1
|
Kim HS, Kim Y, Lee HS. Clinicopathologic Characteristics of Trop Family Proteins (Trop-2 and EpCAM) in Gastric Carcinoma. J Gastric Cancer 2024; 24:391-405. [PMID: 39375055 PMCID: PMC11471318 DOI: 10.5230/jgc.2024.24.e32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 10/09/2024] Open
Abstract
PURPOSE Trop family proteins, including epithelial cell adhesion molecule (EpCAM) and Trop-2, have garnered attention as potential therapeutic and diagnostic targets for various malignancies. This study aimed to elucidate the clinicopathological significance of these proteins in gastric carcinoma (GC) and to reinforce their potential as biomarkers for patient stratification in targeted therapies. MATERIALS AND METHODS Immunohistochemical (IHC) analyses of EpCAM and Trop-2 were performed on GC and precancerous lesions, following rigorous orthogonal validation of the antibodies to ensure specificity and sensitivity. RESULTS Strong membranous staining (3+) for Trop-2 was observed in 49.3% of the GC cases, whereas EpCAM was strongly expressed in almost all cases (93.2%), indicating its widespread expression in GC. A high Trop-2 expression level, characterized by an elevated H-score, was significantly associated with intestinal type by Lauren classification, gastric mucin type, presence of lymph node metastasis, human epidermal growth factor receptor 2-positivity, and Epstein-Barr virus (EBV)-positivity. Patients with a high Trop-2 expression level exhibited poorer survival outcomes on univariate and multivariate analyses. High EpCAM expression levels were prevalent in differentiated histologic type, microsatellite instability-high, and EBV-negative cancer, and were correlated with high densities of CD3 and CD8 T cells and elevated combined positive score for programmed death-ligand 1. CONCLUSIONS These results highlight the differential expression of Trop-2 and EpCAM and their prognostic implications in GC. The use of meticulously validated antibodies ensured the reliability of our IHC data, thereby offering a robust foundation for future therapeutic strategies targeting Trop family members in GC.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Pathology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Younghoon Kim
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
2
|
Hu Y, Zhu Y, Qi D, Tang C, Zhang W. Trop2-targeted therapy in breast cancer. Biomark Res 2024; 12:82. [PMID: 39135109 PMCID: PMC11321197 DOI: 10.1186/s40364-024-00633-6] [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: 05/07/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Human trophoblastic cell surface antigen 2 (Trop2) is a glycoprotein, a cellular marker of trophoblastic and stem cells, and a calcium signaling transducer involved in several signaling pathways, leading to the proliferation, invasion, and metastasis of tumors. It is expressed at a low level in normal epithelial cells, but at a high level in many tumors, making it an ideal target for cancer therapy. According to previous literature, Trop2 is broadly expressed in all breast cancer subtypes, especially in triple negative breast cancer (TNBC). Several clinical trials have demonstrated the effectiveness of Trop2-targeted therapy in breast cancer. Sacituzumab govitecan (SG) is a Trop2-targeted antibody-drug conjugate (ADC) that has been approved for the treatment of metastatic TNBC and hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-) breast cancer. This article reviews the structure and function of Trop2, several major Trop2-targeted ADCs, other appealing novel Trop2-targeted agents and relevant clinical trials to provide a landscape of how Trop2-targeted treatments will develop in the future.
Collapse
Affiliation(s)
- Yixuan Hu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yinxing Zhu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
- Department of Radiation Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Dan Qi
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Cuiju Tang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
3
|
Somboonpatarakun C, Phanthaphol N, Suwanchiwasiri K, Ramwarungkura B, Yuti P, Poungvarin N, Thuwajit P, Junking M, Yenchitsomanus PT. Cytotoxicity of fourth-generation anti-Trop2 CAR-T cells against breast cancer. Int Immunopharmacol 2024; 129:111631. [PMID: 38359664 DOI: 10.1016/j.intimp.2024.111631] [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: 09/20/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024]
Abstract
The treatment of breast cancer (BC) remains a formidable challenge due to the emergence of drug resistance, necessitating the exploration of innovative strategies. Chimeric antigen receptor (CAR)-T cell therapy, a groundbreaking approach in hematologic malignancies, is actively under investigation for its potential application in solid tumors, including BC. Trophoblast cell surface antigen 2 (Trop2) has emerged as a promising immunotherapeutic target in various cancers and is notably overexpressed in BC. To enhance therapeutic efficacy in BC, a fourth-generation CAR (CAR4) construct was developed. This CAR4 design incorporates an anti-Trop2 single-chain variable fragment (scFv) fused with three costimulatory domains -CD28/4-1BB/CD27, and CD3ζ. Comparative analysis with the conventional second-generation CAR (CAR2; 28ζ) revealed that anti-Trop2 CAR4 T cells exhibited heightened cytotoxicity and interferon-gamma (IFN-γ) production against Trop2-expressing MCF-7 cells. Notably, anti-Trop2 CAR4-T cells demonstrated superior long-term cytotoxic functionality and proliferative capacity. Crucially, anti-Trop2 CAR4-T cells displayed specific cytotoxicity against Trop2-positive BC cells (MDA-MB-231, HCC70, and MCF-7) in both two-dimensional (2D) and three-dimensional (3D) culture systems. Following antigen-specific killing, these cells markedly secreted interleukin-2 (IL-2), tumor necrosis factor-alpha (TNF-α), IFN-γ, and Granzyme B compared to non-transduced T cells. This study highlights the therapeutic potential of anti-Trop2 CAR4-T cells in adoptive T cell therapy for BC, offering significant promise for the advancement of BC treatment strategies.
Collapse
Affiliation(s)
- Chalermchai Somboonpatarakun
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nattaporn Phanthaphol
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Kwanpirom Suwanchiwasiri
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Graduate Program in Molecular Medicine, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok 10700, Thailand
| | - Boonyanuch Ramwarungkura
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Graduate Program in Biomedical Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pornpimon Yuti
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Naravat Poungvarin
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
4
|
Sawada A, Ohira M, Hatanaka KC, Matsui H, Ichikawa N, Yoshida T, Fukai M, Matsuno Y, Homma S, Hatanaka Y, Taketomi A. Expression Analysis of Early Metastatic Seeding of Colorectal Cancer. Ann Surg Oncol 2024; 31:2101-2113. [PMID: 38063988 DOI: 10.1245/s10434-023-14714-7] [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: 05/30/2023] [Accepted: 11/20/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Distant metastasis is the leading cause of death in patients with colorectal cancer (CRC). Tumor dissemination for metastasis formation occurs in advanced cancers and also during early stages of tumorigenesis. Here, we investigated the genes involved in early metastatic seeding of CRC using gene expression analysis. PATIENTS AND METHODS We performed a cDNA microarray using specimens resected from stages I-II CRC with and without metachronous metastatic recurrence. For the candidate genes, we immunohistochemically validated protein expression using a tissue microarray of stages I-III CRC. RESULTS The expression of TROP2, VWCE, and BMP7 was upregulated in the recurrence group rather than in the non-recurrence group. Protein expression analysis revealed significant association of these genes with distant metastatic recurrence. The specimens with high expression of BMP7 showed worse recurrence-free survival (RFS; p = 0.02). Those with high expression of TROP2 and VWCE showed worse overall survival (OS) and RFS (TROP2: p = 0.01 and p = 0.03; VWCE: p < 0.05 and p < 0.001, respectively). In the multivariate analysis, high expression of VWCE and BMP7 was an independent predictor of recurrence [VWCE: hazard ratio (HR) 3.41, p < 0.001; BMP7: HR 2.93, p = 0.005]. In contrast, TROP2 was an independent prognostic factor for OS (HR 4.58, p = 0.03). CONCLUSIONS Gene expression analysis revealed that TROP2, VWCE, and BMP7 were involved in early metastatic seeding. The high expression of these genes may warrant careful surveillance or adjuvant therapy, even in stages I-II CRC cases.
Collapse
Affiliation(s)
- Akifumi Sawada
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masafumi Ohira
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Hiroki Matsui
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Nobuki Ichikawa
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tadashi Yoshida
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Moto Fukai
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Shigenori Homma
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
5
|
Nelson BE, Meric-Bernstam F. Leveraging TROP2 Antibody-Drug Conjugates in Solid Tumors. Annu Rev Med 2024; 75:31-48. [PMID: 37758237 DOI: 10.1146/annurev-med-071322-065903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Antibody-drug conjugates (ADCs) have become the cornerstone of effective therapeutics in solid and hematological malignancies by harnessing potent cytotoxic payloads with targeted tumoricidal delivery. Since the monumental shift occurred with HER2-targeted ADCs, the discovery of the TROP2 antigen has revolutionized the landscape of ADC development. Moving beyond the traditional ADC design, multiple novel ADCs have successfully shaped and improved survival outcomes in patients with various tumor histologies. Here we review and contrast the clinical impact of the well-known TROP2 ADCs currently in clinical use. We also shed light on upcoming investigational TROP2 ADCs showing promise with novel ADC platforms.
Collapse
Affiliation(s)
- Blessie Elizabeth Nelson
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA;
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA;
| |
Collapse
|
6
|
Huang W, Liang C, Zhang Y, Zhang D, An S, Wu Q, Li J, Zhao H, Wang C, Cui J, Bao Z, Huang G, Wei W, Liu J. ImmunoPET imaging of Trop2 expression in solid tumors with nanobody tracers. Eur J Nucl Med Mol Imaging 2024; 51:380-394. [PMID: 37792026 DOI: 10.1007/s00259-023-06454-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023]
Abstract
PURPOSE The high expression of the transmembrane glycoprotein trophoblast cell-surface antigen 2 (Trop2) was strongly associated with the progression of solid tumors, including pancreatic and gastric cancers. Our study aimed to construct Trop2-specific immuno-positron emission tomography (immunoPET) probes and assess the diagnostic abilities in preclinical pancreatic and gastric cancer models. METHODS The expression of Trop2 in pancreatic cancer was determined by single-cell sequencing and immunohistochemistry on tissue microarray (TMA). Flow cytometry was used to screen the expression of Trop2 in pancreatic cancer cell lines. Two nanobodies (i.e., RTD98 and RTD01) targeting Trop2 were developed and labeled with gallium-68 (68Ga, T1/2 = 1.1 h) to construct immunoPET imaging probes. The agents were researched in cell-derived pancreatic and patient-derived gastric cancer models expressing varying Trop2. RESULTS Single-cell sequencing results showed high expression of Trop2 in pancreatic ductal cells as well as acinar cells and immunohistochemical staining of TMA from pancreatic cancers showed significantly higher expression of Trop2 in cancerous than in paracancerous tissues. ImmunoPET utilizing [68Ga]Ga-NOTA-RTD98 could clearly delineate subcutaneous tumors, both in cell-derived pancreatic cancer models and patient-derived gastric cancer models, superior to imaging using [18F]-FDG or a non-specific probe [68Ga]Ga-NOTA-RTD161. Another probe with improved pharmacokinetics targeting Trop2, [68Ga]Ga-NOTA-RTD01, was further prepared and showed advantageous diagnostic capabilities in preclinical pancreatic cancer models. CONCLUSION In the work, we reported two nanobody tracers targeting human Trop2 which may facilitate better use of Trop2-targeted therapeutics by noninvasively displaying expression dynamics of the target.
