1
|
Xu Y, Lih TM, De Marzo AM, Li QK, Zhang H. SPOT: spatial proteomics through on-site tissue-protein-labeling. Clin Proteomics 2024; 21:60. [PMID: 39443867 PMCID: PMC11515502 DOI: 10.1186/s12014-024-09505-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/22/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Spatial proteomics seeks to understand the spatial organization of proteins in tissues or at different subcellular localization in their native environment. However, capturing the spatial organization of proteins is challenging. Here, we present an innovative approach termed Spatial Proteomics through On-site Tissue-protein-labeling (SPOT), which combines the direct labeling of tissue proteins in situ on a slide and quantitative mass spectrometry for the profiling of spatially-resolved proteomics. MATERIALS AND METHODS Efficacy of direct TMT labeling was investigated using seven types of sagittal mouse brain slides, including frozen tissues without staining, formalin-fixed paraffin-embedded (FFPE) tissues without staining, deparaffinized FFPE tissues, deparaffinized and decrosslinked FFPE tissues, and tissues with hematoxylin & eosin (H&E) staining, hematoxylin (H) staining, eosin (E) staining. The ability of SPOT to profile proteomes at a spatial resolution was further evaluated on a horizontal mouse brain slide with direct TMT labeling at eight different mouse brain regions. Finally, SPOT was applied to human prostate cancer tissues as well as a tissue microarray (TMA), where TMT tags were meticulously applied to confined regions based on the pathological annotations. After on-site direct tissue-protein-labeling, tissues were scraped off the slides and subject to standard TMT-based quantitative proteomics analysis. RESULTS Tissue proteins on different types of mouse brain slides could be directly labeled with TMT tags. Moreover, the versatility of our direct-labeling approach extended to discerning specific mouse brain regions based on quantitative outcomes. The SPOT was further applied on both frozen tissues on slides and FFPE tissues on TMAs from prostate cancer tissues, where a distinct proteomic profile was observed among the regions with different Gleason scores. CONCLUSIONS SPOT is a robust and versatile technique that allows comprehensive profiling of spatially-resolved proteomics across diverse types of tissue slides to advance our understanding of intricate molecular landscapes.
Collapse
Affiliation(s)
- Yuanwei Xu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - T Mamie Lih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qing Kay Li
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Medical Institutions, Baltimore, MD, USA.
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Kilercik M, Özgür E, Şahin Ş, Şen Doğan B, Mutlu E, Cihan C, Kolay M, Erkal N, Zorlu Ö, Doğanca TS, Kural AR, Tüfek İ, Külah H. Detection of circulating tumor cells in non-metastatic prostate cancer through integration of a microfluidic CTC enrichment system and multiparametric flow cytometry. PLoS One 2024; 19:e0312296. [PMID: 39441869 PMCID: PMC11498670 DOI: 10.1371/journal.pone.0312296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Prostate cancer (PCa) is the second most common cancer among men and the fifth leading cause of cancer death. Circulating tumor cell (CTC) enumeration and characterisation in PCa has been shown to provide valuable information on prognosis of disease, therapy management and detection of resistance. Here, Cellsway's microfluidic platform for high-throughput enrichment of intact CTC populations was used to isolate CTCs from the blood of 20 localised PCa patients and 10 healthy donor samples to evaluate the clinical performance of the technology. To enumerate and characterise CTCs, a multi-parameter flow cytometry analysis was performed on the enriched CTC suspension using CTC-specific biomarkers. CTCs were detected in 17 of 20 patient samples, which corresponds to 85% CTC positivity. The median CTC count per 7.5 ml blood was 2 (1-9). In 80% of patients (n = 16), the number of CTCs ranged from 1 to 5, and in 5% of patients (n = 1) the number of CTCs was above 5. No CTCs were observed in the blood samples of 10 healthy volunteers, demonstrating the high specificity and low risk of false positives of the technology.
