1
|
Liu Y, Huang W, Saladin RJ, Hsu JC, Cai W, Kang L. Trop2-Targeted Molecular Imaging in Solid Tumors: Current Advances and Future Outlook. Mol Pharm 2024; 21:5909-5928. [PMID: 39537365 PMCID: PMC11832138 DOI: 10.1021/acs.molpharmaceut.4c00848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Trophoblast cell surface antigen 2 (Trop2), a transmembrane glycoprotein, plays a dual role in physiological and pathological processes. In healthy tissues, Trop2 facilitates development and orchestrates intracellular calcium signaling. However, its overexpression in numerous solid tumors shifts its function toward driving cell proliferation and metastasis, thus leading to a poor prognosis. The clinical relevance of Trop2 is underscored by its utility as both a biomarker for diagnostic imaging and a target for therapy. Notably, the U.S. Food and Drug Administration (FDA) has approved sacituzumab govitecan (SG), a novel Trop2-targeted agent, for treating triple-negative breast cancer (TNBC) and refractory urothelial cancer, highlighting the significance of Trop2 in clinical oncology. Molecular imaging, a powerful tool for visualizing and quantifying biological phenomena at the molecular and cellular levels, has emerged as a critical technique for studying Trop2. This approach encompasses various modalities, including optical imaging, positron emission tomography (PET), single photon emission computed tomography (SPECT), and targeted antibodies labeled with radioactive isotopes. Incorporating Trop2-targeted molecular imaging into clinical practice is vital for the early detection, prognostic assessment, and treatment planning of a broad spectrum of solid tumors. Our review captures the latest progress in Trop2-targeted molecular imaging, focusing on both diagnostic and therapeutic applications across diverse tumor types, including lung, breast, gastric, pancreatic, prostate, and cervical cancers, as well as salivary gland carcinomas. We critically evaluate the current state by examining the relevant applications, diagnostic accuracy, therapeutic efficacy, and inherent limitations. Finally, we analyze the challenges impeding widespread clinical application and offer insights into strategies for advancing the field, thereby guiding future research endeavors.
Collapse
Affiliation(s)
- Yongshun Liu
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Rachel J Saladin
- Departments of Radiology and Medical Physics, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
2
|
Jiang Y, Zhou H, Liu J, Ha W, Xia X, Li J, Chao T, Xiong H. Progress and Innovative Combination Therapies in Trop-2-Targeted ADCs. Pharmaceuticals (Basel) 2024; 17:652. [PMID: 38794221 PMCID: PMC11125602 DOI: 10.3390/ph17050652] [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: 04/28/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Precise targeting has become the main direction of anti-cancer drug development. Trophoblast cell surface antigen 2 (Trop-2) is highly expressed in different solid tumors but rarely in normal tissues, rendering it an attractive target. Trop-2-targeted antibody-drug conjugates (ADCs) have displayed promising efficacy in treating diverse solid tumors, especially breast cancer and urothelial carcinoma. However, their clinical application is still limited by insufficient efficacy, excessive toxicity, and the lack of biological markers related to effectiveness. This review summarizes the clinical trials and combination therapy strategies for Trop-2-targeted ADCs, discusses the current challenges, and provides new insights for future advancements.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.J.); (H.Z.); (J.L.); (W.H.); (X.X.); (J.L.)
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.J.); (H.Z.); (J.L.); (W.H.); (X.X.); (J.L.)
| |
Collapse
|
3
|
Ning N, Lu J, Li Q, Li M, Cai Y, Wang H, Li J. Single-sEV profiling identifies the TACSTD2 + sEV subpopulation as a factor of tumor susceptibility in the elderly. J Nanobiotechnology 2024; 22:222. [PMID: 38698420 PMCID: PMC11067244 DOI: 10.1186/s12951-024-02456-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/02/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Aging is a very complex physiological phenomenon, and sEVs are involved in the regulation of this mechanism. Serum samples from healthy individuals under 30 and over 60 years of age were collected to analyze differences in sEVs proteomics. RESULTS Based on PBA analysis, we found that sEVs from the serum of elderly individuals highly express TACSTD2 and identified a subpopulation marked by TACSTD2. Using ELISA, we verified the upregulation of TACSTD2 in serum from elderly human and aged mouse. In addition, we discovered that TACSTD2 was significantly increased in samples from tumor patients and had better diagnostic value than CEA. Specifically, 9 of the 13 tumor groups exhibited elevated TACSTD2, particularly for cervical cancer, colon cancer, esophageal carcinoma, liver cancer and thyroid carcinoma. Moreover, we found that serum sEVs from the elderly (especially those with high TACSTD2 levels) promoted tumor cell (SW480, HuCCT1 and HeLa) proliferation and migration. CONCLUSION TACSTD2 was upregulated in the serum of elderly individuals and patients with tumors, and could serve as a dual biomarker for aging and tumors.
Collapse
Affiliation(s)
- Nannan Ning
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, China
| | - Jianying Lu
- School of Public Health, Shandong University, Jinan, China
| | - Qianpeng Li
- Department of Hematology, Weifang People's Hospital, Weifang, China
| | - Mengmeng Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanling Cai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Hongchun Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, China.
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
4
|
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: 10] [Impact Index Per Article: 10.0] [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
|
5
|
Upadhyay SS, Devasahayam Arokia Balaya R, Parate SS, Dagamajalu S, Keshava Prasad TS, Shetty R, Raju R. An assembly of TROP2-mediated signaling events. J Cell Commun Signal 2023; 17:1105-1111. [PMID: 37014471 PMCID: PMC10409939 DOI: 10.1007/s12079-023-00742-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
Trophoblast cell surface antigen 2 (TROP2) is a calcium-transducing transmembrane protein mainly involved in embryo development. The aberrant expression of TROP2 is observed in numerous cancers, including triple-negative breast cancer, gastric, colorectal, pancreatic, squamous cell carcinoma of the oral cavity, and prostate cancers. The main signaling pathways mediated by TROP2 are calcium signaling, PI3K/AKT, JAK/STAT, MAPKs, and β-catenin signaling. However, collective information about the TROP2-mediated signaling pathway is not available for visualization or analysis. In this study, we constructed a TROP2 signaling map with respect to its role in different cancers. The data curation was done manually by following the NetPath annotation criteria. The described map consists of different molecular events, including 8 activation/inhibition, 16 enzyme catalysis, 19 gene regulations, 12 molecular associations, 39 induced-protein expressions, and 2 protein translocation. The data of the TROP2 pathway map is made freely accessible through the WikiPathways Database ( https://www.wikipathways.org/index.php/Pathway:WP5300 ). Development of TROP2 signaling pathway map.
Collapse
Affiliation(s)
- Shubham Sukerndeo Upadhyay
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018 India
| | | | - Sakshi Sanjay Parate
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018 India
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018 India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018 India
| | - Rohan Shetty
- Department of Surgical Oncology, Yenepoya Medical College Hospital, Yenepoya (Deemed to Be University), Mangalore, 575018 India
| | - Rajesh Raju
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018 India
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018 India
| |
Collapse
|
6
|
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: 2.0] [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
|
7
|
Sperger JM, Helzer KT, Stahlfeld CN, Jiang D, Singh A, Kaufmann KR, Niles DJ, Heninger E, Rydzewski NR, Wang L, Wang L, Yang R, Ren Y, Engle JW, Huang P, Kyriakopoulos CE, Slovin SF, Soule HR, Zhao SG, Kohli M, Tagawa ST, Cai W, Dehm SM, Lang JM. Expression and Therapeutic Targeting of TROP-2 in Treatment-Resistant Prostate Cancer. Clin Cancer Res 2023; 29:2324-2335. [PMID: 36939530 PMCID: PMC10261916 DOI: 10.1158/1078-0432.ccr-22-1305] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/29/2022] [Accepted: 03/14/2023] [Indexed: 03/21/2023]
Abstract
PURPOSE Men with metastatic castration-resistant prostate cancer (mCRPC) frequently develop resistance to androgen receptor signaling inhibitor (ARSI) treatment; therefore, new therapies are needed. Trophoblastic cell-surface antigen (TROP-2) is a transmembrane protein identified in prostate cancer and overexpressed in multiple malignancies. TROP-2 is a therapeutic target for antibody-drug conjugates (ADC). EXPERIMENTAL DESIGN TROP-2 gene (TACSTD2) expression and markers of treatment resistance from prostate biopsies were analyzed using data from four previously curated cohorts of mCRPC (n = 634) and the PROMOTE study (dbGaP accession phs001141.v1.p1, n = 88). EPCAM or TROP-2-positive circulating tumor cells (CTC) were captured from peripheral blood for comparison of protein (n = 15) and gene expression signatures of treatment resistance (n = 40). We assessed the efficacy of TROP-2-targeting agents in a mouse xenograft model generated from prostate cancer cell lines. RESULTS We demonstrated that TACSTD2 is expressed in mCRPC from luminal and basal tumors but at lower levels in patients with neuroendocrine prostate cancer. Patients previously treated with ARSI showed no significant difference in TACSTD2 expression, whereas patients with detectable AR-V7 expression showed increased expression. We observed that TROP-2 can serve as a cell surface target for isolating CTCs, which may serve as a predictive biomarker for ADCs. We also demonstrated that prostate cancer cell line xenografts can be targeted specifically by labeled anti-TROP-2 agents in vivo. CONCLUSIONS These results support further studies on TROP-2 as a therapeutic and diagnostic target for mCRPC.