Collapse
Affiliation(s)
- Wei Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Chenyi Liang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Di Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Shuxian An
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Jiajin Li
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Haitao Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Cheng Wang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Jiujie Cui
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Zhouzhou Bao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
- Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China.
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Rd, Shanghai, 200127, China.
| |
Collapse
|
7
|
Koltai T, Fliegel L. The Relationship between Trop-2, Chemotherapeutic Drugs, and Chemoresistance. Int J Mol Sci 2023; 25:87. [PMID: 38203255 PMCID: PMC10779383 DOI: 10.3390/ijms25010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/08/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
Trop-2 is a highly conserved one-pass transmembrane mammalian glycoprotein that is normally expressed in tissues such as the lung, intestines, and kidney during embryonic development. It is overexpressed in many epithelial cancers but is absent in non-epithelial tumors. Trop-2 is an intracellular calcium signal transducer that participates in the promotion of cell proliferation, migration, invasion, metastasis, and probably stemness. It also has some tumor suppressor effects. The pro-tumoral actions have been thoroughly investigated and reported. However, Trop-2's activity in chemoresistance is less well known. We review a possible relationship between Trop-2, chemotherapy, and chemoresistance. We conclude that there is a clear role for Trop-2 in some specific chemoresistance events. On the other hand, there is no clear evidence for its participation in multidrug resistance through direct drug transport. The development of antibody conjugate drugs (ACD) centered on anti-Trop-2 monoclonal antibodies opened the gates for the treatment of some tumors resistant to classic chemotherapies. Advanced urothelial tumors and breast cancer were among the first malignancies for which these ACDs have been employed. However, there is a wide group of other tumors that may benefit from anti-Trop-2 therapy as soon as clinical trials are completed.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires 2199, Argentina;
| | - Larry Fliegel
- Department of Biochemistry, Faculty of Medicine, University of Alberta, 347 Medical Science Bldg., Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
8
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
9
|
He K, Wang Z, Luo M, Li B, Ding N, Li L, He B, Wang H, Cao J, Huang C, Yang J, Chen HN. Metastasis organotropism in colorectal cancer: advancing toward innovative therapies. J Transl Med 2023; 21:612. [PMID: 37689664 PMCID: PMC10493031 DOI: 10.1186/s12967-023-04460-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/19/2023] [Indexed: 09/11/2023] Open
Abstract
Distant metastasis remains a leading cause of mortality among patients with colorectal cancer (CRC). Organotropism, referring to the propensity of metastasis to target specific organs, is a well-documented phenomenon in CRC, with the liver, lungs, and peritoneum being preferred sites. Prior to establishing premetastatic niches within host organs, CRC cells secrete substances that promote metastatic organotropism. Given the pivotal role of organotropism in CRC metastasis, a comprehensive understanding of its molecular underpinnings is crucial for biomarker-based diagnosis, innovative treatment development, and ultimately, improved patient outcomes. In this review, we focus on metabolic reprogramming, tumor-derived exosomes, the immune system, and cancer cell-organ interactions to outline the molecular mechanisms of CRC organotropic metastasis. Furthermore, we consider the prospect of targeting metastatic organotropism for CRC therapy.
Collapse
Affiliation(s)
- Kai He
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhihan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Maochao Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Ning Ding
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lei Li
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Bo He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Han Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiangjun Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Canhua Huang
- School of Basic Medical Sciences and State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jun Yang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Hai-Ning Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
- Department of General Surgery, State Key Laboratory of Biotherapy and Cancer Center, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
10
|
Iwamoto S, Mori Y, Yamashita T, Ojima K, Akita K, Togano S, Kushiyama S, Yashiro M, Yatera Y, Yamaguchi T, Komiyama A, Sago Y, Itano N, Nakada H. Trophoblast cell surface antigen-2 phosphorylation triggered by binding of galectin-3 drives metastasis through down-regulation of E-cadherin. J Biol Chem 2023; 299:104971. [PMID: 37380081 PMCID: PMC10392139 DOI: 10.1016/j.jbc.2023.104971] [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: 10/11/2022] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
The expression of trophoblast cell surface antigen-2 (Trop-2) is enhanced in many tumor tissues and is correlated with increased malignancy and poor survival of patients with cancer. Previously, we demonstrated that the Ser-322 residue of Trop-2 is phosphorylated by protein kinase Cα (PKCα) and PKCδ. Here, we demonstrate that phosphomimetic Trop-2 expressing cells have markedly decreased E-cadherin mRNA and protein levels. Consistently, mRNA and protein of the E-cadherin-repressing transcription factors zinc finger E-Box binding homeobox 1 (ZEB1) were elevated, suggesting transcriptional regulation of E-cadherin expression. The binding of galectin-3 to Trop-2 enhanced the phosphorylation and subsequent cleavage of Trop-2, followed by intracellular signaling by the resultant C-terminal fragment. Binding of β-catenin/transcription factor 4 (TCF4) along with the C-terminal fragment of Trop-2 to the ZEB1 promoter upregulated ZEB1 expression. Of note, siRNA-mediated knockdown of β-catenin and TCF4 increased the expression of E-cadherin through ZEB1 downregulation. Knockdown of Trop-2 in MCF-7 cells and DU145 cells resulted in downregulation of ZEB1 and subsequent upregulation of E-cadherin. Furthermore, wild-type and phosphomimetic Trop-2 but not phosphorylation-blocked Trop-2 were detected in the liver and/or lung of some nude mice bearing primary tumors inoculated intraperitoneally or subcutaneously with wild-type or mutated Trop-2 expressing cells, suggesting that Trop-2 phosphorylation, plays an important role in tumor cell mobility in vivo, too. Together with our previous finding of Trop-2 dependent regulation of claudin-7, we suggest that the Trop-2-mediated cascade involves concurrent derangement of both tight and adherence junctions, which may drive metastasis of epithelial tumor cells.
Collapse
Affiliation(s)
- Shungo Iwamoto
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Yugo Mori
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Tomoko Yamashita
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Kazuki Ojima
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Kaoru Akita
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Shingo Togano
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Shuhei Kushiyama
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yuki Yatera
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Tomoko Yamaguchi
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Akane Komiyama
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Yuki Sago
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Naoki Itano
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Hiroshi Nakada
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan.
| |
Collapse
|
11
|
Poorebrahim M, Quiros-Fernandez I, Marmé F, Burdach SE, Cid-Arregui A. A costimulatory chimeric antigen receptor targeting TROP2 enhances the cytotoxicity of NK cells expressing a T cell receptor reactive to human papillomavirus type 16 E7. Cancer Lett 2023; 566:216242. [PMID: 37217069 DOI: 10.1016/j.canlet.2023.216242] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/11/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023]
Abstract
Immune cells modified to express a tumor-reactive T cell receptor (TCR) have shown limited efficacy as stand-alone therapy against solid tumors. Genital and oropharyngeal carcinomas induced by human papillomavirus (HPV) type 16 express constitutively its E6 and E7 oncoproteins, which makes them convenient targets for adoptive cell immunotherapy. However, viral antigen presentation by tumor cells is low and limits the anti-tumor efficacy of CD8+ T cells. To enhance the functionality of immune effector cells, we have devised a strategy combining a costimulatory chimeric antigen receptor (CAR) with a TCR. We used a clinically tested TCR specific to E7 (E7-TCR) of HPV16 and a newly constructed CAR targeting the trophoblast cell surface antigen 2 (TROP2), which carried the intracellular costimulatory domains CD28 and 4-1BB, but was devoid of the CD3ζ domain. Flow cytometry analyses showed a notable upregulation of activation markers and of cytolytic molecule release by NK-92 cells genetically engineered to express CD3, CD8 and both E7-TCR and TROP2-CAR, after co-incubation with HPV16+ cervical cancer cells. Furthermore, the E7-TCR/TROP2-CAR NK-92 cells demonstrated enhanced antigen-specific activation and augmented cytotoxicity against tumor cells compared with NK-92 cells expressing the E7-TCR alone. A costimulatory TROP2-CAR can synergistically cooperate with the E7-TCR in NK cells thereby enhancing their signaling strength and antigen-specific cytotoxicity. This approach might improve the outcome of adoptive cell immunotherapies for HPV16+ cancer patients that are currently under investigation.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany; Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, Canada
| | - Isaac Quiros-Fernandez
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany; Research Center on Tropical Diseases (CIET)/Research Center on Surgery and Cancer (CICICA), Faculty of Microbiology, Universidad de Costa Rica, San Jose, Costa Rica
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Eg Burdach
- Translational Pediatric Cancer Research Action - Institute of Pathology, TUM School of Medicine - Technical University of Munich, Trogerstr. 22 - RG - 540.0.11, D-81675, München, Germany; Department of Molecular Oncology, British Columbia Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Angel Cid-Arregui
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
12
|
Qiu S, Zhang J, Wang Z, Lan H, Hou J, Zhang N, Wang X, Lu H. Targeting Trop-2 in cancer: Recent research progress and clinical application. Biochim Biophys Acta Rev Cancer 2023; 1878:188902. [PMID: 37121444 DOI: 10.1016/j.bbcan.2023.188902] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/11/2023] [Accepted: 04/26/2023] [Indexed: 05/02/2023]
Abstract
The development of new antitumor drugs depends mainly upon targeting tumor cells precisely. Trophoblast surface antigen 2 (Trop-2) is a type I transmembrane glycoprotein involved in Ca2+ signaling in tumor cells. It is highly expressed in various tumor tissues than in normal tissues and represents a novel and promising molecular target for caner targeted therapy. Up to now, the mechanisms and functions associated with Trop-2 have been extensively studied in a variety of solid tumors. According to these findings, Trop-2 plays an important role in cell proliferation, apoptosis, cell adhesion, epithelial-mesenchymal transition, as well as tumorigenesis and tumor progression. In addition, Trop-2 related drugs are also being developed widely. There are a number of Trop-2 related ADC drugs that have demonstrated potent antitumor activity and are currently been studied, such as Sacituzumab Govitecan (SG) and Datopotamab Deruxtecan (Dato-Dxd). In this study, we reviewed the progress of Trop-2 research in solid tumors. We also sorted out the composition and rationale of Trop-2 related drugs and summarized the related clinical trials. Finally, we discussed the current status of Trop-2 research and expanded our perspectives on its future research directions. Importantly, we found that Trop-2 targeted ADCs have great potential for combination with other antitumor therapies. Trop-2 targeted ADCs can reprogramme tumor microenvironment through multiple signaling pathways, ultimately activating antitumor immunity.