Collapse
Affiliation(s)
- Meltem Kilercik
- Department of Biochemistry, Acibadem University, Istanbul, Turkiye
| | - Ebru Özgür
- Mikro Biyosistemler A.S., Ankara, Turkiye
| | | | | | - Ege Mutlu
- Mikro Biyosistemler A.S., Ankara, Turkiye
| | - Cenay Cihan
- Acıbadem Labmed Clinical Laboratories, Istanbul, Turkiye
| | - Murat Kolay
- Acıbadem Labmed Clinical Laboratories, Istanbul, Turkiye
| | | | - Özge Zorlu
- Mikro Biyosistemler A.S., Ankara, Turkiye
| | | | - Ali Rıza Kural
- Department of Urology, Acibadem University, Istanbul, Turkiye
| | - İlter Tüfek
- Department of Urology, Acibadem University, Istanbul, Turkiye
| | - Haluk Külah
- Mikro Biyosistemler A.S., Ankara, Turkiye
- Department of Electrical and Electronics Engineering, Middle East Technical University, Ankara, Turkiye
| |
Collapse
|
3
|
Xiao D, Xiong M, Wang X, Lyu M, Sun H, Cui Y, Chen C, Jiang Z, Sun F. Regulation of the Function and Expression of EpCAM. Biomedicines 2024; 12:1129. [PMID: 38791091 PMCID: PMC11117676 DOI: 10.3390/biomedicines12051129] [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: 04/09/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) is a single transmembrane protein on the cell surface. Given its strong expression on epithelial cells and epithelial cell-derived tumors, EpCAM has been identified as a biomarker for circulating tumor cells (CTCs) and exosomes and a target for cancer therapy. As a cell adhesion molecule, EpCAM has a crystal structure that indicates that it forms a cis-dimer first and then probably a trans-tetramer to mediate intercellular adhesion. Through regulated intramembrane proteolysis (RIP), EpCAM and its proteolytic fragments are also able to regulate multiple signaling pathways, Wnt signaling in particular. Although great progress has been made, increasingly more findings have revealed the context-specific expression and function patterns of EpCAM and their regulation processes, which necessitates further studies to determine the structure, function, and expression of EpCAM under both physiological and pathological conditions, broadening its application in basic and translational cancer research.
Collapse
Affiliation(s)
- Di Xiao
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Mingrui Xiong
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xin Wang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Mengqing Lyu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Hanxiang Sun
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Yeting Cui
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Chen Chen
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Ziyu Jiang
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Fan Sun
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430081, China; (D.X.); (M.X.); (X.W.); (M.L.); (H.S.); (Y.C.)
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
4
|
Gu R, Kim TD, Song H, Sui Y, Shin S, Oh S, Janknecht R. SET7/9-mediated methylation affects oncogenic functions of histone demethylase JMJD2A. JCI Insight 2023; 8:e164990. [PMID: 37870957 PMCID: PMC10619491 DOI: 10.1172/jci.insight.164990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/05/2023] [Indexed: 10/25/2023] Open
Abstract
The histone demethylase JMJD2A/KDM4A facilitates prostate cancer development, yet how JMJD2A function is regulated has remained elusive. Here, we demonstrate that SET7/9-mediated methylation on 6 lysine residues modulated JMJD2A. Joint mutation of these lysine residues suppressed JMJD2A's ability to stimulate the MMP1 matrix metallopeptidase promoter upon recruitment by the ETV1 transcription factor. Mutation of just 3 methylation sites (K505, K506, and K507) to arginine residues (3xR mutation) was sufficient to maximally reduce JMJD2A transcriptional activity and also decreased its binding to ETV1. Introduction of the 3xR mutation into DU145 prostate cancer cells reduced in vitro growth and invasion and also severely compromised tumorigenesis. Consistently, the 3xR genotype caused transcriptome changes related to cell proliferation and invasion pathways, including downregulation of MMP1 and the NPM3 nucleophosmin/nucleoplasmin gene. NPM3 downregulation phenocopied and its overexpression rescued, to a large degree, the 3xR mutation in DU145 cells, suggesting that NPM3 was a seminal downstream effector of methylated JMJD2A. Moreover, we found that NPM3 was overexpressed in prostate cancer and might be indicative of disease aggressiveness. SET7/9-mediated lysine methylation of JMJD2A may aggravate prostate tumorigenesis in a manner dependent on NPM3, implying that the SET7/9→JMJD2A→NPM3 axis could be targeted for therapy.