Collapse
Affiliation(s)
- Jamie M. Sperger
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | - Kyle T. Helzer
- Department of Human Oncology, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Dawei Jiang
- Department of Radiology, University of Wisconsin, Madison, Wisconsin
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, China
| | - Anupama Singh
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - David J. Niles
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin
| | - Erika Heninger
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | | | | | | | - Rendong Yang
- Masonic Cancer Center and Departments of Laboratory Medicine and Pathology and Urology, University of Minnesota, Minneapolis, Minnesota
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yanan Ren
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| | - Christos E. Kyriakopoulos
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Howard R. Soule
- Department of Science, Prostate Cancer Foundation, Santa Monica, California
| | - Shuang G. Zhao
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
- Department of Human Oncology, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Scott T. Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York
| | - Weibo Cai
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
- Department of Radiology, University of Wisconsin, Madison, Wisconsin
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin
| | - Scott M. Dehm
- Masonic Cancer Center and Departments of Laboratory Medicine and Pathology and Urology, University of Minnesota, Minneapolis, Minnesota
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| |
Collapse
|
8
|
Li Z, Jiao X, Robertson AG, Di Sante G, Ashton AW, DiRocco A, Wang M, Zhao J, Addya S, Wang C, McCue PA, South AP, Cordon-Cardo C, Liu R, Patel K, Hamid R, Parmar J, DuHadaway JB, Jones SJM, Casimiro MC, Schultz N, Kossenkov A, Phoon LY, Chen H, Lan L, Sun Y, Iczkowski KA, Rui H, Pestell RG. The DACH1 gene is frequently deleted in prostate cancer, restrains prostatic intraepithelial neoplasia, decreases DNA damage repair, and predicts therapy responses. Oncogene 2023; 42:1857-1873. [PMID: 37095257 PMCID: PMC10238272 DOI: 10.1038/s41388-023-02668-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 04/26/2023]
Abstract
Prostate cancer (PCa), the second leading cause of death in American men, includes distinct genetic subtypes with distinct therapeutic vulnerabilities. The DACH1 gene encodes a winged helix/Forkhead DNA-binding protein that competes for binding to FOXM1 sites. Herein, DACH1 gene deletion within the 13q21.31-q21.33 region occurs in up to 18% of human PCa and was associated with increased AR activity and poor prognosis. In prostate OncoMice, prostate-specific deletion of the Dach1 gene enhanced prostatic intraepithelial neoplasia (PIN), and was associated with increased TGFβ activity and DNA damage. Reduced Dach1 increased DNA damage in response to genotoxic stresses. DACH1 was recruited to sites of DNA damage, augmenting recruitment of Ku70/Ku80. Reduced Dach1 expression was associated with increased homology directed repair and resistance to PARP inhibitors and TGFβ kinase inhibitors. Reduced Dach1 expression may define a subclass of PCa that warrants specific therapies.
Collapse
Affiliation(s)
- Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, VSZ 4S6, Canada
- Dxige Research, Courtenay, BC, V9N 1C2, Canada
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Anthony W Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - Agnese DiRocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Min Wang
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Sankar Addya
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Chenguang Wang
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Peter A McCue
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Mt. Sinai, Hospital, 1468 Madison Ave., Floor 15, New York, NY, 10029, USA
| | - Runzhi Liu
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Kishan Patel
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Rasha Hamid
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Jorim Parmar
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - James B DuHadaway
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, VSZ 4S6, Canada
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
- Abraham Baldwin Agricultural College, Department of Science and Mathematics, Box 15, 2802 Moore Highway, Tifton, GA, 31794, USA
| | - Nikolaus Schultz
- Human Oncology and Pathogenesis Program, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, 3601 Spruce St., Philadelphia, PA, 19104, USA
| | - Lai Yee Phoon
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Hao Chen
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Li Lan
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA.
- The Wistar Cancer Center, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
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: 19] [Impact Index Per Article: 9.5] [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
|
10
|
Mjaess G, Aoun F, Rassy E, Diamand R, Albisinni S, Roumeguère T. Antibody-Drug Conjugates in Prostate Cancer: Where Are we? Clin Genitourin Cancer 2023; 21:171-174. [PMID: 35999150 DOI: 10.1016/j.clgc.2022.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 02/01/2023]
Abstract
Antibody-drug conjugates (ADCs) reflect a new promising approach in prostate cancer, even more so after the practice-changing results in other malignancies, either hematologic or solid. ADCs consist of monoclonal antibodies (mAb) targeted at specific antigens overly expressed on cancer cells compared to normal cells. A cytotoxic payload is attached to the mAb using a stable linker. In prostate cancer, PSMA, STEAP1, TROP2, CD46 and B7-H3 are antigens currently being studied as targets for ADCs. In this paper, we discuss the composition of ADCs and focus on their application and challenges as treatment options in prostate cancer.
Collapse
Affiliation(s)
- Georges Mjaess
- Department of Urology, Hôpital Universitaire de Bruxelles, Brussels, Belgium.
| | - Fouad Aoun
- Department of Urology, Hotel-Dieu de France, Beirut, Lebanon
| | - Elie Rassy
- Department of Oncology, Gustave Roussy Institute, Paris, France
| | - Romain Diamand
- Department of Urology, Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | - Simone Albisinni
- Department of Urology, Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | - Thierry Roumeguère
- Department of Urology, Hôpital Universitaire de Bruxelles, Brussels, Belgium
| |
Collapse
|
11
|
Li Z, Jiao X, Robertson AG, Sante GD, Ashton AW, DiRocco A, Wang M, Zhao J, Addya S, Wang C, McCue PA, South AP, Cordon-Cardo C, Liu R, Patel K, Hamid R, Parmar J, DuHadaway JB, Jones SJ, Casimiro MC, Schultz N, Kossenkov A, Phoon LY, Chen H, Lan L, Sun Y, Iczkowski KA, Rui H, Pestell RG. The DACH1 gene is frequently deleted in prostate cancer, restrains prostatic intraepithelial neoplasia, decreases DNA damage repair, and predicts therapy responses. RESEARCH SQUARE 2023:rs.3.rs-2423179. [PMID: 36712010 PMCID: PMC9882663 DOI: 10.21203/rs.3.rs-2423179/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Prostate cancer (PCa), the second leading cause of death in American men, includes distinct genetic subtypes with distinct therapeutic vulnerabilities. The DACH1 gene encodes a winged helix/Forkhead DNA-binding protein that competes for binding to FOXM1 sites. Herein, DACH1 gene deletion within the 13q21.31-q21.33 region occurs in up to 18% of human PCa and was associated with increased AR activity and poor prognosis. In prostate OncoMice, prostate-specific deletion of the Dach1 gene enhanced prostatic intraepithelial neoplasia (PIN), and was associated with increased TGFb activity and DNA damage. Reduced Dach1 increased DNA damage in response to genotoxic stresses. DACH1 was recruited to sites of DNA damage, augmenting recruitment of Ku70/Ku80. Reduced Dach1 expression was associated with increased homology directed repair and resistance to PARP inhibitors and TGFb kinase inhibitors. Reduced Dach1 expression may define a subclass of PCa that warrants specific therapies.
Collapse
Affiliation(s)
- Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - A. Gordon Robertson
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Anthony W. Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia; Sydney Medical School Northern, University of Sydney, NSW, 2006, Australia
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA 19096
| | - Agnese DiRocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Min Wang
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Sankar Addya
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Chenguang Wang
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Peter A. McCue
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Andrew P. South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Carlos Cordon-Cardo
- Department of Pathology, Mt. Sinai, Hospital, 1468 Madison Ave., Floor 15, New York, NY, 10029
| | - Runzhi Liu
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Kishan Patel
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Rasha Hamid
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Jorim Parmar
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - James B. DuHadaway
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA 19096
| | - Steven J. Jones
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Mathew C. Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
- Abraham Baldwin Agricultural College, Department of Science and Mathematics, Box 15, 2802 Moore Highway, Tifton, GA, 31794
| | - Nikolaus Schultz
- Human Oncology and Pathogenesis Program, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA
| | - Lai Yee Phoon
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA, and Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Hao Chen
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA, and Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Li Lan
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA, and Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Richard G. Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
- The Wistar Cancer Center, Philadelphia, PA 19107
| |
Collapse
|
12
|
Integrin beta1 (ITGB1) as a prognostic marker in esophageal adenocarcinoma. Sci Rep 2022; 12:20745. [PMID: 36456612 PMCID: PMC9715537 DOI: 10.1038/s41598-022-25071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Today, individual prognosis in patients with adenocarcinoma of the esophagus (EAC) is based on post-surgical TNM staging and valid biomarkers are still not implemented. Integrin beta1 (ITGB1) is widely expressed in epithelial cells and promotes cell adhesion and growth. Its impact on tumor progression was described for different tumor entities before, data on its function as a potential biomarker in EAC is not available. Aim of the study is to evaluate the expression level of ITGB1 in a large collective of EAC and its impact on patients´ prognosis. 640 patients with esophageal adenocarcinoma were analyzed immunohistochemically for ITGB1. The data was correlated with long term outcome, clinical, pathological and molecular data (TP53, HER2/neu, c-myc, GATA6, PIK3CA and KRAS). Of 640 patients to be analyzed, 127 (19.8%) showed expression of ITGB1. ITGB1 expression was associated with lymph node metastasis, expression of integrin alphaV and KRAS mutation status. Patients with high ITGB1 expression showed impaired overall survival (22.5 months (95% CI 15.3-29.7 months), vs. 34.1 months (95% CI 25.3-42.4 months), P = 0.024). This effect was particularly evident in the group of patients undergoing primary surgery without prior neoadjuvant therapy (10.2 months (95% CI 1.9-41.7 months) vs. 31.4 months (95% CI 21.1-144.2 months, P = 0.008). ITGB1 was also an independent prognostic marker in multivariable analysis (HR 1.696 (95% CI 1.084-2.653, P = 0.021) in patients that underwent primary surgery. We demonstrate for the first time the prognostic significance of ITGB1 expression in a large EAC patient population.