Collapse
Affiliation(s)
- Shuying Qiu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Jianping Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China; Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Zhuo Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hui Lan
- Department of Medical Oncology, Affiliated Lishui Hospital of Zhejiang University/Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Jili Hou
- Department of Medical Oncology, Zhuji People's Hospital of Zhejiang Province, Zhuji, China
| | - Nan Zhang
- Department of Medical Oncology, China Coast Guard Hospital of the People's Armed Police Force, Jiaxing, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| | - Haiqi Lu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China.
| |
Collapse
|
13
|
Lombardi P, Filetti M, Falcone R, Altamura V, Paroni Sterbini F, Bria E, Fabi A, Giannarelli D, Scambia G, Daniele G. Overview of Trop-2 in Cancer: From Pre-Clinical Studies to Future Directions in Clinical Settings. Cancers (Basel) 2023; 15:1744. [PMID: 36980630 PMCID: PMC10046386 DOI: 10.3390/cancers15061744] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Trophoblast cell surface antigen-2 (Trop-2) is a glycoprotein that was first described as a membrane marker of trophoblast cells and was associated with regenerative abilities. Trop-2 overexpression was also described in several tumour types. Nevertheless, the therapeutic potential of Trop-2 was widely recognized and clinical studies with drug-antibody conjugates have been initiated in various cancer types. Recently, these efforts have been rewarded with the approval of sacituzumab govitecan from both the Food and Drug Administration (FDA) and European Medicines Agency (EMA), for metastatic triple-negative breast cancer patients. In our work, we briefly summarize the various characteristics of cancer cells overexpressing Trop-2, the pre-clinical activities of specific inhibitors, and the role of anti-Trop-2 therapy in current clinical practice. We also review the ongoing clinical trials to provide a snapshot of the future developments of these therapies.
Collapse
Affiliation(s)
- Pasquale Lombardi
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Filetti
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Rosa Falcone
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Altamura
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Universitá Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alessandra Fabi
- Precision Medicine in Senology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Diana Giannarelli
- Facility of Epidemiology and Biostatistics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Scambia
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Gennaro Daniele
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
14
|
Sakach E, Sacks R, Kalinsky K. Trop-2 as a Therapeutic Target in Breast Cancer. Cancers (Basel) 2022; 14:5936. [PMID: 36497418 PMCID: PMC9735829 DOI: 10.3390/cancers14235936] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
The emergence of Trop-2 as a therapeutic target has given rise to new treatment paradigms for the treatment of patients with advanced and metastatic breast cancer. Trop-2 is most highly expressed in triple negative breast cancer (TNBC), but the receptor is found across all breast cancer subtypes. With sacituzumab govitecan, the first FDA-approved, Trop-2 inhibitor, providing a survival benefit in patients with both metastatic TNBC and hormone receptor positive breast cancer, additional Trop-2 directed therapies are under investigation. Ongoing studies of combination regimens with immunotherapy, PARP inhibitors, and other targeted agents aim to further harness the effect of Trop-2 inhibition. Current investigations are also underway in the neoadjuvant and adjuvant setting to evaluate the therapeutic benefit of Trop-2 inhibition in patients with early stage disease. This review highlights the significant impact the discovery Trop-2 has had on our patients with heavily pretreated breast cancer, for whom few treatment options exist, and the future direction of novel Trop-2 targeted therapies.
Collapse
Affiliation(s)
- Elizabeth Sakach
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
15
|
Liu X, Deng J, Yuan Y, Chen W, Sun W, Wang Y, Huang H, Liang B, Ming T, Wen J, Huang B, Xing D. Advances in Trop2-targeted therapy: Novel agents and opportunities beyond breast cancer. Pharmacol Ther 2022; 239:108296. [PMID: 36208791 DOI: 10.1016/j.pharmthera.2022.108296] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022]
Abstract
Trop2 is a transmembrane glycoprotein and calcium signal transducer with limited expression in normal human tissues. It is consistently overexpressed in a variety of malignant tumors and participates in several oncogenic signaling pathways that lead to tumor development, invasion, and metastasis. As a result, Trop2 has become an attractive therapeutic target in cancer treatment. The anti-Trop2 antibody-drug conjugate (Trodelvy™, sacituzumab govitecan) has been approved to treat metastatic triple-negative breast cancer. However, it is still unclear whether the success observed in Trop2-positive breast cancer could be replicated in other tumor types, owing to the differences in the expression levels and functions of Trop2 across cancer types. In this review, we summarize the recent progress on the structures and functions of Trop2 and highlight the potential diagnostic and therapeutic value of Trop2 beyond breast cancer. In addition, the promising novel Trop2-targeted agents in the clinic were discussed, which will likely alter the therapeutic landscape of Trop2-positive tumors in the future.
Collapse
Affiliation(s)
- Xinlin Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Junwen Deng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yang Yuan
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Wenshe Sun
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yanhong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Haiming Huang
- Shanghai Asia United Antibody Medical Co., Ltd, Shanghai 201203, China
| | - Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Tao Ming
- Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100094, China
| | - Jialian Wen
- School of Social Science, The University of Manchester, Manchester, UK
| | - Binghuan Huang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
16
|
Fu Z, Li S, Han S, Shi C, Zhang Y. Antibody drug conjugate: the "biological missile" for targeted cancer therapy. Signal Transduct Target Ther 2022; 7:93. [PMID: 35318309 PMCID: PMC8941077 DOI: 10.1038/s41392-022-00947-7] [Citation(s) in RCA: 496] [Impact Index Per Article: 248.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/08/2023] Open
Abstract
Antibody-drug conjugate (ADC) is typically composed of a monoclonal antibody (mAbs) covalently attached to a cytotoxic drug via a chemical linker. It combines both the advantages of highly specific targeting ability and highly potent killing effect to achieve accurate and efficient elimination of cancer cells, which has become one of the hotspots for the research and development of anticancer drugs. Since the first ADC, Mylotarg® (gemtuzumab ozogamicin), was approved in 2000 by the US Food and Drug Administration (FDA), there have been 14 ADCs received market approval so far worldwide. Moreover, over 100 ADC candidates have been investigated in clinical stages at present. This kind of new anti-cancer drugs, known as "biological missiles", is leading a new era of targeted cancer therapy. Herein, we conducted a review of the history and general mechanism of action of ADCs, and then briefly discussed the molecular aspects of key components of ADCs and the mechanisms by which these key factors influence the activities of ADCs. Moreover, we also reviewed the approved ADCs and other promising candidates in phase-3 clinical trials and discuss the current challenges and future perspectives for the development of next generations, which provide insights for the research and development of novel cancer therapeutics using ADCs.
Collapse
Affiliation(s)
- Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, (Parkville Campus) 381 Royal Parade,, Parkville, VIC, 3052, Australia
- Faculty of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, People's Republic of China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
17
|
Mathew Thomas V, Tripathi N, Agarwal N, Swami U. Current and emerging role of sacituzumab govitecan in the management of urothelial carcinoma. Expert Rev Anticancer Ther 2022; 22:335-341. [PMID: 35249433 DOI: 10.1080/14737140.2022.2049763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite rapid advances in the treatment landscape of urothelial cancer, there is a substantial unmet need for safe and effective therapies for patients with locally advanced and metastatic urothelial cancer. Sacituzumab govitecan (SG) is an antibody-drug conjugate, consisting of a Trop-2 directed monoclonal antibody linked to SN-38, the active metabolite of irinotecan. Trop-2 is a glycoprotein overexpressed in various carcinomas, including urothelial carcinomas. AREAS COVERED We review the available data on SG, including mechanism of action, pharmacology, efficacy, safety, and clinical studies regarding locally advanced or metastatic urothelial cancer. EXPERT OPINION SG performed well in the TROPHY-U-01 phase II trial with an objective response rate of 27%. The most common adverse effects were diarrhea, nausea, fatigue, alopecia, and neutropenia, with the most common grade ≥ 3 treatment-related AEs being neutropenia, leukopenia, anemia, diarrhea, and febrile neutropenia. However, these effects were managed effectively with supportive care. SG currently has an accelerated approval for patients with locally advanced or metastatic urothelial cancer who have received platinum-based chemotherapy and either programmed cell death receptor-1 (PD-1) or programmed death-ligand 1 (PD-L1) inhibitor. Several studies are evaluating SG in urothelial cancers as single-agent or in combination with other agents.
Collapse
Affiliation(s)
- Vinay Mathew Thomas
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Nishita Tripathi
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Umang Swami
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
18
|
Zhou H, Liu Z, Wang Y, Wen X, Amador EH, Yuan L, Ran X, Xiong L, Ran Y, Chen W, Wen Y. Colorectal liver metastasis: molecular mechanism and interventional therapy. Signal Transduct Target Ther 2022; 7:70. [PMID: 35246503 PMCID: PMC8897452 DOI: 10.1038/s41392-022-00922-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring malignancy tumors with a high morbidity additionally, CRC patients may develop liver metastasis, which is the major cause of death. Despite significant advances in diagnostic and therapeutic techniques, the survival rate of colorectal liver metastasis (CRLM) patients remains very low. CRLM, as a complex cascade reaction process involving multiple factors and procedures, has complex and diverse molecular mechanisms. In this review, we summarize the mechanisms/pathophysiology, diagnosis, treatment of CRLM. We also focus on an overview of the recent advances in understanding the molecular basis of CRLM with a special emphasis on tumor microenvironment and promise of newer targeted therapies for CRLM, further improving the prognosis of CRLM patients.
Collapse
Affiliation(s)
- Hui Zhou
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Zhongtao Liu
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Yongxiang Wang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Xiaoyong Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Eric H Amador
- Department of Physics, The University of Texas, Arlington, TX, 76019, USA
| | - Liqin Yuan
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Xin Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China.
| | - Yuping Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wei Chen
- Department of Physics, The University of Texas, Arlington, TX, 76019, USA.
- Medical Technology Research Centre, Chelmsford Campus, Anglia Ruskin University, Chelmsford, CM1 1SQ, UK.
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
19
|
Liao S, Wang B, Zeng R, Bao H, Chen X, Dixit R, Xing X. Recent advances in trophoblast cell-surface antigen 2 targeted therapy for solid tumors. Drug Dev Res 2021; 82:1096-1110. [PMID: 34462935 DOI: 10.1002/ddr.21870] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 12/27/2022]
Abstract
Trophoblast cell-surface antigen 2 (Trop 2) is a transmembrane glycoprotein that is highly expressed in various cancer types with relatively low or no baseline expression in most normal tissues. Its overexpression is associated with tumor growth and poor prognosis; Trop 2 is, therefore, an ideal therapeutic target for epithelial cancers. Several Trop 2 targeted therapeutics have recently been developed for the treatment of cancers, such as anti-Trop 2 antibodies and antibody-drug conjugates (ADCs), as well as Trop 2-specific cell therapy. In particular, the safety and clinical benefit of Trop 2-based ADCs have been demonstrated in clinical trials across multiple tumor types, including those with limited treatment options, such as triple-negative breast cancer, platinum-resistant urothelial cancer, and heavily pretreated non-small cell lung cancer. In this review, we elaborate on recent advances in Trop 2 targeted modalities and provide an overview of novel insights for future developments in this field.