Collapse
Affiliation(s)
| | | | | | | | - Sook Shin
- Department of Cell Biology
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Sangphil Oh
- Department of Cell Biology
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Ralf Janknecht
- Department of Cell Biology
- Department of Pathology, and
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
5
|
Zaarour RF, Ribeiro M, Azzarone B, Kapoor S, Chouaib S. Tumor microenvironment-induced tumor cell plasticity: relationship with hypoxic stress and impact on tumor resistance. Front Oncol 2023; 13:1222575. [PMID: 37886168 PMCID: PMC10598765 DOI: 10.3389/fonc.2023.1222575] [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: 05/14/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The role of tumor interaction with stromal components during carcinogenesis is crucial for the design of efficient cancer treatment approaches. It is widely admitted that tumor hypoxic stress is associated with tumor aggressiveness and thus impacts susceptibility and resistance to different types of treatments. Notable biological processes that hypoxia functions in include its regulation of tumor heterogeneity and plasticity. While hypoxia has been reported as a major player in tumor survival and dissemination regulation, the significance of hypoxia inducible factors in cancer stem cell development remains poorly understood. Several reports indicate that the emergence of cancer stem cells in addition to their phenotype and function within a hypoxic tumor microenvironment impacts cancer progression. In this respect, evidence showed that cancer stem cells are key elements of intratumoral heterogeneity and more importantly are responsible for tumor relapse and escape to treatments. This paper briefly reviews our current knowledge of the interaction between tumor hypoxic stress and its role in stemness acquisition and maintenance. Our review extensively covers the influence of hypoxia on the formation and maintenance of cancer stem cells and discusses the potential of targeting hypoxia-induced alterations in the expression and function of the so far known stem cell markers in cancer therapy approaches. We believe that a better and integrated understanding of the effect of hypoxia on stemness during carcinogenesis might lead to new strategies for exploiting hypoxia-associated pathways and their targeting in the clinical setting in order to overcome resistance mechanisms. More importantly, at the present time, efforts are oriented towards the design of innovative therapeutical approaches that specifically target cancer stem cells.
Collapse
Affiliation(s)
- RF. Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - M. Ribeiro
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - B. Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - S. Kapoor
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - S. Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, Villejuif, France
| |
Collapse
|
6
|
Voss G, Cassidy JR, Ceder Y. Functional consequences of A-to-I editing of miR-379 in prostate cancer cells. Sci Rep 2023; 13:16602. [PMID: 37789115 PMCID: PMC10547749 DOI: 10.1038/s41598-023-43775-7] [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: 03/22/2023] [Accepted: 09/28/2023] [Indexed: 10/05/2023] Open
Abstract
Prostate cancer is the predominant cause of cancer in men, but there is still a lack of biomarkers and treatments for metastatic spread. The initial promise of microRNAs to provide avenues to solve these problems has been dampened by the realisation that microRNAs co-exist in multiple functionally distinct isoforms, for example due to A-to-I editing. We recently found that A-to-I-editing of microRNA-379 (miR-379) was associated with prostate cancer, and that only the unedited isoform was negatively correlated with aggressive disease. Here, we set out to decipher the biological effects of unedited and edited miR-379 in prostate cancer cells. After transfection of four different prostate cancer cell lines with isoform-specific miR-379 mimics, we performed assays for cell growth, colony formation, migration, cell-cell adhesion, and analysed epithelial-mesenchymal transition (EMT) and stemness markers. We found that unedited miR-379 affected cell growth, with a promoting function in androgen receptor (AR)-negative cells and an inhibiting effect in AR-positive cells. This is supported by our in silico analysis that found unedited miR-379 targets are predicted to be predominantly involved in cellular proliferation whereas the targets of edited miR-379 are not. We further found that both miR-379 isoforms could promote colony formation, migration, and cell-cell adhesion. Overall, our data suggests that editing of miR-379 attenuates the growth-suppressive function of unedited miR-379 in androgen-sensitive prostate cancer cells, thereby promoting tumor growth.
Collapse
Affiliation(s)
- Gjendine Voss
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - James R Cassidy
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Yvonne Ceder
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
7
|
Sonam Dongsar T, Tsering Dongsar T, Molugulu N, Annadurai S, Wahab S, Gupta N, Kesharwani P. Targeted therapy of breast tumor by PLGA-based nanostructures: The versatile function in doxorubicin delivery. ENVIRONMENTAL RESEARCH 2023; 233:116455. [PMID: 37356522 DOI: 10.1016/j.envres.2023.116455] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/15/2023] [Accepted: 06/17/2023] [Indexed: 06/27/2023]
Abstract
Breast carcinoma is a molecularly diverse illness, and it is among the most prominent and often reported malignancies in female across the globe. Surgical intervention, chemotherapy, immunotherapy, gene therapy, and endocrine treatment are among the currently viable treatment options for the carcinoma of breast. Chemotherapy is among the most prevalent cancer management strategy. Doxorubicin (DOX) widely employed as a cytostatic medication for the treatment of a variety of malignancies. Despite its widespread acceptance and excellent efficacy against an extensive line up of neoplasia, it has a variety of shortcomings that limit its therapeutic potential in the previously mentioned indications. Employment of nanoparticulate systems has come up as a unique chemo medication delivery strategy and are being considerably explored for the amelioration of breast carcinoma. Polylactic-co-glycolic acid (PLGA)-based nano systems are being utilized in a number of areas within the medical research and medication delivery constitutes one of the primary functions for PLGA given their inherent physiochemical attributes, including their aqueous solubility, biocompatibility, biodegradability, versatility in formulation, and limited toxicity. Herein along with the different application of PLGA-based nano formulations in cancer therapy, the present review intends to describe the various research investigations that have been conducted to enumerate the effectiveness of DOX-encapsulated PLGA nanoparticles (DOX-PLGA NPs) as a feasible treatment option for breast cancer.