Collapse
|
13
|
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: 48] [Impact Index Per Article: 16.0] [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
|
14
|
ATF2 loss promotes tumor invasion in colorectal cancer cells via upregulation of cancer driver TROP2. Cell Mol Life Sci 2022; 79:423. [PMID: 35838828 PMCID: PMC9287261 DOI: 10.1007/s00018-022-04445-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
In cancer, the activating transcription factor 2 (ATF2) has pleiotropic functions in cellular responses to growth stimuli, damage, or inflammation. Due to only limited studies, the significance of ATF2 in colorectal cancer (CRC) is not well understood. We report that low ATF2 levels correlated with worse prognosis and tumor aggressiveness in CRC patients. NanoString gene expression and ChIP analysis confirmed trophoblast cell surface antigen 2 (TROP2) as a novel inhibitory ATF2 target gene. This inverse correlation was further observed in primary human tumor tissues. Immunostainings revealed that high intratumoral heterogeneity for ATF2 and TROP2 expression was sustained also in liver metastasis. Mechanistically, our in vitro data of CRISPR/Cas9-generated ATF2 knockout (KO) clones revealed that high TROP2 levels were critical for cell de-adhesion and increased cell migration without triggering EMT. TROP2 was enriched in filopodia and displaced Paxillin from adherens junctions. In vivo imaging, micro-computer tomography, and immunostainings verified that an ATF2KO/TROP2high status triggered tumor invasiveness in in vivo mouse and chicken xenograft models. In silico analysis provided direct support that ATF2low/TROP2high expression status defined high-risk CRC patients. Finally, our data demonstrate that ATF2 acts as a tumor suppressor by inhibiting the cancer driver TROP2. Therapeutic TROP2 targeting might prevent particularly the first steps in metastasis, i.e., the de-adhesion and invasion of colon cancer cells.
Collapse
|
15
|
Wolf I, Gratzke C, Wolf P. Prostate Cancer Stem Cells: Clinical Aspects and Targeted Therapies. Front Oncol 2022; 12:935715. [PMID: 35875084 PMCID: PMC9304860 DOI: 10.3389/fonc.2022.935715] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Despite decades of research and successful improvements in diagnosis and therapy, prostate cancer (PC) remains a major challenge. In recent years, it has become clear that PC stem cells (PCSCs) are the driving force in tumorigenesis, relapse, metastasis, and therapeutic resistance of PC. In this minireview, we discuss the impact of PCSCs in the clinical practice. Moreover, new therapeutic approaches to combat PCSCs are presented with the aim to achieve an improved outcome for patients with PC.
Collapse
Affiliation(s)
- Isis Wolf
- Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- *Correspondence: Philipp Wolf,
| |
Collapse
|
16
|
Feng S, Lou K, Zou X, Zou J, Zhang G. The Potential Role of Exosomal Proteins in Prostate Cancer. Front Oncol 2022; 12:873296. [PMID: 35747825 PMCID: PMC9209716 DOI: 10.3389/fonc.2022.873296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/16/2022] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer is the most prevalent malignant tumor in men across developed countries. Traditional diagnostic and therapeutic methods for this tumor have become increasingly difficult to adapt to today’s medical philosophy, thus compromising early detection, diagnosis, and treatment. Prospecting for new diagnostic markers and therapeutic targets has become a hot topic in today’s research. Notably, exosomes, small vesicles characterized by a phospholipid bilayer structure released by cells that is capable of delivering different types of cargo that target specific cells to regulate biological properties, have been extensively studied. Exosomes composition, coupled with their interactions with cells make them multifaceted regulators in cancer development. Numerous studies have described the role of prostate cancer-derived exosomal proteins in diagnosis and treatment of prostate cancer. However, so far, there is no relevant literature to systematically summarize its role in tumors, which brings obstacles to the later research of related proteins. In this review, we summarize exosomal proteins derived from prostate cancer from different sources and summarize their roles in tumor development and drug resistance.
Collapse
Affiliation(s)
- Shangzhi Feng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, China
| | - Kecheng Lou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, China
| | - Xiaofeng Zou
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, The First Affiliated Hospital of Ganna Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, The First Affiliated Hospital of Ganna Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, Jiangxi, China
- *Correspondence: Junrong Zou, ; Guoxi Zhang,
| | - Guoxi Zhang
- Department of Urology, The First Affiliated hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, The First Affiliated Hospital of Ganna Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, Jiangxi, China
- *Correspondence: Junrong Zou, ; Guoxi Zhang,
| |
Collapse
|
17
|
Hayashi T, Hinata N. Current status and future prospects of antibody-drug conjugates in urological malignancies. Int J Urol 2022; 29:1100-1108. [PMID: 35581739 DOI: 10.1111/iju.14925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/25/2022] [Indexed: 12/16/2022]
Abstract
Antibody-drug conjugates represent a promising new treatment option that uses the targeting ability of an antibody to deliver cytotoxic drugs directly to tumors. Antibody-drug conjugates provide the opportunity to deliver drugs to antigen-expressing cancer cells while minimizing toxicity to normal tissue, achieving wider therapeutic windows. To date, three antibody-drug conjugates have been approved by the US Food and Drug Administration, and many antibody-drug conjugates are under clinical development for urological malignancies. In this paper, we review the mechanism, history, and development of antibody-drug conjugates, and review the current landscape of antibody-drug conjugates in urological malignancies including 12 targets and 18 antibody-drug conjugates in prostate cancer, renal cancer, and urothelial cancer. Furthermore, we review the rational combination of antibody-drug conjugates with immune checkpoint inhibitors and consider future prospects to enhance the therapeutic activity of antibody-drug conjugates in urological malignancies.
Collapse
Affiliation(s)
- Tetsutaro Hayashi
- Department of Urology, Hiroshima University Graduate School of Biomedical and Health Science, Hiroshima, Japan
| | - Nobuyuki Hinata
- Department of Urology, Hiroshima University Graduate School of Biomedical and Health Science, Hiroshima, Japan
| |
Collapse
|
18
|
Antibody-Drug Conjugates in Uro-Oncology. Target Oncol 2022; 17:203-221. [PMID: 35567672 DOI: 10.1007/s11523-022-00872-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Abstract
Currently available treatment options for patients with refractory metastatic prostate, bladder, or kidney cancers are limited with the prognosis remaining poor. Advances in the pathobiology of tumors has led to the discovery of cancer antigens that may be used as the target for cancer treatment. Antibody-drug conjugates (ADCs) are a relatively new concept in cancer treatment that broaden therapeutic landscape. ADCs are examples of a 'drug delivery into the tumor' system composed of an antigen-directed antibody linked to a cytotoxic drug that may release cytotoxic components after binding to the antigen located on the surface of tumor cells. The clinical properties of drugs are influenced by every component of ADCs. Regarding uro-oncology, enfortumab vedotin (EV) and sacituzumab govitecan (SG) are currently registered for patients with locally advanced or metastatic urothelial cancer following previous treatment with an immune checkpoint inhibitor (iCPI; programmed death receptor-1 [PD-1] or programmed death-ligand 1 [PD-L1]) inhibitor) and platinum-containing chemotherapy. The EV-301 trial showed that EV significantly prolonged the overall survival compared with classic chemotherapy. The TROPHY-U-01 trial conducted to evaluate SG demonstrated promising results as regards the objective response rate and duration of response. The safety and efficacy of ADCs in monotherapy and polytherapy (mainly with iCPIs) for different cancer stages and tumor types are assessed in numerous ongoing clinical trials. The aim of this review is to present new molecular biomarkers, specific mechanisms of action, and ongoing clinical trials of ADCs in genitourinary cancers. In the expert discussion, we assess the place of ADCs in uro-oncology and discuss their clinical value.
Collapse
|
19
|
Trop-2, Na+/K+ ATPase, CD9, PKCα, cofilin assemble a membrane signaling super-complex that drives colorectal cancer growth and invasion. Oncogene 2022; 41:1795-1808. [DOI: 10.1038/s41388-022-02220-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/10/2022] [Accepted: 01/27/2022] [Indexed: 12/20/2022]
|
20
|
Kamble PR, Patkar SR, Breed AA, Pathak BR. N-glycosylation status of Trop2 impacts its surface density, interaction with claudin-7 and exosomal release. Arch Biochem Biophys 2021; 714:109084. [PMID: 34774484 DOI: 10.1016/j.abb.2021.109084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 12/31/2022]
Abstract
Trophoblast antigen 2 (Trop2) is a type I transmembrane protein post-translationally modified by N-linked glycosylation. It was originally detected in trophoblasts but was later shown to be frequently overexpressed in many epithelial cancers. Recently, anti-Trop2 antibody-drug conjugate has been FDA approved for the treatment of metastatic triple-negative breast and urothelial carcinomas, making it an important tumor antigen. The current study explored the significance of N-glycosylation of Trop2 by substituting specific N-glycan addition sites by site-directed mutagenesis. The mutant proteins were characterized in transiently transfected HEK293 cells. The N-glycosylation mutants did not affect protein expression, stability, dimerization ability and matriptase mediated cleavage. However, N120A and N208A mutants showed decreased interaction with its binding partner claudin-7. Our earlier reported Trop2 mutant V194A, which shows aberrant glycosylation, also displayed hampered interaction with claudin-7. To further characterize the mutants, stable clones expressing wild type and mutant Trop2 were generated in OVCAR3 cell line. Interestingly, surface biotinylation assay showed significantly higher surface expression of N120A and N208A mutants whereas surface localization was drastically reduced for V194A Trop2 mutant. Though overexpression of wild type Trop2 did not cause any change in fibronectin-mediated FAK (Focal adhesion kinase) signaling; expression of N120A mutant, surprisingly downregulated FAK signaling. Furthermore, exosomal release of Trop2 was also decreased in N120A and N208A mutants. This data suggests that site-specific N-glycan addition determines Trop2 surface density, claudin-7 interaction and exosomal release.