Collapse
Affiliation(s)
- Shutan Liao
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Bing Wang
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Rong Zeng
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Haifeng Bao
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Xiaomin Chen
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| | - Rakesh Dixit
- Department of Consultation, Bionavigen LLC, Gaithersburg, Maryland, USA
| | - Xiaoyan Xing
- Department of Consultation, Amador Bioscience Ltd, Hangzhou, China
| |
Collapse
|
20
|
Chen H, Wei F, Yin M, Zhao Q, Liu Z, Yu B, Huang Z. CD27 enhances the killing effect of CAR T cells targeting trophoblast cell surface antigen 2 in the treatment of solid tumors. Cancer Immunol Immunother 2021; 70:2059-2071. [PMID: 33439295 PMCID: PMC10992360 DOI: 10.1007/s00262-020-02838-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 12/20/2020] [Indexed: 12/25/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy, a type of adoptive cell therapy, has been successfully used when treating lymphoma malignancies, but not nearly as successful in treating solid tumors. Trophoblast cell surface antigen 2 (Trop2) is expressed in various solid tumors and plays a role in tumor growth, invasion, and metastasis. In this study, a CAR targeting Trop2 (T2-CAR) was developed with different co-stimulatory intercellular domains. T2-CAR T cells demonstrated a powerful killing ability in the presence of Trop2-positive cells following an in vitro assay. Moreover, T2-CAR T cells produced multiple effector cytokines under antigen stimulation. In tumor-bearing mouse models, the CD27-based T2-CAR T cells showed a higher antitumor activity. Additionally, more CD27-based T2-CAR T cells survived in tumor-bearing mice spleens as well as in the tumor tissue. CD27-based T2-CAR T cells were also found to upregulate IL-7Rα expression, while downregulating PD-1 expression. In conclusion, the CD27 intercellular domain can enhance the T2-CAR T cell killing effect via multiple mechanisms, thus indicating that a CD27-based T2-CAR T cell approach is suitable for clinical applications.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Apoptosis
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Cell Adhesion Molecules/antagonists & inhibitors
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Proliferation
- Female
- Humans
- Immunotherapy, Adoptive/methods
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Receptors, Chimeric Antigen/immunology
- Tumor Cells, Cultured
- Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Huanpeng Chen
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, N1311 Rm, No. 10 Bld, 74 Zhongshan 2nd Rd, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Fengjiao Wei
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, N1311 Rm, No. 10 Bld, 74 Zhongshan 2nd Rd, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Meng Yin
- Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qingyu Zhao
- Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhonghua Liu
- Laboratory Animal Center, South China Agricultural University, Guangzhou, China
| | - Bolan Yu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, BioResource Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhaofeng Huang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, N1311 Rm, No. 10 Bld, 74 Zhongshan 2nd Rd, Guangzhou, 510080, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, China.
| |
Collapse
|
21
|
Lenárt S, Lenárt P, Šmarda J, Remšík J, Souček K, Beneš P. Trop2: Jack of All Trades, Master of None. Cancers (Basel) 2020; 12:E3328. [PMID: 33187148 PMCID: PMC7696911 DOI: 10.3390/cancers12113328] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Trophoblast cell surface antigen 2 (Trop2) is a widely expressed glycoprotein and an epithelial cell adhesion molecule (EpCAM) family member. Although initially identified as a transmembrane protein, other subcellular localizations and processed forms were described. Its congenital mutations cause a gelatinous drop-like corneal dystrophy, a disease characterized by loss of barrier function in corneal epithelial cells. Trop2 is considered a stem cell marker and its expression associates with regenerative capacity in various tissues. Trop2 overexpression was described in tumors of different origins; however, functional studies revealed both oncogenic and tumor suppressor roles. Nevertheless, therapeutic potential of Trop2 was recognized and clinical studies with drug-antibody conjugates have been initiated in various cancer types. One of these agents, sacituzumab govitecan, has been recently granted an accelerated approval for therapy of metastatic triple-negative breast cancer. In this article, we review the current knowledge about the yet controversial function of Trop2 in homeostasis and pathology.
Collapse
Affiliation(s)
- Sára Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
| | - Peter Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
| | - Ján Remšík
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Karel Souček
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 65 Brno, Czech Republic
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Petr Beneš
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic; (S.L.); (P.L.); (J.Š.); (K.S.)
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| |
Collapse
|
22
|
Nagahara Y, Tsujikawa M, Koto R, Uesugi K, Sato S, Kawasaki S, Maruyama K, Nishida K. Corneal Opacity Induced by Light in a Mouse Model of Gelatinous Drop-Like Corneal Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2330-2342. [PMID: 33011110 DOI: 10.1016/j.ajpath.2020.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 11/29/2022]
Abstract
Gelatinous drop-like corneal dystrophy (GDLD) is a severe inherited corneal dystrophy characterized by subepithelial corneal amyloid deposition. We had previously succeeded in identifying the responsible gene, TACSTD2, and subsequently found that the epithelial barrier function is significantly decreased. As with GDLD patients, the knockout mice showed severe loss of tight junction, progressive opacity, and neovascularization in the cornea. We devised an easy method to confirm the loss of the corneal barrier function even before corneal opacity is observed. Furthermore, by using knockout mice, we were able to verify clinical findings, such as the wound healing delay and light-induced acceleration of the disease. This mouse model should prove to be a highly useful tool for investigating the pathology of GDLD and for developing new therapies.
Collapse
Affiliation(s)
- Yukiko Nagahara
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Motokazu Tsujikawa
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan; Department of Biomedical Informatics, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Ryota Koto
- Department of Biomedical Informatics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koji Uesugi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan; Menicon Co., Kasugai, Japan
| | - Shigeru Sato
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan; Department of Biomedical Informatics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Kawasaki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuichi Maruyama
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
23
|
Fagotto F, Aslemarz A. EpCAM cellular functions in adhesion and migration, and potential impact on invasion: A critical review. Biochim Biophys Acta Rev Cancer 2020; 1874:188436. [PMID: 32976980 DOI: 10.1016/j.bbcan.2020.188436] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/19/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022]
Abstract
EpCAM has long been known as a cell surface protein highly expressed in carcinomas. It has since become one of the key cancer biomarkers. Despite its high fame, its actual role in cancer development is still controversial. Beyond a flurry of correlative studies, which point either to a positive or a negative link with tumour progression, there has been surprisingly few studies on the actual cellular mechanisms of EpCAM and on their functional consequences. Clearly, EpCAM plays multiple important roles, in cell proliferation as well as in cell adhesion and migration. The two latter functions, directly relevant for metastasis, are the focus of this review. We attempt here to bring together the available experimental data to build a global coherent view of EpCAM functions. We also include in this overview EpCAM2/Trop2, the close relative of EpCAM. At the core of EpCAM (and EpCAM2/Trop2) function stands the ability to repress contractility of the actomyosin cell cortex. This activity appears to involve direct inhibition by EpCAM of members of the novel PKC family and of a specific downstream PKD-Erk cascade. We will discuss how this activity can result in a variety of adhesive and migratory phenotypes, thus potentially explaining at least part of the apparent inconsistencies between different studies. The picture remains fragmented, and we will highlight some of the conflicting evidence and the many unsolved issues, starting with the controversy around its original description as a cell-cell adhesion molecule.
Collapse
Affiliation(s)
- François Fagotto
- CRBM, University of Montpellier and CNRS, Montpellier 34293, France.
| | - Azam Aslemarz
- CRBM, University of Montpellier and CNRS, Montpellier 34293, France; Department of Biology, McGill University, Montreal, QC H3A1B1, Canada
| |
Collapse
|
24
|
Cocco S, Piezzo M, Calabrese A, Cianniello D, Caputo R, Di Lauro V, Fusco G, di Gioia G, Licenziato M, de Laurentiis M. Biomarkers in Triple-Negative Breast Cancer: State-of-the-Art and Future Perspectives. Int J Mol Sci 2020; 21:E4579. [PMID: 32605126 PMCID: PMC7369987 DOI: 10.3390/ijms21134579] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous group of tumors characterized by aggressive behavior, high risk of distant recurrence, and poor survival. Chemotherapy is still the main therapeutic approach for this subgroup of patients, therefore, progress in the treatment of TNBC remains an important challenge. Data derived from molecular technologies have identified TNBCs with different gene expression and mutation profiles that may help developing targeted therapies. So far, however, only a few of these have shown to improve the prognosis and outcomes of TNBC patients. Robust predictive biomarkers to accelerate clinical progress are needed. Herein, we review prognostic and predictive biomarkers in TNBC, discuss the current evidence supporting their use, and look at the future of this research field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michelino de Laurentiis
- Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola, 53, 80131 Napoli NA, Italy; (S.C.); (M.P.); (A.C.); (D.C.); (R.C.); (V.D.L.); (G.F.); (G.d.G.); (M.L.)
| |
Collapse
|
25
|
Hidalgo-Estévez AM, Stamatakis K, Jiménez-Martínez M, López-Pérez R, Fresno M. Cyclooxygenase 2-Regulated Genes an Alternative Avenue to the Development of New Therapeutic Drugs for Colorectal Cancer. Front Pharmacol 2020; 11:533. [PMID: 32410997 PMCID: PMC7201075 DOI: 10.3389/fphar.2020.00533] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common and recurrent types of cancer, with high mortality rates. Several clinical trials and meta-analyses have determined that the use of pharmacological inhibitors of cyclooxygenase 2 (COX-2), the enzyme that catalyses the rate-limiting step in the synthesis of prostaglandins (PG) from arachidonic acid, can reduce the incidence of CRC as well as the risk of recurrence of this disease, when used together with commonly used chemotherapeutic agents. These observations suggest that inhibition of COX-2 may be useful in the treatment of CRC, although the current drugs targeting COX-2 are not widely used since they increase the risk of health complications. To overcome this difficulty, a possibility is to identify genes regulated by COX-2 activity that could give an advantage to the cells to form tumors and/or metastasize. The modulation of those genes as effectors of COX-2 may cancel the beneficial effects of COX-2 in tumor transformation and metastasis. A review of the available databases and literature and our own data have identified some interesting molecules induced by prostaglandins or COX-2 that have been also described to play a role in colon cancer, being thus potential pharmacological targets in colon cancer. Among those mPGES-1, DUSP4, and 10, Programmed cell death 4, Trop2, and many from the TGFβ and p53 pathways have been identified as genes upregulated in response to COX-2 overexpression or PGs in colon carcinoma lines and overexpressed in colon tumor tissue. Here, we review the available evidence of the potential roles of those molecules in colon cancer in the context of PG/COX signaling pathways that could be critical mediators of some of the tumor growth and metastasis advantage induced by COX-2. At the end, this may allow defining new therapeutic targets/drugs against CRC that could act specifically against tumor cells and would be effective in the prevention and treatment of CRC, lacking the unwanted side effects of COX-2 pharmacological inhibitors, providing alternative approaches in colon cancer.