Collapse
Affiliation(s)
- Tenzin Sonam Dongsar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Tenzin Tsering Dongsar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nagashekhara Molugulu
- School of Pharmacy, Monash University, Bandar Sunway, Jalan Lagoon Selatan, 47500, Malaysia
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Neelima Gupta
- Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
8
|
Rashid K, Ahmad A, Meerasa SS, Khan AQ, Wu X, Liang L, Cui Y, Liu T. Cancer stem cell-derived exosome-induced metastatic cancer: An orchestra within the tumor microenvironment. Biochimie 2023; 212:1-11. [PMID: 37011805 DOI: 10.1016/j.biochi.2023.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/20/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Although the mechanisms as well as pathways associated with cancer stem cell (CSC) maintenance, expansion, and tumorigenicity have been extensively studied and the role of tumor cell (TC)-derived exosomes in this process is well understood, there is a paucity of research focusing specifically on the functional mechanisms of CSC-derived exosomes (CSC-Exo)/-exosomal-ncRNAs and their impact on malignancy. This shortcoming needs to be addressed, given that these vesicular and molecular components of CSCs could have a great impact on the cancer initiation, progression, and recurrence through their interaction with other key tumor microenvironment (TME) components, such as MSCs/MSC-Exo and CAFs/CAF-Exo. In particular, understanding CSCs/CSC-Exo and its crosstalk with MSCs/MSC-Exo or CAFs/CAF-Exo that are associated with the proliferation, migration, differentiation, angiogenesis, and metastasis through an enhanced process of self-renewal, chemotherapy as well as radiotherapy resistance may aid cancer treatment. This review contributes to this endeavor by summarizing the characteristic features and functional mechanisms of CSC-Exo/MSC-Exo/CAF-Exo and their mutual impact on cancer progression and therapy resistance.
Collapse
Affiliation(s)
- Khalid Rashid
- Department of Cancer Biology, Faculty of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Aqeel Ahmad
- Department of Medical Biochemistry, College of Medicine, Shaqra University, Shaqra, Saudi Arabia.
| | - Semmal Syed Meerasa
- Department of Physiology, College of Medicine, Shaqra University, Shaqra, Saudi Arabia
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Xiaobo Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Liang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Mirzaei R, Shafiee S, Vafaei R, Salehi M, Jalili N, Nazerian Z, Muhammadnajad A, Yadegari F, Reza Esmailinejad M, Farahmand L. Production of novel recombinant anti-EpCAM antibody as targeted therapy for breast cancer. Int Immunopharmacol 2023; 122:110656. [PMID: 37473710 DOI: 10.1016/j.intimp.2023.110656] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND The utilization of monoclonal antibodies (moAbs), an issue correlated with the biopharmaceutical professions, is developing and maturing. Coordinated with this conception, we produced the appealingly modeled anti-EpCAM scFv for breast cancer tumors. METHODS Afterward cloning and expression of recombinant antibody in Escherichia coli bacteria, the correctness of the desired antibody was checked by western blotting. Flow cytometry was utilized to determine the capacity of the recombinant antibody to append to the desired receptors in the malignant breast cancer (BC)cell line. The recombinant antibody (anti-EpCAM scFv) was examined for preclinical efficacy in reducing tumor growth, angiogenesis, and invasiveness (in vitro- in vivo). FINDINGS A target antibody-mediated attenuation of migration and invasion in the examined cancer cell lines was substantiated (P-value < 0.05). Grafted tumors from breast cancer in mice indicated significant and compelling suppression of tumor growth and decrement in blood supply in reaction to the recombinant anti-EpCAM intervention. Evaluations of immunohistochemical and histopathological findings revealed an enhanced response rate to the treatment. CONCLUSION The desired anti-EpCAM scFv can be a therapeutic tool to reduce invasion and proliferation in malignant breast cancer.