Collapse
Affiliation(s)
- Pradnya R Kamble
- Cellular and Structural Biology Division, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Shivali R Patkar
- Cellular and Structural Biology Division, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Ananya A Breed
- Cellular and Structural Biology Division, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Bhakti R Pathak
- Cellular and Structural Biology Division, ICMR-National Institute for Research in Reproductive Health, Mumbai, India.
| |
Collapse
|
21
|
Pavšič M. Trop2 Forms a Stable Dimer with Significant Structural Differences within the Membrane-Distal Region as Compared to EpCAM. Int J Mol Sci 2021; 22:ijms221910640. [PMID: 34638982 PMCID: PMC8508679 DOI: 10.3390/ijms221910640] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 02/05/2023] Open
Abstract
Trop2 is a cell-surface transmembrane glycoprotein involved in the maintenance of epithelial tissue integrity and is an important carcinoma marker. It shares similar claudin-interaction capacity with its paralogue EpCAM, and both are implicated in signaling triggered by proteolytic cleavage within the ectodomain. However, the cell proliferation-regulating interactions with IGF-1, neuregulin-1, and α5β1 integrin appear to be Trop2-specific. To illuminate the structural differences between Trop2 and EpCAM, we report the first crystal structure of a Trop2 ectodomain dimer and compare it to the analogous part of EpCAM. While the overall fold of the two proteins is similar, the dimers differ. In Trop2, the inter-subunit contacts are more extensive than in EpCAM, and there are two major differences in the membrane-distal regions. The immunogenic N-terminal domain is in Trop2 almost colinear with the dimer interface plain and consequently more laterally exposed, and the cleft of yet unknown functionality between the two subunits is almost absent. Furthermore, the site of initial signaling-associated proteolytic cleavage in Trop2 is accessible in the dimeric state, while in EpCAM dimer destabilization is required. The structural differences highlight the divergent evolutionary path of the two proteins and pave the way for their structure-based utilization in therapy.
Collapse
Affiliation(s)
- Miha Pavšič
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna Pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
22
|
Heninger E, Kosoff D, Rodems TS, Sethakorn N, Singh A, Gungurthi H, Carlson KN, Yang B, Gilsdorf C, Pasch CA, Deming DA, Ellis L, Beebe DJ, Jarrard DF, Lang JM. Live cell molecular analysis of primary prostate cancer organoids identifies persistent androgen receptor signaling. Med Oncol 2021; 38:135. [PMID: 34581895 PMCID: PMC8478748 DOI: 10.1007/s12032-021-01582-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/12/2021] [Indexed: 11/29/2022]
Abstract
Prostate Cancer (PC) is a disease with remarkable tumor heterogeneity that often manifests in significant intra-patient variability with regards to clinical outcomes and treatment response. Commonly available PC cell lines do not accurately reflect the complexity of this disease and there is critical need for development of new models to recapitulate the intricate hierarchy of tumor pathogenesis. In current study, we established ex vivo primary patient-derived cancer organoid (PDCO) cultures from prostatectomy specimens of patients with locally advanced PC. We then performed a comprehensive multi-parameter characterization of the cellular composition utilizing a novel approach for live-cell staining and direct imaging in the integrated microfluidic Stacks device. Using orthogonal flow cytometry analysis, we demonstrate that primary PDCOs maintain distinct subsets of epithelial cells throughout culture and that these cells conserve expression of androgen receptor (AR)-related elements. Furthermore, to confirm the tumor-origin of the PDCOs we have analyzed the expression of PC-associated epigenetic biomarkers including promoter methylation of the GSTP1, RASSF1 and APC and RARb genes by employing a novel microfluidic rare-event screening protocol. These results demonstrate that this ex vivo PDCO model recapitulates the complexity of the epithelial tumor microenvironment of multifocal PC using orthogonal analyses. Furthermore, we propose to leverage the Stacks microfluidic device as a high-throughput, translational platform to interrogate phenotypic and molecular endpoints with the capacity to incorporate a complex tumor microenvironment.
Collapse
Affiliation(s)
- Erika Heninger
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Tamara S Rodems
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Nan Sethakorn
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Anupama Singh
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Harshitha Gungurthi
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Kristin N Carlson
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Bing Yang
- Department of Urology, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Cole Gilsdorf
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Cheri A Pasch
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Dustin A Deming
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Leigh Ellis
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, 1111 Highland Ave., Madison, WI, 53705, USA
| | - David F Jarrard
- Department of Urology, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA. .,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,Wisconsin Institutes for Medical Research, Rm 7151, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
23
|
Corrao G, Zaffaroni M, Bergamaschi L, Augugliaro M, Volpe S, Pepa M, Bonizzi G, Pece S, Amodio N, Mistretta FA, Luzzago S, Musi G, Alessi S, La Fauci FM, Tordonato C, Tosoni D, Cattani F, Gandini S, Petralia G, Pravettoni G, De Cobelli O, Viale G, Orecchia R, Marvaso G, Jereczek-Fossa BA. Exploring miRNA Signature and Other Potential Biomarkers for Oligometastatic Prostate Cancer Characterization: The Biological Challenge behind Clinical Practice. A Narrative Review. Cancers (Basel) 2021; 13:cancers13133278. [PMID: 34208918 PMCID: PMC8267686 DOI: 10.3390/cancers13133278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/16/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The oligometastatic prostate cancer state is defined as the presence of a number of lesions ≤ 5 and has been significantly correlated with better survival if compared to a number of metastases > 5. In particular, patients in an oligometastatic setting could benefit from a metastates directed therapy, which could control the disease delaying the start of systemic therapies. For this reason, the selection of true-oligometastatic patients who could benefit from such approach is particularly important in this setting. The aim of the present narrative review is to report the current state of the art on the liquid biopsy-derived analytes and their reliability as biomarkers in the clinics for the identification of true-oligometastatic patients. This kind of molecular profiling could refine current developments in the era of precision oncology allowing patients’ stratification and leading to more refined therapeutic strategies. Abstract In recent years, a growing interest has been directed towards oligometastatic prostate cancer (OMPC), as patients with three to five metastatic lesions have shown a significantly better survival as compared with those harboring a higher number of lesions. The efficacy of local ablative treatments directed on metastatic lesions (metastases-directed treatments) was extensively investigated, with the aim of preventing further disease progression and delaying the start of systemic androgen deprivation therapies. Definitive diagnosis of prostate cancer is traditionally based on histopathological analysis. Nevertheless, a bioptic sample—static in nature—inevitably fails to reflect the dynamics of the tumor and its biological response due to the dynamic selective pressure of cancer therapies, which can profoundly influence spatio-temporal heterogeneity. Furthermore, even with new imaging technologies allowing an increasingly early detection, the diagnosis of oligometastasis is currently based exclusively on radiological investigations. Given these premises, the development of minimally-invasive liquid biopsies was recently promoted and implemented as predictive biomarkers both for clinical decision-making at pre-treatment (baseline assessment) and for monitoring treatment response during the clinical course of the disease. Through liquid biopsy, different biomarkers, commonly extracted from blood, urine or saliva, can be characterized and implemented in clinical routine to select targeted therapies and assess treatment response. Moreover, this approach has the potential to act as a tissue substitute and to accelerate the identification of novel and consistent predictive analytes cost-efficiently. However, the utility of tumor profiling is currently limited in OMPC due to the lack of clinically validated predictive biomarkers. In this scenario, different ongoing trials, such as the RADIOSA trial, might provide additional insights into the biology of the oligometastatic state and on the identification of novel biomarkers for the outlining of true oligometastatic patients, paving the way towards a wider ideal approach of personalized medicine. The aim of the present narrative review is to report the current state of the art on the solidity of liquid biopsy-related analytes such as CTCs, cfDNA, miRNA and epi-miRNA, and to provide a benchmark for their further clinical implementation. Arguably, this kind of molecular profiling could refine current developments in the era of precision oncology and lead to more refined therapeutic strategies in this subset of oligometastatic patients.
Collapse
Affiliation(s)
- Giulia Corrao
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
| | - Mattia Zaffaroni
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
| | - Luca Bergamaschi
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
| | - Matteo Augugliaro
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
- Correspondence:
| | - Stefania Volpe
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
| | - Matteo Pepa
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
| | - Giuseppina Bonizzi
- Department of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Salvatore Pece
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
- Novel Diagnostics Program, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | | | - Stefano Luzzago
- Department of Urology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (F.A.M.); (S.L.)
| | - Gennaro Musi
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
- Department of Urology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (F.A.M.); (S.L.)
| | - Sarah Alessi
- Division of Radiology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Francesco Maria La Fauci
- Unit of Medical Physics IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (F.M.L.F.); (F.C.)
| | - Chiara Tordonato
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Daniela Tosoni
- Novel Diagnostics Program, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Federica Cattani
- Unit of Medical Physics IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (F.M.L.F.); (F.C.)