Collapse
Affiliation(s)
| | - Konstantinos Stamatakis
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto Sanitario de Investigación Princesa, Madrid, Spain
| | - Marta Jiménez-Martínez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ricardo López-Pérez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto Sanitario de Investigación Princesa, Madrid, Spain
| |
Collapse
|
26
|
Oczko-Wojciechowska M, Pfeifer A, Jarzab M, Swierniak M, Rusinek D, Tyszkiewicz T, Kowalska M, Chmielik E, Zembala-Nozynska E, Czarniecka A, Jarzab B, Krajewska J. Impact of the Tumor Microenvironment on the Gene Expression Profile in Papillary Thyroid Cancer. Pathobiology 2020; 87:143-154. [PMID: 32320975 DOI: 10.1159/000507223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/12/2020] [Indexed: 11/19/2022] Open
Abstract
Transcriptome of papillary thyroid cancer (PTC) is well characterized and correlates with some prognostic and genotypic factors, but data addressing the interaction between PTC and tumor microenvironment (TME) are scarce. Therefore, in the present study, we aimed to assess the impact of TME on gene expression profile in PTC. We evaluated the gene expression profile in PTC and normal thyroid cells isolated by laser capture microdissection and in whole tissue slides corresponding to the entire tumor. We included 26 microdissected samples for gene expression analysis (HG-U133 PLUS 2.0, Affymetrix, currently Thermo Fisher Scientific USA): 15 PTC samples, 11 samples of normal thyrocytes, and 30 whole slides (15 PTC and 15 normal thyroid). Transcripts were divided into three groups: differentially expressed both in microdissected and whole slides, transcripts differently expressed in microdissected samples and not changed in whole slides, and transcripts differentially expressed in whole slides and not changed in microdissected samples. Eleven genes were selected for validation in an independent set of samples; among them, four genes differentiated only microdissected PTC and normal cells. Two genes (PTCSC and CTGF) were confirmed. One gene (FOS) was not confirmed by the validation, whereas EGR1 was also significant in whole slide analysis. The other seven genes (TFF3, FN1, MPPED2, MET, KCNJ2, TACSTD2, and GALE) showed differentiated expression in microdissected thyrocytes and in whole tumor slides. Most of identified genes were related to the tumor-microenvironment interaction and confirmed the crosstalk between TME and cancer cells.
Collapse
Affiliation(s)
- Malgorzata Oczko-Wojciechowska
- Genetic and Molecular Diagnostics of Cancer Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland,
| | - Aleksandra Pfeifer
- Genetic and Molecular Diagnostics of Cancer Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Michal Jarzab
- Breast Unit, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Michał Swierniak
- Center of New Technologies, University of Warsaw, Warsaw, Poland
| | - Dagmara Rusinek
- Genetic and Molecular Diagnostics of Cancer Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Tomasz Tyszkiewicz
- Genetic and Molecular Diagnostics of Cancer Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Malgorzata Kowalska
- Genetic and Molecular Diagnostics of Cancer Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Ewa Chmielik
- Tumor Pathology Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Ewa Zembala-Nozynska
- Tumor Pathology Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Agnieszka Czarniecka
- Oncologic and Reconstructive Surgery Clinic, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Barbara Jarzab
- Nuclear Medicine and Endocrine Oncology Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Jolanta Krajewska
- Nuclear Medicine and Endocrine Oncology Department, M. Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| |
Collapse
|
27
|
Sui JSY, Martin P, Gray SG. Pre-clinical models of small cell lung cancer and the validation of therapeutic targets. Expert Opin Ther Targets 2020; 24:187-204. [PMID: 32068452 DOI: 10.1080/14728222.2020.1732353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Small-cell lung cancer (SCLC) is an aggressive form of lung cancer that has a dismal prognosis. One of the factors hindering therapeutic developments for SCLC is that most SCLC is not surgically resected resulting in a paucity of material for analysis. To address this, significant efforts have been made by investigators to develop pre-clinical models of SCLC allowing for downstream target identification in this difficult to treat cancer.Areas covered: In this review, we describe the current pre-clinical models that have been developed to interrogate SCLC, and outline the benefits and limitations associated with each. Using examples we show how each has been used to (i) improve our knowledge of this intractable cancer, and (ii) identify and validate potential therapeutic targets that (iii) are currently under development and testing within the clinic.Expert opinion: The large numbers of preclinical models that have been developed have dramatically improved the ways in which we can examine SCLC and test therapeutic targets/interventions. The newer models are rapidly providing novel avenues for the design and testing of new therapeutics. Despite this many of these models have inherent flaws that limit the possibility of their use for individualized therapy decision-making for SCLC.
Collapse
Affiliation(s)
- Jane S Y Sui
- Thoracic Oncology Research Group, Laboratory Medicine and Molecular Pathology, Central Pathology Laboratory, St. James's Hospital, Dublin, Ireland.,Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Petra Martin
- Thoracic Oncology Research Group, Laboratory Medicine and Molecular Pathology, Central Pathology Laboratory, St. James's Hospital, Dublin, Ireland
| | - Steven G Gray
- Thoracic Oncology Research Group, Laboratory Medicine and Molecular Pathology, Central Pathology Laboratory, St. James's Hospital, Dublin, Ireland.,Labmed Directorate, St. James's Hospital, Dublin, Ireland.,School of Biological Sciences, Dublin Institute of Technology, Dublin, Ireland
| |
Collapse
|
28
|
Sahin M, Yetim A, Ates F. The role of tacstd-2 level in hepatitis C patients (controlled clinical research). Libyan J Med 2019; 14:1622363. [PMID: 31146656 PMCID: PMC6567178 DOI: 10.1080/19932820.2019.1622363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 05/18/2019] [Indexed: 11/01/2022] Open
Abstract
Hepatitis C virus is one of the leading causes of liver cirrhosis and hepatocellular carcinoma. The tumor-associated calcium signal transducer 2 (Tacstd-2) molecule is thought to be involved in the expression of a number of molecules that facilitate transport of hepatitis C into the cell. The aim of this study was to investigate Tacstd-2 concentrations in hepatitis C patients, with and without cirrhosis, and compare with uninfected controls. Sixty-one hepatitis C patients and twenty-nine control (hepatitis C antibody negative patients with dyspeptic complaints) cases were recruited between 2014 and 2016. Tacstd-2 concentrations in all hepatitis C and control patients were measured and compared. In addition, cirrhotic and non-cirrhotic hepatitis C patients were compared in terms of Tacstd-2 concentration, and comparison was made between patients with high and low concentrations of Tacstd-2. The mean Tacstd-2 concentration of patients with Hepatitis C was 691.2 ± 473.3 ng/U was significantly higher (p = 0.043) than in the healthy control group (524 ± 290.1 ng/U). Although the Tacstd-2 value was higher in cirrhotic than the non-cirrhotic patient group, the difference was not statistically significant (p = 0.78). Liver transferase concentrations were higher in hepatitis C patients with a Tacstd-2 concentration <500 ng/U compared to those with a Tacstd-2 concentration >500 ng/U. In patients with hepatitis C, Tacstd-2 level was detected at higher serum concentrations than healthy individuals. The introduction of hepatitis C virus into the cell can be relatively easy in people with a higher serum concentration of Tacstd-2.
Collapse
Affiliation(s)
- Memduh Sahin
- Department of Gastroenterology, Saglik Bilimleri University Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey
| | - Aylin Yetim
- Department of Adeloscent Health, Istanbul University, Istanbul, Turkey
| | - Fehmi Ates
- Department of Gastroenterology, Mersin University, Mersin, Turkey
| |
Collapse
|
29
|
Nishimura T, Mitsunaga M, Sawada R, Saruta M, Kobayashi H, Matsumoto N, Kanke T, Yanai H, Nakamura K. Photoimmunotherapy targeting biliary-pancreatic cancer with humanized anti-TROP2 antibody. Cancer Med 2019; 8:7781-7792. [PMID: 31674732 PMCID: PMC6912056 DOI: 10.1002/cam4.2658] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/30/2019] [Accepted: 10/13/2019] [Indexed: 11/15/2022] Open
Abstract
Photoimmunotherapy (PIT) is a new type of tumor‐specific treatment utilizing monoclonal antibody (mAb)‐photosensitizer conjugates and near‐infrared (NIR) light irradiation. One potential PIT target, the type I transmembrane protein TROP2, is expressed at high levels in many cancers, including pancreatic carcinoma (PC) and cholangiocarcinoma (CC), in which its expression is correlated with poor prognosis and tumor aggressiveness. In this study, we assessed the efficacy of PIT utilizing newly developed humanized anti‐TROP2 mAb conjugated to the photosensitizer IR700 (TROP2‐IR700) for PC and CC. Immunohistochemistry on PC and CC tissue microarrays confirmed that TROP2 is overexpressed in about half of PC and CC specimens. Using cultured PC and CC cells, TROP2‐IR700 localized TROP2‐specific and target‐specific cell killing was observed after NIR light irradiation. In addition, TROP2‐IR700 was localized to mouse xenograft tumors expressing TROP2 after intravenous injection. PC and CC xenograft tumor growth was significantly inhibited by TROP2‐targeted PIT relative to controls. The efficacy of TROP2‐targeted PIT in vitro and against xenografted tumors in vivo suggests promise as a therapy for human PC and CC, both of which currently have dismal prognoses and limited therapeutic options.
Collapse
Affiliation(s)
- Takashi Nishimura
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Makoto Mitsunaga
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ryoichi Sawada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Toru Kanke
- Drug Discovery Laboratories, Chiome Bioscience, Kawasaki, Japan
| | - Hiroyuki Yanai
- Drug Discovery Laboratories, Chiome Bioscience, Kawasaki, Japan
| | - Koji Nakamura
- Drug Discovery Laboratories, Chiome Bioscience, Kawasaki, Japan
| |
Collapse
|
30
|
Fenn KM, Kalinsky K. Sacituzumab govitecan: antibody-drug conjugate in triple-negative breast cancer and other solid tumors. Drugs Today (Barc) 2019; 55:575-585. [PMID: 31584574 DOI: 10.1358/dot.2019.55.9.3039669] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Patients with metastatic triple-negative breast cancer (mTNBC) that has progressed on first-line therapy have a poor prognosis with limited therapeutic options. Sacituzumab govitecan (SG) is a novel antibody-drug conjugate (ADC) that has shown promising efficacy in mTNBC. SG is comprised of SN-38, the active metabolite of irinotecan, conjugated via a hydrolyzable linker to the humanized RS7 antibody targeting trophoblast cell surface antigen 2 (Trop-2), a glycoprotein that is expressed at high levels in many epithelial solid tumors. It has received breakthrough therapy status by the U.S. Food and Drug Administration (FDA) for the treatment of patients with pretreated mTNBC. In this review, we summarize available data regarding the pharmacology, pharmacokinetics, safety and efficacy of SG and describe ongoing and future clinical studies investigating this agent.