Collapse
Affiliation(s)
- Roya Mirzaei
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Soodabeh Shafiee
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Rana Vafaei
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Malihe Salehi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Neda Jalili
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Zahra Nazerian
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Ahad Muhammadnajad
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Yadegari
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohamad Reza Esmailinejad
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
10
|
He M, Zhang D, Cao Y, Chi C, Zeng Z, Yang X, Yang G, Sharma K, Hu K, Enikeev M. Chimeric antigen receptor-modified T cells therapy in prostate cancer: A comprehensive review on the current state and prospects. Heliyon 2023; 9:e19147. [DOI: https:/doi.org/10.1016/j.heliyon.2023.e19147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
|
11
|
He M, Zhang D, Cao Y, Chi C, Zeng Z, Yang X, Yang G, Sharma K, Hu K, Enikeev M. Chimeric antigen receptor-modified T cells therapy in prostate cancer: A comprehensive review on the current state and prospects. Heliyon 2023; 9:e19147. [PMID: 37664750 PMCID: PMC10469587 DOI: 10.1016/j.heliyon.2023.e19147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023] Open
Abstract
Recent immunotherapy research has focused on chimeric antigen receptor-modified T cells (CAR-Ts). CAR-T therapies have been clinically applied to manage hematologic malignancies with satisfactory effectiveness. However, the application of CAR-T immunotherapy in solid tumors remains challenging. Even so, current CAR-T immunotherapies for prostate cancer (PCa) have shown some promise, giving hope to patients with advanced metastatic PCa. This review aimed to elucidate different types of prostate tumor-associated antigen targets, such as prostate-specific membrane antigen and prostate stem cell antigen, and their effects. The current status of the corresponding targets in clinical research through their applications was also discussed. To improve the efficacy of CAR-T immunotherapy, we addressed the possible applications of multimodal immunotherapy, chemotherapy, and CAR-T combined therapies. The obstacles of solid tumors were concisely elaborated. Further studies should aim to discover novel potential targets and establish new models by overcoming the inherent barriers of solid tumors, such as tumor heterogeneity and the immunosuppressive nature of the tumor microenvironment.
Collapse
Affiliation(s)
- Mingze He
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia
| | - Dongqi Zhang
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), 130000, Changchun, China
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Changliang Chi
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), 130000, Changchun, China
| | - Zitong Zeng
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Guodong Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Kritika Sharma
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Kebang Hu
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), 130000, Changchun, China
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia
| |
Collapse
|
12
|
Padmyastuti A, Sarmiento MG, Dib M, Ehrhardt J, Schoon J, Somova M, Burchardt M, Roennau C, Pinto PC. Microfluidic-based prostate cancer model for investigating the secretion of prostate-specific antigen and microRNAs in vitro. Sci Rep 2023; 13:11623. [PMID: 37468746 DOI: 10.1038/s41598-023-38834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/16/2023] [Indexed: 07/21/2023] Open
Abstract
The study of prostate cancer in vitro relies on established cell lines that lack important physiological characteristics, such as proper polarization and expression of relevant biomarkers. Microphysiological systems (MPS) can replicate cancer microenvironments and lead to cellular phenotypic changes that better represent organ physiology in vitro. In this study, we developed an MPS model comprising conventional prostate cancer cells to evaluate their activity under dynamic culture conditions. Androgen-sensitive (LNCaP) and androgen-insensitive (PC3) cells were grown in conventional and 3D cultures, both static and dynamic. Cell morphology, the secretion of prostate-specific antigen, and the expression of key prostate markers and microRNAs were analyzed. LNCaP formed spheroids in 3D and MPS cultures, with morphological changes supported by the upregulation of cytokeratins and adhesion proteins. LNCaP also maintained a constant prostate-specific antigen secretion in MPS. PC3 cells did not develop complex structures in 3D and MPS cultures. PSA expression at the gene level was downregulated in LNCaP-MPS and considerably upregulated in PC3-MPS. MicroRNA expression was altered by the 3D static and dynamic culture, both intra- and extracellularly. MicroRNAs associated with prostate cancer progression were mostly upregulated in LNCaP-MPS. Overall dynamic cell culture substantially altered the morphology and expression of LNCaP cells, arguably augmenting their prostate cancer phenotype. This novel approach demonstrates that microRNA expression in prostate cancer cells is sensitive to external stimuli and that MPS can effectively promote important physiological changes in conventional prostate cancer models.