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Giuseppe Petralia
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
- Division of Radiology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Gabriella Pravettoni
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
- Applied Research Division for Cognitive and Psychological Science, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Ottavio De Cobelli
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
- Department of Urology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (F.A.M.); (S.L.)
| | - Giuseppe Viale
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
- Department of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Roberto Orecchia
- Scientific Direction, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Giulia Marvaso
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (G.C.); (M.Z.); (L.B.); (S.V.); (M.P.); (G.M.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (S.P.); (G.M.); (C.T.); (G.P.); (G.P.); (O.D.C.); (G.V.)
| |
Collapse
|
24
|
Huang D, Chen J, Hu D, Xie F, Yang T, Li Z, Wang X, Xiao Y, Zhong J, Jiang Y, Zhang X, Zhong T. Advances in Biological Function and Clinical Application of Small Extracellular Vesicle Membrane Proteins. Front Oncol 2021; 11:675940. [PMID: 34094979 PMCID: PMC8172959 DOI: 10.3389/fonc.2021.675940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Small extracellular vesicles are membrane-bound vesicles secreted into extracellular spaces by virtually all types of cells. These carry a large number of membrane proteins on their surface that are incorporated during their biogenesis in cells. The composition of the membrane proteins hence bears the signature of the cells from which they originate. Recent studies have suggested that the proteins on these small extracellular vesicles can serve as biomarkers and target proteins for the diagnosis and treatment of diseases. This article classifies small extracellular vesicle membrane proteins and summarizes their pathophysiological functions in the diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Defa Huang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jie Chen
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Die Hu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fangfang Xie
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tong Yang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhengzhe Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiaoxing Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yongwei Xiao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jianing Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Xiaokang Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Department of Preventive Medicine, Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.,Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
25
|
Gao Z, Pang B, Li J, Gao N, Fan T, Li Y. Emerging Role of Exosomes in Liquid Biopsy for Monitoring Prostate Cancer Invasion and Metastasis. Front Cell Dev Biol 2021; 9:679527. [PMID: 34017837 PMCID: PMC8129505 DOI: 10.3389/fcell.2021.679527] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the most common solid tumor in men. While patients with local PCa have better prognostic survival, patients with metastatic PCa have relatively high mortality rates. Existing diagnostic methods for PCa rely on tissue biopsy and blood prostate-specific antigen (PSA) detection; however, the PSA test does not detect aggressive PCa. Liquid biopsy is a promising technique to overcome tumor heterogeneity in diagnosis, provide more comprehensive information, and track tumor progression over time, allowing for the development of treatment options at all stages of PCa. Exosomes containing proteins and nucleic acids are potential sources of tumor biomarkers. Accumulating evidence indicates that exosomes play important roles in cell communication and tumor progression and are suitable for monitoring PCa progression and metastasis. In this review, we summarize recent advances in the use of exosomal proteins and miRNAs as biomarkers for monitoring PCa invasion and metastasis and discuss their feasibility in clinical diagnosis.
Collapse
Affiliation(s)
- Zhengfan Gao
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Bairen Pang
- Faculty of Medicine, St George and Sutherland Clinical School, St George Hospital, UNSW Sydney, Kensington, NSW, Australia
| | - Jing Li
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Na Gao
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Tianli Fan
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China
| | - Yong Li
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou, China.,Faculty of Medicine, St George and Sutherland Clinical School, St George Hospital, UNSW Sydney, Kensington, NSW, Australia
| |
Collapse
|
26
|
Shen M, Liu S, Stoyanova T. The role of Trop2 in prostate cancer: an oncogene, biomarker, and therapeutic target. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2021; 9:73-87. [PMID: 33816696 PMCID: PMC8012837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
Prostate cancer remains the second leading cause of cancer-associated deaths amongst American men. Trop2, a cell surface glycoprotein, correlates with poor clinical outcome and is highly expressed in metastatic, treatment-resistant prostate cancer. High levels of Trop2 are prognostic for biochemical recurrence. Trop2 regulates tumor growth and metastatic ability of prostate cancer. Moreover, overexpression of Trop2 drives the transdifferentiation to neuroendocrine phenotype in prostate cancer. In addition, Trop2 is overexpressed across epithelial cancers and has emerged as a promising therapeutic target in various solid epithelial cancers. The FDA (Food and Drug Administration) recently approved the use of a Trop2-targeting ADC (antibody-drug conjugate), Sacituzumab Govitecan (IMMU-132), for metastatic, triple-negative breast cancer with at least two prior therapies. Here, we review the role of Trop2 in prostate tumorigenesis and its potential as a promising biomarker and therapeutic target for prostate cancer.
Collapse
Affiliation(s)
- Michelle Shen
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University USA
| | - Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University USA
| |
Collapse
|
27
|
Rosellini M, Santoni M, Mollica V, Rizzo A, Cimadamore A, Scarpelli M, Storti N, Battelli N, Montironi R, Massari F. Treating Prostate Cancer by Antibody-Drug Conjugates. Int J Mol Sci 2021; 22:ijms22041551. [PMID: 33557050 PMCID: PMC7913806 DOI: 10.3390/ijms22041551] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/26/2021] [Accepted: 01/30/2021] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer is the most frequent malignancy in the worldwide male population; it is also one of the most common among all the leading cancer-related death causes. In the last two decades, the therapeutic scenario of metastatic castration-resistant prostate cancer has been enriched by the use of chemotherapy and androgen receptor signaling inhibitors (ARSI) and, more recently, by immunotherapy and poly(ADP–ribose) polymerase (PARP) inhibitors. At the same time, several trials have shown the survival benefits related to the administration of novel ARSIs among patients with non-castration-resistant metastatic disease along with nonmetastatic castration-resistant cancer too. Consequently, the therapeutic course of this malignancy has been radically expanded, ensuring survival benefits never seen before. Among the more recently emerging agents, the so-called “antibody–drug conjugates” (ADCs) are noteworthy because of their clinical practice changing outcomes obtained in the management of other malignancies (including breast cancer). The ADCs are novel compounds consisting of cytotoxic agents (also known as the payload) linked to specific antibodies able to recognize antigens expressed over cancer cells’ surfaces. As for prostate cancer, researchers are focusing on STEAP1, TROP2, PSMA, CD46 and B7-H3 as optimal antigens which may be targeted by ADCs. In this paper, we review the pivotal trials that have currently changed the therapeutic approach to prostate cancer, both in the nonmetastatic castration-resistant and metastatic settings. Therefore, we focus on recently published and ongoing trials designed to investigate the clinical activity of ADCs against prostate malignancy, characterizing these agents. Lastly, we briefly discuss some ADCs-related issues with corresponding strategies to overwhelm them, along with future perspectives for these promising novel compounds.
Collapse
Affiliation(s)
- Matteo Rosellini
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.R.); (V.M.); (A.R.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
- Correspondence: (M.S.); (F.M.)
| | - Veronica Mollica
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.R.); (V.M.); (A.R.)
| | - Alessandro Rizzo
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.R.); (V.M.); (A.R.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Nadia Storti
- Direzione Sanitaria Azienda Sanitaria Unica Regionale, 60122 Ancona, Italy;
| | | | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Francesco Massari
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.R.); (V.M.); (A.R.)
- Correspondence: (M.S.); (F.M.)
| |
Collapse
|
28
|
Barok M, Puhka M, Yazdi N, Joensuu H. Extracellular vesicles as modifiers of antibody-drug conjugate efficacy. J Extracell Vesicles 2021; 10:e12070. [PMID: 33613875 PMCID: PMC7881363 DOI: 10.1002/jev2.12070] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a new class of anti-cancer drugs that consist of a monoclonal antibody, a highly potent small-molecule cytotoxic drug, and a chemical linker between the two. ADCs can selectively deliver cytotoxic drugs to cancer cells leading to a reduced systemic exposure and a wider therapeutic window. To date, nine ADCs have received marketing approval, and over 100 are being investigated in nearly 600 clinical trials. The target antigens of at least eight out of the nine approved anti-cancer ADCs and of 69 investigational ADCs are present on extracellular vesicles (EVs) (tiny particles produced by almost all types of cells) that may carry their contents into local and distant cells. Therefore, the EVs have a potential to mediate both the anti-cancer effects and the adverse effects of ADCs. In this overview, we discuss the mechanisms of action of ADCs and the resistance mechanisms to them, the EV-mediated resistance mechanisms to small molecule anti-cancer drugs and anti-cancer monoclonal antibodies, and the EVs as modifiers of ADC efficacy and safety.
Collapse
Affiliation(s)
- Mark Barok
- Helsinki University Hospital and University of HelsinkiHelsinkiFinland
- Laboratory of Molecular OncologyUniversity of HelsinkiBiomedicumHelsinkiFinland
| | - Maija Puhka
- Institute for Molecular Medicine FIMMEV and HiPrep CoreUniversity of HelsinkiHelsinkiFinland
| | - Narjes Yazdi
- Helsinki University Hospital and University of HelsinkiHelsinkiFinland
- Laboratory of Molecular OncologyUniversity of HelsinkiBiomedicumHelsinkiFinland
| | - Heikki Joensuu
- Helsinki University Hospital and University of HelsinkiHelsinkiFinland
- Laboratory of Molecular OncologyUniversity of HelsinkiBiomedicumHelsinkiFinland
| |
Collapse
|
29
|
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: 79] [Impact Index Per Article: 15.8] [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
|
30
|
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: 28] [Impact Index Per Article: 5.6] [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
|
31
|
Fagotto F. EpCAM as Modulator of Tissue Plasticity. Cells 2020; 9:E2128. [PMID: 32961790 PMCID: PMC7563481 DOI: 10.3390/cells9092128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 01/01/2023] Open
Abstract
The Epithelial Cell Adhesion Molecule or EpCAM is a well-known marker highly expressed in carcinomas and showing a strong correlation with poor cancer prognosis. While its name relates to its proposed function as a cell adhesion molecule, EpCAM has been shown to have various signalling functions. In particular, it has been identified as an important positive regulator of cell adhesion and migration, playing an essential role in embryonic morphogenesis as well as intestinal homeostasis. This activity is not due to its putative adhesive function, but rather to its ability to repress myosin contractility by impinging on a PKC signalling cascade. This mechanism confers EpCAM the unique property of favouring tissue plasticity. I review here the currently available data, comment on possible connections with other properties of EpCAM, and discuss the potential significance in the context of cancer invasion.