Collapse
Affiliation(s)
- K M Fenn
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - K Kalinsky
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.
| |
Collapse
|
31
|
Mori Y, Akita K, Ojima K, Iwamoto S, Yamashita T, Morii E, Nakada H. Trophoblast cell surface antigen 2 (Trop-2) phosphorylation by protein kinase C α/δ (PKCα/δ) enhances cell motility. J Biol Chem 2019; 294:11513-11524. [PMID: 31177095 DOI: 10.1074/jbc.ra119.008084] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/28/2019] [Indexed: 01/05/2023] Open
Abstract
Dysfunction of tight junctions is a critical step during the initial stage of tumor progression. Trophoblast cell surface antigen 2 (Trop-2) belongs to the family of tumor-associated calcium signal transducer (TACSTD) and is required for the stability of claudin-7 and claudin-1, which are often dysregulated or lost in carcinogenesis. Here, we investigated the effects of Trop-2 phosphorylation on cell motility. Analyses using HCT116 cells expressing WT Trop-2 (HCT116/WT) or Trop-2 alanine-substituted at Ser-303 (HCT116/S303A) or Ser-322 (HCT116/S322A) revealed that Trop-2 is phosphorylated at Ser-322. Furthermore, coimmunoprecipitation and Transwell assays indicated that Trop-2 S322A interacted with claudin-7 the strongest, and a phosphomimetic variant, Trop-2 S322E, the weakest and that HCT116/S322E cells have the highest motility and HCT116/S322A cells the lowest. All cell lines had similar levels of claudin-7 mRNA, but levels of claudin-7 protein were markedly decreased in the HCT116/S322E cells, suggesting posttranscriptional control of claudin-7. Moreover, claudin-7 was clearly localized to cell-cell borders in HCT116/S322A cells but was diffusely distributed on the membrane and partially localized in the cytoplasm of HCT116/S322E and HCT116/WT cells. These observations suggested that Trop-2 phosphorylation plays a role in the decrease or mislocalization of claudin-7. Using protein kinase C (PKC) inhibitors and PKC-specific siRNAs, we found that PKCα and PKCδ are responsible for Trop-2 phosphorylation. Of note, chemical PKC inhibition and PKCα- and PKCδ-specific siRNAs reduced motility. In summary, our findings provide evidence that Trop-2 is phosphorylated at Ser-322 by PKCα/δ and that this phosphorylation enhances cell motility and decreases claudin-7 localization to cellular borders.
Collapse
Affiliation(s)
- Yugo Mori
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Kaoru Akita
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Kazuki Ojima
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Shungo Iwamoto
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Tomoko Yamashita
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Hiroshi Nakada
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| |
Collapse
|
32
|
Dong W, Shi J, Yuan T, Qi B, Yu J, Dai J, He L. Antibody-drug conjugates of 7-ethyl-10-hydroxycamptothecin: Sacituzumab govitecan and labetuzumab govitecan. Eur J Med Chem 2019; 167:583-593. [DOI: 10.1016/j.ejmech.2019.02.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/27/2022]
|
33
|
Zaman S, Jadid H, Denson AC, Gray JE. Targeting Trop-2 in solid tumors: future prospects. Onco Targets Ther 2019; 12:1781-1790. [PMID: 30881031 PMCID: PMC6402435 DOI: 10.2147/ott.s162447] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Trop-2 is a transmembrane glycoprotein that is upregulated in all cancer types independent of baseline levels of Trop-2 expression. Trop-2 is an ideal candidate for targeted therapeutics due to it being a transmembrane protein with an extracellular domain overexpressed on a wide variety of tumors as well as its upregulated expression relative to normal cells. As a result, several Trop-2-targeted therapeutics have recently been developed for clinical use, such as anti-Trop-2 antibodies and Trop-2-targeted antibody-drug conjugates (ADC). Subsequently, multiple early-phase clinical trials have demonstrated safety and clinical benefit of Trop-2-based ADCs across multiple tumor types. This includes clinical benefit and tolerability in tumor types with limited treatment options, such as triple-negative breast cancer, platinum-resistant urothelial cancer, and small-cell lung cancer. In this review, we elaborate on all clinical trials involving Trop-2.
Collapse
Affiliation(s)
- Saif Zaman
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Hassan Jadid
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| | - Aaron C Denson
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| |
Collapse
|
34
|
Oeyen E, Hoekx L, De Wachter S, Baldewijns M, Ameye F, Mertens I. Bladder Cancer Diagnosis and Follow-Up: The Current Status and Possible Role of Extracellular Vesicles. Int J Mol Sci 2019; 20:ijms20040821. [PMID: 30769831 PMCID: PMC6412916 DOI: 10.3390/ijms20040821] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Diagnostic methods currently used for bladder cancer are cystoscopy and urine cytology. Cystoscopy is an invasive tool and has low sensitivity for carcinoma in situ. Urine cytology is non-invasive, is a low-cost method, and has a high specificity but low sensitivity for low-grade urothelial tumors. Despite the search for urinary biomarkers for the early and non-invasive detection of bladder cancer, no biomarkers are used at the present in daily clinical practice. Extracellular vesicles (EVs) have been recently studied as a promising source of biomarkers because of their role in intercellular communication and tumor progression. In this review, we give an overview of Food and Drug Administration (FDA)-approved urine tests to detect bladder cancer and why their use is not widespread in clinical practice. We also include non-FDA approved urinary biomarkers in this review. We describe the role of EVs in bladder cancer and their possible role as biomarkers for the diagnosis and follow-up of bladder cancer patients. We review recently discovered EV-derived biomarkers for the diagnosis of bladder cancer.
Collapse
Affiliation(s)
- Eline Oeyen
- Flemish Institute for Technological Research (VITO), 2400 Mol, Belgium.
- Centre for Proteomics (CFP), University of Antwerp, 2020 Antwerp, Belgium.
| | - Lucien Hoekx
- Urology Department, Antwerp University Hospital (UZA), 2650 Edegem, Belgium.
| | - Stefan De Wachter
- Urology Department, Antwerp University Hospital (UZA), 2650 Edegem, Belgium.
| | - Marcella Baldewijns
- Pathological Anatomy Department, Antwerp University Hospital (UZA), 2650 Edegem, Belgium.
| | - Filip Ameye
- Urology Department, General Hospital Maria Middelares Ghent, 9000 Ghent, Belgium.
| | - Inge Mertens
- Flemish Institute for Technological Research (VITO), 2400 Mol, Belgium.
- Centre for Proteomics (CFP), University of Antwerp, 2020 Antwerp, Belgium.
| |
Collapse
|
35
|
A Cell Proliferation and Inflammatory Signature Is Induced by Lawsonia intracellularis Infection in Swine. mBio 2019; 10:mBio.01605-18. [PMID: 30696739 PMCID: PMC6355989 DOI: 10.1128/mbio.01605-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lawsonia intracellularis causes porcine proliferative enteropathy. This is an enteric disease characterized by thickening of the wall of the ileum that leads to decreased growth of animals and diarrhea. In this study, we investigated the host response to L. intracellularis infection by performing transcriptomic and pathway analysis of intestinal tissue samples from groups of infected and noninfected animals at 14, 21, and 28 days postchallenge. At the peak of infection, when animals developed the most severe lesions, infected animals had higher levels of several gene transcripts involved in cellular proliferation and inflammation, including matrix metalloproteinase-7 (MMP7), transglutaminase-2 (TGM2), and oncostatin M (OSM). Histomorphology also revealed general features of intestinal inflammation. This study identified important pathways associated with the host response in developing and resolving lesions due to L. intracellularis infection.IMPORTANCE Lawsonia intracellularis is among the most important enteric pathogens of swine, and it can also infect other mammalian species. Much is still unknown regarding its pathogenesis and the host response, especially at the site of infection. In this study, we uncovered several novel genes and pathways associated with infection. Differentially expressed transcripts, in addition to histological changes in infected tissue, revealed striking similarities between L. intracellularis infection and cellular proliferation mechanisms described in some cancers and inflammatory diseases of the gastrointestinal tract. This research sheds important light into the pathogenesis of L. intracellularis and the host response associated with the lesions caused by infection.
Collapse
|
36
|
Yahyazadeh Mashhadi SM, Kazemimanesh M, Arashkia A, Azadmanesh K, Meshkat Z, Golichenari B, Sahebkar A. Shedding light on the EpCAM: An overview. J Cell Physiol 2019; 234:12569-12580. [DOI: 10.1002/jcp.28132] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Seyed Muhammad Yahyazadeh Mashhadi
- Department of Virology Pasteur Institute of Iran Tehran Iran
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Production Expert at Samandaroo 8 (Biotech Pharmaceutical) Co. Mashhad Iran
| | | | - Arash Arashkia
- Department of Virology Pasteur Institute of Iran Tehran Iran
| | | | - Zahra Meshkat
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Behrouz Golichenari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhosein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Neurogenic inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
37
|
Peng J, Ou Q, Deng Y, Xiao B, Zhang L, Li J, Li Y, Wan D, Lu Z, Fang Y. TROP2 overexpression in colorectal liver oligometastases is associated with poor prognosis after liver resection. Ther Adv Med Oncol 2019; 11:1758835919897543. [PMID: 35173815 PMCID: PMC8842308 DOI: 10.1177/1758835919897543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 12/05/2019] [Indexed: 12/24/2022] Open
Abstract
Background: The aim of this study was to assess trophoblast antigen protein 2 (TROP2)
expression in liver oligometastases and its prognostic value for colorectal
liver oligometastasis (CLO) patients undergoing liver resection. Methods: We retrospectively selected 129 consecutive CLO patients who underwent
curative liver resection between June 1999 and December 2016.
Immunohistochemistry (IHC) was performed to detect TROP2 expression in
paraffin-embedded specimens. Recurrence-free survival (RFS) and overall
survival (OS) were analyzed using the Kaplan–Meier method and log-rank test,
and independent prognostic factors were identified with Cox regression
modeling. Results: TROP2 was expressed in 72.9% (94/129) of liver oligometastatic tissues. TROP2
expression in primary tumors and liver oligometastases was significantly
positively correlated (r = 0.758,
p < 0.001). Survival analysis indicated that CLO
patients with high TROP2 expression had worse 3-year RFS (44.2%
versus 66.4%, p = 0.007) and 3-year OS
rates (70.3% versus 85.4%, p = 0.035) than
did those with low TROP2 expression. Multivariate analysis indicated that
high TROP2 expression was independently associated with poor RFS [hazard
ratio (HR) = 2.017; 95% confidence interval (CI) 1.198–3.396;
p = 0.023] and OS (HR = 2.090; 95% CI 1.037–4.214;
p = 0.039). Gene expression profile analysis indicated
that high TROP2 expression was associated with TNFα signaling
via NF-κB, the inflammatory response and
epithelial–mesenchymal transition (EMT). Conclusions: TROP2 overexpression was associated with an unfavorable oncologic prognosis
in patients with CLO undergoing liver resection. Detecting TROP2 expression
may be valuable for guiding postoperative treatment among CLO patients.
