), such as depatuxizumab mafodotin (Depatux-M), is a promising therapeutic strategy for glioblastoma (GBM) but recent clinical trials did not demonstrate a survival benefit. Understanding the mechanisms of failure for this promising strategy is critically important. METHODS PDX models were employed to study efficacy of systemic vs intracranial delivery of Depatux-M. Immunofluorescence and MALDI-MSI were performed to detect drug levels in the brain. EGFR levels and compensatory pathways were studied using quantitative flow cytometry, Western blots, RNAseq, FISH, and phosphoproteomics. RESULTS Systemic delivery of Depatux-M was highly effective in nine of 10 EGFR-amplified heterotopic PDXs with survival extending beyond one year in eight PDXs. Acquired resistance in two PDXs (GBM12 and GBM46) was driven by suppression of EGFR expression or emergence of a novel short-variant of EGFR lacking the epitope for the Depatux-M antibody. In contrast to the profound benefit observed in heterotopic tumors, only two of seven intrinsically sensitive PDXs were responsive to Depatux-M as intracranial tumors. Poor efficacy in orthotopic PDXs was associated with limited and heterogeneous distribution of Depatux-M into tumor tissues, and artificial disruption of the BBB or bypass of the BBB by direct intracranial injection of Depatux-M into orthotopic tumors markedly enhanced the efficacy of drug treatment. CONCLUSIONS Despite profound intrinsic sensitivity to Depatux-M, limited drug delivery into brain tumor may have been a key contributor to lack of efficacy in recently failed clinical trials.
Collapse
Affiliation(s)
- Bianca-Maria Marin
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kendra A Porath
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonia Jain
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Minjee Kim
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jason E Conage-Pough
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ju-Hee Oh
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Caitlyn L Miller
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Surabhi Talele
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gaspar J Kitange
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shulan Tian
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shiv K Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul A Decker
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Madison H McMinn
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael S Regan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lihong He
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Katrina Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Terence C Burns
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Caterina Giannini
- Department of Laboratory Medicine and Pathology; Mayo Clinic, Rochester, Minnesota, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - William F Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rachael A Vaubel
- Department of Laboratory Medicine and Pathology; Mayo Clinic, Rochester, Minnesota, USA
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA,Corresponding Author: Jann N. Sarkaria, MD, Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Mayo Clinic, Rochester, MN 55902, USA ()
| |
Collapse
|