Collapse
Affiliation(s)
- François Fagotto
- CRBM, University of Montpellier and CNRS, 34293 Montpellier, France
| |
Collapse
|
32
|
Vicente‐Ruiz S, Serrano‐Martí A, Armiñán A, Vicent MJ. Nanomedicine for the Treatment of Advanced Prostate Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sonia Vicente‐Ruiz
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Antoni Serrano‐Martí
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - Ana Armiñán
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia 46012 Spain
| |
Collapse
|
33
|
Kamble PR, Rane S, Breed AA, Joseph S, Mahale SD, Pathak BR. Proteolytic cleavage of Trop2 at Arg87 is mediated by matriptase and regulated by Val194. FEBS Lett 2020; 594:3156-3169. [PMID: 32761920 DOI: 10.1002/1873-3468.13899] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/22/2020] [Accepted: 07/13/2020] [Indexed: 01/08/2023]
Abstract
Proteolytic processing is an important post-translational modification affecting protein activity and stability. In the current study, we investigate the N-terminal cleavage of Trop2, a protein which is overexpressed in many cancers. We demonstrate that Trop2 is cleaved at Arg87 by a transmembrane serine protease, matriptase. Homology modeling and site-directed mutagenesis of amino acids in close proximity to the matriptase cleavage site reveal the importance of Val194 in regulating Trop2 cleavage. Co-immunoprecipitation studies confirm that amino acid substitutions at Arg87, Thr88, Lys189, Val194, and His195 do not affect Trop2 dimerization. However, cleavage of wild-type Trop2 by matriptase is inhibited when it is allowed to dimerize with a V194 A mutant monomer, further confirming the role of Val194 in matriptase-mediated N-terminal cleavage.
Collapse
Affiliation(s)
- Pradnya R Kamble
- Division of Structural Biology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Sanjana Rane
- Division of Structural Biology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Ananya A Breed
- Division of Structural Biology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Shaini Joseph
- Genetic Research Center, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Smita D Mahale
- Division of Structural Biology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Bhakti R Pathak
- Division of Structural Biology, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
34
|
Xu J, Liao K, Fu Z, Xiong Z. Screening differentially expressed genes of pancreatic cancer between Mongolian and Han people using bioinformatics technology. BMC Cancer 2020; 20:298. [PMID: 32272917 PMCID: PMC7147062 DOI: 10.1186/s12885-020-06722-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 03/06/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND To screen and analyze differentially expressed genes in pancreatic carcinoma tissues taken from Mongolian and Han patients by Affymetrix Genechip. METHODS Pancreatic ductal cell carcinoma tissues were collected from the Mongolian and Han patients undergoing resection in the Second Affiliated Hospital of Nanchang University from March 2015 to May 2018 and the total RNA was extracted. Differentially expressed genes were selected from the total RNA qualified by Nanodrop 2000 and Agilent 2100 using Affymetrix and a cartogram was drawn; The gene ontology (GO) analysis and Pathway analysis were used for the collection and analysis of biological information of these differentially expressed genes. Finally, some differentially expressed genes were verified by real-time PCR. RESULTS Through the microarray analysis of gene expression, 970 differentially expressed genes were detected by comparing pancreatic cancer tissue samples between Mongolian and Han patients. A total of 257 genes were significantly up-regulated in pancreatic cancer tissue samples in Mongolian patients; while a total of 713 genes were down-regulated. In the Gene Ontology database, 815 differentially expressed genes were identified with clear GO classification, and CPB1 gene showed the highest increase in expression level (multiple difference: 31.76). The pathway analysis detected 28 signaling pathways that included these differentially expressed genes, involving a total of 178 genes. Among these pathways, the enrichment of differentially expressed genes in the FAK signaling pathway was the strongest and COL11A1 gene showed the highest multiple difference (multiple difference: 5.02). The expression of differentially expressed genes CPB1, COL11A1、ITGA4、BIRC3、PAK4、CPA1、CLPS、PIK3CG and HLA-DPA1 determined by real-time PCR were consistent with the results of gene microarray analysis. CONCLUSIONS The results of microarray analysis of gene expression profiles showed that there are a large number of differentially expressed genes in pancreatic cancer tissue samples comparing Mongolian and Han population. These genes are closely related to the cell proliferation, differentiation, invasion, metastasis and multi-drug resistance in pancreatic cancer. They are also involved in the regulation of multiple important signaling pathways in organisms.
Collapse
Affiliation(s)
- Jiasheng Xu
- Department of Pathology, the First Affiliated Hospital of Nanchang University, No.17 YONGWAIZHENG Street, Nanchang, 330006, Jiangxi, China
| | - Kaili Liao
- Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Zhonghua Fu
- Department of Burns, the First Affiliated Hospital of Nanchang University, No.17 YONGWAIZHENG Street, Nanchang, 330006, Jiangxi, China.
| | - Zhenfang Xiong
- Department of Pathology, the First Affiliated Hospital of Nanchang University, No.17 YONGWAIZHENG Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
35
|
Saber SH, Ali HEA, Gaballa R, Gaballah M, Ali HI, Zerfaoui M, Abd Elmageed ZY. Exosomes are the Driving Force in Preparing the Soil for the Metastatic Seeds: Lessons from the Prostate Cancer. Cells 2020; 9:E564. [PMID: 32121073 PMCID: PMC7140426 DOI: 10.3390/cells9030564] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Exosomes are nano-membrane vesicles that various cell types secrete during physiological and pathophysiological conditions. By shuttling bioactive molecules such as nucleic acids, proteins, and lipids to target cells, exosomes serve as key regulators for multiple cellular processes, including cancer metastasis. Recently, microvesicles have emerged as a challenge in the treatment of prostate cancer (PCa), encountered either when the number of vesicles increases or when the vesicles move into circulation, potentially with an ability to induce drug resistance, angiogenesis, and metastasis. Notably, the exosomal cargo can induce the desmoplastic response of PCa-associated cells in a tumor microenvironment (TME) to promote PCa metastasis. However, the crosstalk between PCa-derived exosomes and the TME remains only partially understood. In this review, we provide new insights into the metabolic and molecular signatures of PCa-associated exosomes in reprogramming the TME, and the subsequent promotion of aggressive phenotypes of PCa cells. Elucidating the molecular mechanisms of TME reprogramming by exosomes draws more practical and universal conclusions for the development of new therapeutic interventions when considering TME in the treatment of PCa patients.
Collapse
Affiliation(s)
- Saber H. Saber
- Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt;
| | - Hamdy E. A. Ali
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Rofaida Gaballa
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Mohamed Gaballah
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Hamed I. Ali
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Mourad Zerfaoui
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Zakaria Y. Abd Elmageed
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| |
Collapse
|
36
|
Trop2 is a driver of metastatic prostate cancer with neuroendocrine phenotype via PARP1. Proc Natl Acad Sci U S A 2020; 117:2032-2042. [PMID: 31932422 PMCID: PMC6994991 DOI: 10.1073/pnas.1905384117] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
NEPC is a highly aggressive subtype of prostate cancer that is increasing in incidence, likely due to use of new secondary androgen deprivation therapies. Here, we demonstrate that Trop2 is significantly elevated in CRPC and NEPC and represents a driver of metastatic NEPC. Trop2 overexpression increases tumor growth, drives metastasis and neuroendocrine phenotype, and significantly increases PARP1 levels. Inhibition of PARP1 in Trop2-driven NEPC significantly decreases neuroendocrine features, tumor growth, and metastatic colonization in vivo, suggesting that PARP1 inhibitors may represent a promising therapeutic strategy for metastatic prostate cancer expressing high levels of Trop2. Resistance to androgen deprivation therapy, or castration-resistant prostate cancer (CRPC), is often accompanied by metastasis and is currently the ultimate cause of prostate cancer-associated deaths in men. Recently, secondary hormonal therapies have led to an increase of neuroendocrine prostate cancer (NEPC), a highly aggressive variant of CRPC. Here, we identify that high levels of cell surface receptor Trop2 are predictive of recurrence of localized prostate cancer. Moreover, Trop2 is significantly elevated in CRPC and NEPC, drives prostate cancer growth, and induces neuroendocrine phenotype. Overexpression of Trop2 induces tumor growth and metastasis while loss of Trop2 suppresses these abilities in vivo. Trop2-driven NEPC displays a significant up-regulation of PARP1, and PARP inhibitors significantly delay tumor growth and metastatic colonization and reverse neuroendocrine features in Trop2-driven NEPC. Our findings establish Trop2 as a driver and therapeutic target for metastatic prostate cancer with neuroendocrine phenotype and suggest that high Trop2 levels could identify cancers that are sensitive to Trop2-targeting therapies and PARP1 inhibition.
Collapse
|
37
|
Wang J, Ni J, Beretov J, Thompson J, Graham P, Li Y. Exosomal microRNAs as liquid biopsy biomarkers in prostate cancer. Crit Rev Oncol Hematol 2019; 145:102860. [PMID: 31874447 DOI: 10.1016/j.critrevonc.2019.102860] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed solid-organ cancer in males. The PSA testing may cause overdiagnosis and overtreatment for PCa patients. There is an urgent need for new biomarkers with greater discriminative precision for diagnosis and risk-stratification, to select for prostate biopsy and treatment of PCa. Liquid biopsy is a promising field with the potential to provide comprehensive information on the genetic landscape at diagnosis and to track genomic evolution over time in order to tailor the therapeutic choices at all stages of PCa. Exosomes, containing RNAs, DNAs and proteins, have been shown to be involved in tumour progression and a rich potential source of tumour biomarkers, especially for profiling analysis of their miRNAs content. In this review, we summarise the exosomal miRNAs in PCa diagnosis, prognosis and management, and further discuss their possible technical challenges associated with isolating PCa-specific exosomes.