Collapse
Affiliation(s)
- Jianhong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Qingjian Ou
- Department of Colorectal Surgery, Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Yuxiang Deng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Binyi Xiao
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Lin Zhang
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Jibin Li
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Yin Li
- Department Endoscopic and Laser Cancer Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Desen Wan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong, P. R. China
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong 510060, P. R. China
| | - Yujing Fang
- Department of Colorectal Surgery, Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine Guangzhou, Guangdong 510060, P. R. China
| |
Collapse
|
38
|
Tray N, Adams S, Esteva FJ. Antibody-drug conjugates in triple negative breast cancer. Future Oncol 2018; 14:2651-2661. [PMID: 30175620 DOI: 10.2217/fon-2018-0131] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogeneous disease that comprises 15-20% of all breast cancers and is more frequently seen in younger women, African-Americans, and BRCA1 expression. Advanced TNBC carries aggressive features and is associated with overall poor outcomes. Unfortunately, there are no targeted therapies available for non-BRCA associated TNBC, which remains a high unmet therapeutic need. One emerging treatment modality includes antibody-drug conjugates which are highly selective monoclonal antibodies conjugated to cytotoxic agents, designed to deliver cytotoxic drugs to antigen-expressing tumor cells. This review will highlight three antibody-drug conjugates currently being evaluated in TNBC (CDX-011, SGN-LIV1a, IMMU-132), including one that has been given Breakthrough Therapy designation from the US FDA.
Collapse
Affiliation(s)
- Nancy Tray
- Perlmutter Cancer Center at New York University Langone Health, New York, NY 10016, USA
| | - Sylvia Adams
- Perlmutter Cancer Center at New York University Langone Health, New York, NY 10016, USA
| | - Francisco J Esteva
- Perlmutter Cancer Center at New York University Langone Health, New York, NY 10016, USA
| |
Collapse
|
39
|
Matsui S, Harada K, Miyata N, Okochi H, Miyajima A, Tanaka M. Characterization of Peribiliary Gland–Constituting Cells Based on Differential Expression of Trophoblast Cell Surface Protein 2 in Biliary Tract. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2059-2073. [DOI: 10.1016/j.ajpath.2018.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 12/18/2022]
|
40
|
Alehabib E, Jamshidi J, Ghaedi H, Emamalizadeh B, Andarva M, Daftarian N, Rezaei Kanavi M, Mohammadi Torbati P, Espandar G, Alinaghi S, Johari AH, Saghally M, Mohajerani F, Darvish H. Novel Mutations in TACSTD2 Gene in Families with Gelatinous Drop-like Corneal Dystrophy (GDLD). INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 6:204-211. [PMID: 29988226 PMCID: PMC6004293 DOI: 10.22088/bums.6.4.204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/29/2017] [Indexed: 11/12/2022]
Abstract
In the current study, we conducted a mutation screening of tumor-associated calcium signal transducer 2 (TACSTD2) gene in six consanguineous Iranian families with gelatinous drop-like corneal dystrophy (GDLD), in order to find the causative mutations. Detailed eye examination was performed by ophthalmologist to confirm GDLD in patients. To detect the possible mutations, direct Sanger sequencing was performed for the only exon of TACSTD2 gene, and its boundary regions in all patients. In the patients with GDLD, the corneal surface showed lesions with different shapes from mild to severe forms depending on the progress of the disease. The patients showed grayish corneal deposits as a typical mulberry form, corneal dystrophy along with corneal lipid deposition, and vascularization. Targeted Sanger sequencing in TACSTD2 gene revealed the causative mutations in this gene in all studied families. Our study expanded the mutational spectrum of TACSTD2 which along with the related symptoms could help with the diagnosis, and management of the disease.
Collapse
Affiliation(s)
- Elham Alehabib
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Jamshidi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Emamalizadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monavvar Andarva
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narsis Daftarian
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyman Mohammadi Torbati
- Department of Pathology, Labbafi-Nezhad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Goldis Espandar
- Department of Pathology, Labbafi-Nezhad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Alinaghi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Johari
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mansoor Saghally
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohajerani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Darvish
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Goldenberg DM, Stein R, Sharkey RM. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget 2018; 9:28989-29006. [PMID: 29989029 PMCID: PMC6034748 DOI: 10.18632/oncotarget.25615] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
TROP-2 is a glycoprotein first described as a surface marker of trophoblast cells, but subsequently shown to be increased in many solid cancers, with lower expression in certain normal tissues. It regulates cancer growth, invasion and spread by several signaling pathways, and has a role in stem cell biology and other diseases. This review summarizes TROP-2's properties, especially in cancer, and particularly its role as a target for antibody-drug conjugates (ADC) or immunotherapy. When the irinotecan metabolite, SN-38, is conjugated to a humanized anti-TROP-2 antibody (sacituzumab govitecan), it shows potent broad anticancer activity in human cancer xenografts and in patients with advanced triple-negative breast, non-small cell and small-cell lung, as well as urothelial cancers.
Collapse
Affiliation(s)
- David M. Goldenberg
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
- IBC Pharmaceuticals, Inc., Morris Plains, NJ, USA
| | - Rhona Stein
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
| | - Robert M. Sharkey
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
- Immunomedics, Inc., Morris Plains, NJ, USA
| |
Collapse
|
42
|
Zhang L, Yang G, Zhang R, Dong L, Chen H, Bo J, Xue W, Huang Y. Curcumin inhibits cell proliferation and motility via suppression of TROP2 in bladder cancer cells. Int J Oncol 2018; 53:515-526. [PMID: 29901071 PMCID: PMC6017220 DOI: 10.3892/ijo.2018.4423] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022] Open
Abstract
Bladder cancer (BC) has become a serious health prob-lem and represents the second most commonly diagnosed urological tumor. Curcumin is a principal active natural component of turmeric and has long been used in Asia as a traditional herbal medicine. Curcumin suppresses cell growth in various types of cancer, including BC, by regulating numerous molecular signaling pathways. The human trophoblast cell surface antigen 2 (Trop2) belongs to the tumor-associated calcium signal transducer gene family. Trop2 has been described as a cancer driver and is deregulated in various types of cancer. However, whether Trop2 is involved in curcumin-induced BC cell inhibition remains to be elucidated. The present study hypothesized that Trop2 may be a promising target of curcumin in BC cells. It was found that Trop2 was closely involved in curcumin-induced cell proliferation suppression, mobility inhibition, apoptosis, and cell cycle arrest in BC cells. Curcumin decreased the expression of Trop2 and its downstream target cyclin E1, and increased the level of p27. The overexpression of Trop2 enhanced the oncogenic activity of BC cells, whereas downregulation of the expression of Trop2 suppressed cell proliferation and mobility, increased apoptosis, and sensitized BC cells to curcumin treatment. Therefore, Trop2 may be a promising target of curcumin in BC cells and the inhibition of Trop2 may be an important method for the therapeutic management of patients with BC.
Collapse
Affiliation(s)
- Lianhua Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Guoliang Yang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Ruiyun Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Liang Dong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Haige Chen
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Juanjie Bo
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
43
|
Abstract
Objectives: Human trophoblast cell-surface marker (TROP-2) has been reported to be overexpressed in various human carcinomas (CAs) and suggested to be a prognostic marker for some CAs. The diagnostic utility of TROP-2 in CAs has not been explored. Methods: Immunohistochemical evaluation of TROP-2 expression on tissue microarray sections of 136 thyroid neoplasms, surgical specimens of 61 atypical thyroid follicular-patterned lesions [including 33 papillary thyroid carcinomas (PTCs), 17 atypical follicular neoplasms (AFNs), and 11 adenomatoid nodules with focal nuclear atypia (ANFNA)], and 20 benign thyroid lesions, as well as 10 cytology specimens of PTCs was performed. For comparison, immunoassay for Hector Battifora mesothelial-1 (HBME-1), galectin-3, and cytokeratin 19 was performed on the 61 atypical thyroid follicular-patterned lesions. Results: Strong membranous staining with TROP-2 was seen in 94% (33/35) of classic PTCs and 81% (30/37) of confirmed follicular variant PTCs on tissue microarray and routine surgical sections, as well as 100% (10/10) of PTCs on cytology specimens; it was not observed in follicular adenomas (n=51) or CAs (n=37), AFNs or ANFNA (n=28), benign (n=20) or normal (n=15) thyroid tissue. In contrast, the expression of HBME-1 and galectin-3 was identified in 100% (33/33) of surgical cases of PTCs and in 57% (16/28) and 50% (14/28) of AFNs and ANFNA, respectively. Conclusions: Our findings demonstrate that a membranous TROP-2 staining pattern is highly specific for PTC, which may serve as a potential diagnostic marker aiding in the accurate classification of morphologically equivocal thyroid follicular-patterned lesions.
Collapse
|
44
|
Incorporation of CD40 ligand enhances the immunogenicity of tumor‑associated calcium signal transducer 2 virus‑like particles against lung cancer. Int J Mol Med 2018; 41:3671-3679. [PMID: 29568866 DOI: 10.3892/ijmm.2018.3570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/05/2018] [Indexed: 11/05/2022] Open
Abstract
The cell surface glycoprotein Trop‑2 is overexpressed in various types of cancer, including in lung cancer, and has recently been used as an effective immunotherapeutic target. CD40 ligand (CD40L), a tumor necrosis factor superfamily member, is a promising immune adjuvant. Human immunodeficiency virus (HIV) gag‑based virus‑like particles (VLPs) are highly immunogenic, and foreign antigens can be incorporated onto their membrane envelope for cancer vaccine development. In the present study, a HIV gag‑based VLP strategy and Bac‑to‑Bac system were utilized to construct Trop‑2, CD40L and gag recombinant baculoviruses, which were then used to infect TN5 cells in order to form Trop‑2 VLPs or Trop‑2‑CD40L VLPs. These VLPs were characterized using transmission electron microscopy and western blot analysis methods. VLPs incorporating murine Trop‑2 only or incorporating Trop‑2 and CD40L were used to immunize C57BL/6 mice. Immunized mice demonstrated high humoral and cellular immunity responses, whereas the Trop‑2‑CD40L VLPs led to higher immune responses in comparison with Trop‑2 only VLPs. Immunization with Trop‑2‑CD40L VLPs also reduced tumor growth more effectively compared with Trop‑2 VLPs. Furthermore, Trop‑2‑CD40L VLP immunization increased the survival rate of Lewis tumor‑bearing mice more significantly when compared with Trop‑2 only VLPs. In conclusion, the present study provided a novel vaccine design by combination of a tumor antigen and an immune adjuvant based on a VLP strategy, which may be potentially applied as an alternative immunotherapeutic option in the treatment of lung cancer.