Collapse
Affiliation(s)
- Jingpu Wang
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
| | - Jie Ni
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
| | - Julia Beretov
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia; Anatomical Pathology, NSW Health Pathology, St. George Hospital, Kogarah, NSW, Australia
| | - James Thompson
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia; Department of Urology, St. George Hospital, Kogarah, NSW, Australia; Prostate Clinical Research Group, Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - Peter Graham
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
| | - Yong Li
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia; School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
38
|
Remšík J, Binó L, Kahounová Z, Kharaishvili G, Šimecková Š, Fedr R, Kucírková T, Lenárt S, Muresan XM, Slabáková E, Knopfová L, Bouchal J, Král M, Beneš P, Soucek K. Trop-2 plasticity is controlled by epithelial-to-mesenchymal transition. Carcinogenesis 2019; 39:1411-1418. [PMID: 30010814 DOI: 10.1093/carcin/bgy095] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 07/13/2018] [Indexed: 12/23/2022] Open
Abstract
The cell surface glycoprotein Trop-2 is commonly overexpressed in carcinomas and represents an exceptional antigen for targeted therapy. Here, we provide evidence that surface Trop-2 expression is functionally connected with an epithelial phenotype in breast and prostate cell lines and in patient tumor samples. We further show that Trop-2 expression is suppressed epigenetically or through the action of epithelial-to-mesenchymal transition transcription factors and that deregulation of Trop-2 expression is linked with cancer progression and poor patient prognosis. Moreover, our data suggest that the cancer plasticity-driven intratumoral heterogeneity in Trop-2 expression may significantly contribute to response and resistance to therapies targeting Trop-2-expressing cells.
Collapse
Affiliation(s)
- Ján Remšík
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lucia Binó
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Gvantsa Kharaishvili
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Šárka Šimecková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Radek Fedr
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Tereza Kucírková
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Sára Lenárt
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Ximena Maria Muresan
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Eva Slabáková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Lucia Knopfová
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Milan Král
- Department of Urology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Petr Beneš
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karel Soucek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
39
|
A systematic review of contemporary management of oligometastatic prostate cancer: fighting a challenge or tilting at windmills? World J Urol 2019; 37:2343-2353. [PMID: 30706122 DOI: 10.1007/s00345-019-02652-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Amongst the unanswered questions regarding prostate cancer (PCa), the optimal management of oligometastatic disease remains one of the major concerns of the scientific community. The very existence of this category is still subject to controversy. Aim of this systematic review is to summarize current available data on the most appropriate management of oligometastatic PCa. EVIDENCE ACQUISITION All relevant studies published in English up to November the 1st were identified through systematic searches in PubMed, EMBASE, Cochrane Library, CINAHL, Google Scholar and Ovid database. A search was performed including the combination of following words: (prostate cancer) and (metastatic) and [(oligo) or (PSMA) or (cytoreductive) or (stereotaxic radiotherapy) or (prostatectomy)]. 3335 articles were reviewed. After title screening and abstract reading, 118 papers were considered for full reading, leaving a total of 36 articles for the systematic review. EVIDENCE SYNTHESIS There is still no consensus on the definition of oligometastatic disease, nor on the imaging modalities used for its detection. While retrospective studies suggest an added benefit with the treatment the primitive tumor by cytoreductive prostatectomy (55% survival rate vs 21%, p < 0.001), prospective studies do not validate the same outcome. Nonetheless, most studies have reported a reduction in local complications after cytoreductive prostatectomy (< 10%) compared to the best systemic treatment (25-30%). Concerning radiotherapy, an overall survival benefit for patients with a low metastatic burden was found in STAMPEDE (HR 0.68, 95% CI 0.52-0.90; p = 0.007) and suggested in subgroup analysis of the HORRAD trial. Regarding the impact of metastases-directed therapy (MDT), the STOMP and ORIOLE trials suggested that metastatic disease control might improve androgen deprivation therapy-free survival (in STOMP: 21 vs 13 months for MDT vs standard of care). Nonetheless, the impact of MDT on long-term oncologic results remains unclear. Finally, oligometastatic disease appears to be a biologically different entity compared to high-burden metastatic disease. New findings on exosomes appear to make them intriguing biomarkers in the early phases of oligometastatic PCa. CONCLUSION Oligometastatic PCa is today a poorly understood disease. The implementation of new imaging techniques as whole-body MRI and PSMA PET/CT has increased exponentially the number of oligometastatic patients detected. Data of available trials suggest a benefit from cytoreductive prostatectomy to reduce local complication, though its impact on survival remains unknown. Radiotherapy may be beneficial for patients with low-burden metastatic PCa, while MDT may delay the need for androgen deprivation therapy. Results from ongoing trials data are eagerly awaited to draw reliable recommendations.
Collapse
|
40
|
Wang SH, Liou GG, Liu SH, Chang JS, Hsiao JR, Yen YC, Chen YL, Wu WL, Chang JY, Chen YW. Laminin γ2-enriched extracellular vesicles of oral squamous cell carcinoma cells enhance in vitro lymphangiogenesis via integrin α3-dependent uptake by lymphatic endothelial cells. Int J Cancer 2019; 144:2795-2810. [PMID: 30485433 DOI: 10.1002/ijc.32027] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022]
Abstract
Oral squamous cell carcinoma (OSCC) LN1-1 cells previously showed greater capacities for lymphangiogenesis and lymph node metastasis compared to their parental OEC-M1 cells, in addition to an ability to enhance the migration and tube formation of lymphatic endothelial cells (LECs). Purified by a series of differential centrifugations and characterized using electron microscopy, dynamic light scattering and western blot, LN1-1 cell-derived extracellular vesicles (LN1-1 EVs) were shown to promote LEC migration, tube formation and uptake by LECs more effectively than did OEC-M1 cell-derived EVs (OEC-M1 EVs). Using stable isotope labeling with amino acids in cell culture/liquid chromatography-tandem mass spectrometry-based proteomic platform, the laminin-332 proteins, including laminin α3, β3 and γ2, were validated as highly expressed proteins in LN1-1 EVs. Clinically, a higher level of laminin-332 was detected in plasma EVs from OSCC patients with lymph node metastasis than in both healthy controls and OSCC patients without lymphatic metastasis, suggesting EV-borne laminin-332 as a novel and noninvasive biomarker for the detection of lymph node metastasis in OSCC. The knockdown of laminin γ2 and inhibition by anti-laminin-332 neutralizing antibodies impaired LN1-1 EV-mediated LEC migration, tube formation and uptake by LECs. Importantly, laminin γ2-deficient EVs showed a reduced ability to drain into lymph nodes in comparison with the control EVs. In addition, the laminin 332/γ2-mediated EV uptake was dependent on integrin α3 but not β1, β4 or α6. Collectively, the uptake of laminin γ2-enriched EVs by LECs enhanced in vitro lymphangiogenesis and EV-borne laminin-332 is thus a viable biomarker for OSCC.
Collapse
Affiliation(s)
- Ssu-Han Wang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Gunn-Guang Liou
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Szu-Heng Liu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jeffrey S Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Jenn-Ren Hsiao
- Department of Otolaryngology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chen Yen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Lin Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Wan-Ling Wu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Wen Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.,Ph.D. Program for Aging, Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
41
|
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: 4.8] [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
|
42
|
Sin STK, Li Y, Liu M, Yuan YF, Ma S, Guan XY. Down-regulation of TROP-2 Predicts Poor Prognosis of Hepatocellular Carcinoma Patients. Hepatol Commun 2018; 2:1408-1414. [PMID: 30411086 PMCID: PMC6211329 DOI: 10.1002/hep4.1242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/12/2018] [Indexed: 01/16/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and lethal cancer types worldwide, especially in Asian countries. Genetic alterations, including hyperactivation of oncogenes and loss of expression of tumor suppressor genes, greatly contribute to the initiation and progression of HCC. Here we report that down-regulation of trophoblast cell surface antigen 2 (TROP-2) was frequently detected in HCC. Transcriptome sequencing of non-tumor and HCC patient samples revealed down-regulation of TROP-2 in tumor tissues. Immunohistochemical staining showed nearly undetectable levels of TROP-2 in HCC tissues but distinct and strong staining of TROP-2 in adjacent non-tumor tissues. The frequent down-regulation of TROP-2 expression was further confirmed in an in-house cohort of 205 pairs of HCC patient samples and in the Cancer Genome Atlas (TCGA) databases. Furthermore, the down-regulation of TROP-2 was associated with poor overall survival of HCC patients, severe adjacent organ invasion, and poor differentiation of HCC. Using bisulfite genomic sequencing and methylation-specific polymerase chain reaction analyses, we show that higher levels of promoter methylation were detected in the DNA samples of HCC tissues (low TROP-2 expression) than that of the non-tumor tissues (high TROP-2 expression). Conclusion: Taken together, our data suggest that promoter hypermethylation contributes to the frequent down-regulation of TROP-2 in HCC, and that TROP-2 down-regulation predicts poor prognosis of HCC patients.