Collapse
|
45
|
Sekhar V, Pollicino T, Diaz G, Engle RE, Alayli F, Melis M, Kabat J, Tice A, Pomerenke A, Altan-Bonnet N, Zamboni F, Lusso P, Emerson SU, Farci P. Infection with hepatitis C virus depends on TACSTD2, a regulator of claudin-1 and occludin highly downregulated in hepatocellular carcinoma. PLoS Pathog 2018. [PMID: 29538454 PMCID: PMC5882150 DOI: 10.1371/journal.ppat.1006916] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Entry of hepatitis C virus (HCV) into hepatocytes is a complex process that involves numerous cellular factors, including the scavenger receptor class B type 1 (SR-B1), the tetraspanin CD81, and the tight junction (TJ) proteins claudin-1 (CLDN1) and occludin (OCLN). Despite expression of all known HCV-entry factors, in vitro models based on hepatoma cell lines do not fully reproduce the in vivo susceptibility of liver cells to primary HCV isolates, implying the existence of additional host factors which are critical for HCV entry and/or replication. Likewise, HCV replication is severely impaired within hepatocellular carcinoma (HCC) tissue in vivo, but the mechanisms responsible for this restriction are presently unknown. Here, we identify tumor-associated calcium signal transducer 2 (TACSTD2), one of the most downregulated genes in primary HCC tissue, as a host factor that interacts with CLDN1 and OCLN and regulates their cellular localization. TACSTD2 gene silencing disrupts the typical linear distribution of CLDN1 and OCLN along the cellular membrane in both hepatoma cells and primary human hepatocytes, recapitulating the pattern observed in vivo in primary HCC tissue. Mechanistic studies suggest that TACSTD2 is involved in the phosphorylation of CLDN1 and OCLN, which is required for their proper cellular localization. Silencing of TACSTD2 dramatically inhibits HCV infection with a pan-genotype effect that occurs at the level of viral entry. Our study identifies TACSTD2 as a novel regulator of two major HCV-entry factors, CLDN1 and OCLN, which is strongly downregulated in malignant hepatocytes. These results provide new insights into the complex process of HCV entry into hepatocytes and may assist in the development of more efficient cellular systems for HCV propagation in vitro.
Collapse
Affiliation(s)
- Vandana Sekhar
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Teresa Pollicino
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of Clinical and Molecular Hepatology, Department of Human Pathology, University of Messina, Messina, Italy
| | - Giacomo Diaz
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ronald E. Engle
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Farah Alayli
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Marta Melis
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Juraj Kabat
- Biological Imaging Facility/Research Technologies Branch, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ashley Tice
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anna Pomerenke
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fausto Zamboni
- Liver Transplantation Center, Brotzu Hospital, Cagliari, Italy
| | - Paolo Lusso
- Viral Pathogenesis Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Suzanne U. Emerson
- Molecular Hepatitis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Patrizia Farci
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Redlich N, Robinson AM, Nickel KP, Stein AP, Wheeler DL, Adkins DR, Uppaluri R, Kimple RJ, Van Tine BA, Michel LS. Anti-Trop2 blockade enhances the therapeutic efficacy of ErbB3 inhibition in head and neck squamous cell carcinoma. Cell Death Dis 2018; 9:5. [PMID: 29305574 PMCID: PMC5849045 DOI: 10.1038/s41419-017-0029-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/02/2017] [Accepted: 09/20/2017] [Indexed: 12/22/2022]
Abstract
ErbB3 has been widely implicated in treatment resistance, but its role as a primary treatment target is less clear. Canonically ErbB3 requires EGFR or ErbB2 for activation, whereas these two established treatment targets are thought to signal independently of ErbB3. In this study, we show that ErbB3 is essential for tumor growth of treatment-naive HNSCC patient-derived xenografts. This ErbB3 dependency occurs via ErbB3-mediated control of EGFR activation and HIF1α stabilization, which require ErbB3 and its ligand neuregulin-1. Here, we show that ErbB3 antibody treatment selects for a population of ErbB3-persister cells that express high levels of the transmembrane protein Trop2 that we previously identified as an inhibitor of ErbB3. Co-treatment with anti-ErbB3 and anti-Trop2 antibodies is synergistic and produces a greater anti-tumor response than either antibody alone. Collectively, these data both compel a revision of ErbB-family signaling and delineate a strategy for its effective inhibition in HNSCC.
Collapse
Affiliation(s)
| | - Anthony M Robinson
- Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Kwangok P Nickel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Andrew P Stein
- School of Medicine, Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, USA
| | - Deric L Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Douglas R Adkins
- Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | | | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, and University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| | - Brian A Van Tine
- Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| | - Loren S Michel
- Memorial Sloan-Kettering Cancer Center, Monmouth, NJ, USA
| |
Collapse
|
47
|
Mao Y, Wang X, Zheng F, Wang C, Tang Q, Tang X, Xu N, Zhang H, Zhang D, Xiong L, Liang J, Zhu J. The tumor-inhibitory effectiveness of a novel anti-Trop2 Fab conjugate in pancreatic cancer. Oncotarget 2017; 7:24810-23. [PMID: 27050150 PMCID: PMC5029744 DOI: 10.18632/oncotarget.8529] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/28/2016] [Indexed: 12/26/2022] Open
Abstract
Human trophoblastic cell surface antigen 2 (Trop2) has been reported to act oncogenically. In this study, one-step quantitative real-time polymerase chain reaction (qPCR) test and immunohistochemistry (IHC) analysis with were employed to evaluate the relationship between Trop2 expression and the clinicopathological features of patients with PC. Then a novel anti-Trop2 Fab antibody was conjugated with Doxorubicin (DOX) to form Trop2Fab-DOX, an antibody-drug conjugate. This Trop2Fab-DOX conjugate was characterized by cell ELISA and immunofluorescence assay. MTT and wound healing analyses were used to evaluate the inhibitory effect of Trop2Fab-DOX on PC cell growth in vitro, while xenograft nude mice model was established to examine the tumor-inhibitory effects of PC in vivo. High Trop2 expression was observed in PC tissues and Trop2 expression was associated with several malignant attributes of PC patients, including overall survival. Trop2Fab-DOX can bind to the Trop2-expressing PC cells and provide an improved releasing type of DOX. In addition, Trop2Fab-DOX inhibited the proliferation and suppressed the migration of PC cells in a dose-dependent manner in vitro, while inhibited the growth of PC xenografts in vivo. Trop2 is a specific marker for PC, and a novel Trop2Fab-DOX ADC has a potent antitumor activity
Collapse
Affiliation(s)
- Yuan Mao
- Department of Oncology, Jiangsu Province Geriatric Hospital, Nanjing 210024, China
| | - Xiaoying Wang
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China.,Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Feng Zheng
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Changjun Wang
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Qi Tang
- Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Xiaojun Tang
- Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Ning Xu
- Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Huiling Zhang
- Department of Gynecology and Obstetrics, Nanjing Maternal and Children Care Hospital Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Dawei Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Lin Xiong
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Jie Liang
- Department of Pathology, Wuxi Nanjing Maternal and Children Care Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China.,Department of Pathology and The Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
48
|
Merk D, Schubert-Zsilavecz M. The Linker Approach. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527674381.ch8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Daniel Merk
- Goethe University Frankfurt; Institute of Pharmaceutical Chemistry; Max-von-Laue-Str. 9 60438 Frankfurt Germany
| | - Manfred Schubert-Zsilavecz
- Goethe University Frankfurt; Institute of Pharmaceutical Chemistry; Max-von-Laue-Str. 9 60438 Frankfurt Germany
| |
Collapse
|
49
|
Bardia A, Mayer IA, Diamond JR, Moroose RL, Isakoff SJ, Starodub AN, Shah NC, O’Shaughnessy J, Kalinsky K, Guarino M, Abramson V, Juric D, Tolaney SM, Berlin J, Messersmith WA, Ocean AJ, Wegener WA, Maliakal P, Sharkey RM, Govindan SV, Goldenberg DM, Vahdat LT. Efficacy and Safety of Anti-Trop-2 Antibody Drug Conjugate Sacituzumab Govitecan (IMMU-132) in Heavily Pretreated Patients With Metastatic Triple-Negative Breast Cancer. J Clin Oncol 2017. [DOI: 10.1200/jco.2016.70.8297.2017.2.test] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Aditya Bardia
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Ingrid A. Mayer
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Jennifer R. Diamond
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Rebecca L. Moroose
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Steven J. Isakoff
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Alexander N. Starodub
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Nikita C. Shah
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Joyce O’Shaughnessy
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Kevin Kalinsky
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Michael Guarino
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Vandana Abramson
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Dejan Juric
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Sara M. Tolaney
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Jordan Berlin
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Wells A. Messersmith
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Allyson J. Ocean
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - William A. Wegener
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Pius Maliakal
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Robert M. Sharkey
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Serengulam V. Govindan
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - David M. Goldenberg
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Linda T. Vahdat
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| |
Collapse
|
50
|
Heist RS, Guarino MJ, Masters G, Purcell WT, Starodub AN, Horn L, Scheff RJ, Bardia A, Messersmith WA, Berlin J, Ocean AJ, Govindan SV, Maliakal P, Mudenda B, Wegener WA, Sharkey RM, Goldenberg DM, Camidge DR. Therapy of Advanced Non-Small-Cell Lung Cancer With an SN-38-Anti-Trop-2 Drug Conjugate, Sacituzumab Govitecan. J Clin Oncol 2017; 35:2790-2797. [PMID: 28548889 DOI: 10.1200/jco.2016.72.1894] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose Trop-2, expressed in most solid cancers, may be a target for antibody-drug conjugates (ADCs) in non-small-cell lung cancer (NSCLC). We studied sacituzumab govitecan (IMMU-132), a Trop-2 ADC, for the targeting of SN-38. Patients and Methods We evaluated IMMU-132 in a single-arm multicenter trial in patients with pretreated metastatic NSCLC who received either 8 or 10 mg/kg on days 1 and 8 of 21-day cycles. The primary end points were safety and objective response rate (ORR). Progression-free survival and overall survival were secondary end points. Results Fifty-four patients were treated. In the response-assessable study population (n = 47), which had a median of three prior therapies (range, two to seven), the ORR was 19%; median response duration, 6.0 months (95% CI, 4.8 to 8.3 months); and clinical benefit rate (complete response + partial response + stable disease ≥ 4 months), 43%. ORR in the intention-to-treat (ITT) population was 17% (nine of 54). Responses occurred with a median onset of 3.8 months, including patients who had relapsed or progressed after immune checkpoint inhibitor therapy. Median ITT progression-free survival was 5.2 months (95% CI, 3.2 to 7.1 months) and median ITT overall survival, 9.5 months (95% CI, 5.9 to 16.7 months). Grade 3 or higher adverse events included neutropenia (28%), diarrhea (7%), nausea (7%), fatigue (6%), and febrile neutropenia (4%). One patient developed a transient immune response, despite patients receiving a median of 10 doses. More than 90% of 26 assessable archival tumor specimens were highly positive (2+, 3+) for Trop-2 by immunohistochemistry, which suggests that Trop-2 is not a predictive biomarker for response. Conclusion IMMU-132 was well-tolerated and induced durable responses in heavily pretreated patients with metastatic NSCLC. This ADC should be studied further in this disease and in other patients with Trop-2-expressing tumors.
Collapse
Affiliation(s)
- Rebecca Suk Heist
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Michael J Guarino
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Gregory Masters
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - W Thomas Purcell
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Alexander N Starodub
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Leora Horn
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Ronald J Scheff
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Aditya Bardia
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Wells A Messersmith
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Jordan Berlin
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Allyson J Ocean
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Serengulam V Govindan
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Pius Maliakal
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Boyd Mudenda
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - William A Wegener
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Robert M Sharkey
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - David M Goldenberg
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - D Ross Camidge
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| |
Collapse
|