Collapse
Affiliation(s)
- Sarah T K Sin
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine the University of Hong Kong Hong Kong.,State Key Laboratory for Liver Research, Li Ka Shing Faculty of Medicine the University of Hong Kong Hong Kong
| | - Yan Li
- Department of Biology South University of Science and Technology of China Shenzhen China
| | - Ming Liu
- School of Basic Sciences Guangzhou Medical University Guangzhou China
| | - Yun-Fei Yuan
- State Key Laboratory of Oncology in South China Sun Yat-Sen University Cancer Center Guangzhou China
| | - Stephanie Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine the University of Hong Kong Hong Kong
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine the University of Hong Kong Hong Kong.,State Key Laboratory for Liver Research, Li Ka Shing Faculty of Medicine the University of Hong Kong Hong Kong.,State Key Laboratory of Oncology in South China Sun Yat-Sen University Cancer Center Guangzhou China
| |
Collapse
|
43
|
Prostate cancer sheds the αvβ3 integrin in vivo through exosomes. Matrix Biol 2018; 77:41-57. [PMID: 30098419 DOI: 10.1016/j.matbio.2018.08.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/03/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022]
Abstract
The αvβ3 integrin has been shown to promote aggressive phenotypes in many types of cancers, including prostate cancer. We show that GFP-labeled αvβ3 derived from cancer cells circulates in the blood and is detected in distant lesions in NOD scid gamma (NSG) mice. We, therefore, hypothesized that αvβ3 travels through exosomes and tested its levels in pools of vesicles, which we designate extracellular vesicles highly enriched in exosomes (ExVs), and in exosomes isolated from the plasma of prostate cancer patients. Here, we show that the αvβ3 integrin is found in patient blood exosomes purified by sucrose or iodixanol density gradients. In addition, we provide evidence that the αvβ3 integrin is transferred through ExVs isolated from prostate cancer patient plasma to β3-negative recipient cells. We also demonstrate the intracellular localization of β3-GFP transferred via cancer cell-derived ExVs. We show that the ExVs present in plasma from prostate cancer patients contain higher levels of αvβ3 and CD9 as compared to plasma ExVs from age-matched subjects who are not affected by cancer. Furthermore, using PSMA antibody-bead mediated immunocapture, we show that the αvβ3 integrin is expressed in a subset of exosomes characterized by PSMA, CD9, CD63, and an epithelial-specific marker, Trop-2. Finally, we present evidence that the levels of αvβ3, CD63, and CD9 remain unaltered in ExVs isolated from the blood of prostate cancer patients treated with enzalutamide. Our results suggest that detecting exosomal αvβ3 integrin in prostate cancer patients could be a clinically useful and non-invasive biomarker to follow prostate cancer progression. Moreover, the ability of αvβ3 integrin to be transferred from ExVs to recipient cells provides a strong rationale for further investigating the role of αvβ3 integrin in the pathogenesis of prostate cancer and as a potential therapeutic target.
Collapse
|
44
|
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: 184] [Impact Index Per Article: 26.3] [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
|
45
|
Inamura K, Yokouchi Y, Kobayashi M, Ninomiya H, Sakakibara R, Subat S, Nagano H, Nomura K, Okumura S, Shibutani T, Ishikawa Y. Association of tumor TROP2 expression with prognosis varies among lung cancer subtypes. Oncotarget 2018; 8:28725-28735. [PMID: 28404926 PMCID: PMC5438686 DOI: 10.18632/oncotarget.15647] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/27/2017] [Indexed: 01/04/2023] Open
Abstract
TROP2 is a transmembrane glycoprotein that is overexpressed in various cancers. Emerging evidence suggests that TROP2-targeting therapies are efficacious and safe in patients with multiple prior treatments. TROP2 is a promising target for lung cancer treatment; however, little is known regarding the association of TROP2 expression with clinicopathological/molecular features, including prognosis, in lung cancer. We examined consecutive cases of adenocarcinoma, squamous cell carcinoma (SqCC), and high-grade neuroendocrine tumor (HGNET) for the membranous expression of TROP2 using immunohistochemistry. High TROP2 expression was observed in 64% (172/270) of adenocarcinomas, 75% (150/201) of SqCCs, and 18% (21/115) of HGNETs. Intriguingly, the association of TROP2 expression with mortality was dependent on the lung cancer subtype. High TROP2 expression was associated with higher lung cancer-specific mortality in adenocarcinomas [univariable hazard ratio (HR) = 1.60, 95% confidence interval (CI) = 1.07–2.44, P = 0.022)], but not in SqCCs (univariable HR = 0.79, 95% CI = 0.35–1.94, P = 0.79). In HGNETs, high TROP2 expression was associated with lower lung cancer-specific mortality in both univariable and multivariable analyses (multivariable HR = 0.13, 95% CI = 0.020–0.44, P = 0.0003). Our results suggest a differential role for TROP2 in different lung cancer subtypes.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Yusuke Yokouchi
- Translational Medicine & Clinical Pharmacology Department, Daiichi Sankyo Co., Ltd., Shinagawa-ku, Tokyo 140-0005, Japan
| | - Maki Kobayashi
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Hironori Ninomiya
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Rie Sakakibara
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan.,Department of Integrated Pulmonology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Sophia Subat
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Hiroko Nagano
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Kimie Nomura
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Sakae Okumura
- Thoracic Oncology Center, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Tomoko Shibutani
- Translational Medicine & Clinical Pharmacology Department, Daiichi Sankyo Co., Ltd., Shinagawa-ku, Tokyo 140-0005, Japan
| | - Yuichi Ishikawa
- Division of Pathology, The Cancer Institute; Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| |
Collapse
|
46
|
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: 0.1] [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
|
47
|
Sayeed A, Lu H, Liu Q, Deming D, Duffy A, McCue P, Dicker AP, Davis RJ, Gabrilovich D, Rodeck U, Altieri DC, Languino LR. β1 integrin- and JNK-dependent tumor growth upon hypofractionated radiation. Oncotarget 2018; 7:52618-52630. [PMID: 27438371 PMCID: PMC5288136 DOI: 10.18632/oncotarget.10522] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 06/15/2016] [Indexed: 12/17/2022] Open
Abstract
Radiation therapy is an effective cancer treatment modality although tumors invariably become resistant. Using the transgenic adenocarcinoma of mouse prostate (TRAMP) model system, we report that a hypofractionated radiation schedule (10 Gy/day for 5 consecutive days) effectively blocks prostate tumor growth in wild type (β1wt /TRAMP) mice as well as in mice carrying a conditional ablation of β1 integrins in the prostatic epithelium (β1pc-/- /TRAMP). Since JNK is known to be suppressed by β1 integrins and mediates radiation-induced apoptosis, we tested the effect of SP600125, an inhibitor of c-Jun amino-terminal kinase (JNK) in the TRAMP model system. Our results show that SP600125 negates the effect of radiation on tumor growth in β1pc-/- /TRAMP mice and leads to invasive adenocarcinoma. These effects are associated with increased focal adhesion kinase (FAK) expression and phosphorylation in prostate tumors in β1pc-/- /TRAMP mice. In marked contrast, radiation-induced tumor growth suppression, FAK expression and phosphorylation are not altered by SP600125 treatment of β1wt /TRAMP mice. Furthermore, we have reported earlier that abrogation of insulin-like growth factor receptor (IGF-IR) in prostate cancer cells enhances the sensitivity to radiation. Here we further explore the β1/IGF-IR crosstalk and report that β1 integrins promote cell proliferation partly by enhancing the expression of IGF-IR. In conclusion, we demonstrate that β1 integrin-mediated inhibition of JNK signaling modulates tumor growth rate upon hypofractionated radiation.
Collapse
Affiliation(s)
- Aejaz Sayeed
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Huimin Lu
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Qin Liu
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - David Deming
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Alexander Duffy
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam P Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dmitry Gabrilovich
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Translational Tumor Immunology Program, The Wistar Institute, Philadelphia, PA, USA
| | - Ulrich Rodeck
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
48
|
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: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/metabolism
- Cell Adhesion Molecules/metabolism
- Cell Line, Tumor
- Cetuximab/pharmacology
- Cetuximab/therapeutic use
- Cobalt/pharmacology
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Mice
- Neuregulin-1/antagonists & inhibitors
- Neuregulin-1/genetics
- Neuregulin-1/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/immunology
- Receptor, ErbB-3/metabolism
- Signal Transduction/drug effects
- Squamous Cell Carcinoma of Head and Neck/drug therapy
- Squamous Cell Carcinoma of Head and Neck/metabolism
- Squamous Cell Carcinoma of Head and Neck/pathology
- Transplantation, Heterologous
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
|
49
|
Xu P, Zhao Y, Liu K, Lin S, Liu X, Wang M, Yang P, Tian T, Zhu YY, Dai Z. Prognostic role and clinical significance of trophoblast cell surface antigen 2 in various carcinomas. Cancer Manag Res 2017; 9:821-837. [PMID: 29276405 PMCID: PMC5731441 DOI: 10.2147/cmar.s147033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Trophoblast cell surface antigen 2 (TROP2) has been linked to disease prognosis in various human cancers and plays a critical role in tumor development, progression, and metastasis. A number of relevant studies have been published on this topic. A meta-analysis of the latest literature to evaluate the value of TROP2 as a predictive prognosticator of cancer was performed. METHODS Several online databases were searched, and relevant articles were retrieved. Overall and subcategory meta-analyses were performed, and results were collated. RESULTS Twenty-seven articles, including 29 studies, were included, involving 4,852 cancer patients, and results showed that the above-baseline expression of TROP2 was significantly associated with poorer overall survival (OS) (pooled hazard ratio [HR]: 1.84, 95% confidence interval [CI]: 1.45-2.35), disease-free survival (DFS) (pooled HR: 2.77, 95% CI: 1.73-4.42), and progression-free survival (PFS) (pooled HR: 1.71, 95% CI: 1.25-2.35). The following clinical characteristics were also significantly linked with TROP2 overexpression: moderate/poor differentiation (pooled HR: 3.03, 95% CI: 1.99-4.63), distant metastasis (pooled HR: 2.46, 95% CI: 1.05-5.75), lymph node metastasis (pooled HR: 2.47, 95%: CI 1.72-3.56), and advanced TNM stage (pooled HR: 2.02, 95% CI: 1.38-2.95). CONCLUSION TROP2 overexpression was predictive of poor prognosis in human cancers and may be an independent prognostic predictive biomarker. Further studies should be performed to confirm the significance of TROP2 in clinical practice.
Collapse
Affiliation(s)
- Peng Xu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yang Zhao
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Kang Liu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xinghan Liu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Pengtao Yang
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Tian Tian
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yu-yao Zhu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
50
|
Conti A, D’Elia C, Cheng M, Santoni M, Piva F, Brunelli M, Lopez-Beltran A, Giulietti M, Scarpelli M, Pycha A, Galosi AB, Artibani W, Cheng L, Montironi R, Battelli N, Lusuardi L. Oligometastases in Genitourinary Tumors: Recent Insights and Future Molecular Diagnostic Approach. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.eursup.2017.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|