Collapse
Affiliation(s)
- Adventina Padmyastuti
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Marina Garcia Sarmiento
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Maria Dib
- Department of Ear, Nose and Throat Surgery, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Jens Ehrhardt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Janosch Schoon
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, Fleichmannstraße 8, 17475, Greifswald, Germany
| | - Maryna Somova
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Martin Burchardt
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Cindy Roennau
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - Pedro Caetano Pinto
- Department of Urology, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany.
| |
Collapse
|
13
|
Černe K, Kelhar N, Resnik N, Herzog M, Vodnik L, Veranič P, Kobal B. Characteristics of Extracellular Vesicles from a High-Grade Serous Ovarian Cancer Cell Line Derived from a Platinum-Resistant Patient as a Potential Tool for Aiding the Prediction of Responses to Chemotherapy. Pharmaceuticals (Basel) 2023; 16:907. [PMID: 37375854 DOI: 10.3390/ph16060907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Platinum-resistant high-grade serous ovarian cancer (HGSOC) is invariably a fatal disease. A central goal of ovarian cancer research is therefore to develop new strategies to overcome platinum resistance. Treatment is thus moving towards personalized therapy. However, validated molecular biomarkers that predict patients' risk of developing platinum resistance are still lacking. Extracellular vesicles (EVs) are promising candidate biomarkers. EpCAM-specific EVs are largely unexplored biomarkers for predicting chemoresistance. Using transmission electron microscopy, nanoparticle tracking analysis and flow cytometry, we compared the characteristics of EVs released from a cell line derived from a clinically confirmed cisplatin-resistant patient (OAW28) and EVs released from two cell lines from tumors sensitive to platinum-based chemotherapy (PEO1 and OAW42). We demonstrated that EVs released from the HGSOC cell line of chemoresistant patients exhibited greater size heterogeneity, a larger proportion of medium/large (>200 nm) Evs and a higher number of released EpCAM-positive EVs of different sizes, although the expression of EpCAM was predominant in EVs larger than 400 nm. We also found a strong positive correlation between the concentration of EpCAM-positive EVs and the expression of cellular EpCAM. These results may contribute to the prediction of platinum resistance in the future, although they should first be validated in clinical samples.
Collapse
Affiliation(s)
- Katarina Černe
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Nuša Kelhar
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Maruša Herzog
- Division of Gynecology and Obstetrics, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
- Department of Gynecology and Obstetrics, Faculty of Medicine, University Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Lana Vodnik
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Borut Kobal
- Division of Gynecology and Obstetrics, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
- Department of Gynecology and Obstetrics, Faculty of Medicine, University Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
14
|
Li YN, Li YY, Wang SX, Ma XY. Efficacy of Bispecific Antibody Targeting EpCAM and CD3 for Immunotherapy in Ovarian Cancer Ascites: An Experimental Study. Curr Med Sci 2023:10.1007/s11596-023-2753-2. [PMID: 37119369 DOI: 10.1007/s11596-023-2753-2] [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: 03/03/2023] [Accepted: 04/11/2023] [Indexed: 05/01/2023]
Abstract
OBJECTIVE This study aimed to explore the value of M701, targeting epithelial cell adhesion molecule (EpCAM) and CD3, in the immunotherapy of ovarian cancer ascites by the in vitro assay. METHODS The expression of EpCAM in ovarian cancer tissues was analyzed by databases. The EpCAM expression and immune cell infiltration in different foci of ovarian cancer were detected by 8-channel flow cytometry. The toxic effect of M701 on OVCAR3 was tested using the in vitro cytotoxicity assay. The 3D cell culture and drug intervention experiments were performed to evaluate the therapeutic effect of M701 in ovarian cancer specimens. Flow cytometry was used to examine the effect of M701 on the binding of immune cells to tumor cells and the activation capacity of T cells. RESULTS The results of the bioinformatic analysis showed that the expression of EpCAM in ovarian cancer tissue was significantly higher than that in normal ovarian tissue. The 8-channel flow cytometry of clinical samples showed that the EpCAM expression and lymphocyte infiltration were significantly heterogeneous among ovarian cancer patients and lesions at different sites. The in vitro experiment results showed that M701 had a significant killing effect on OVCAR3 cells. M701 also obviously killed primary tumor cells derived from some patients with ovarian cancer ascites. M701 could mediate the binding of CD3+ T cells to EpCAM+ tumor cells and induce T cell activation in a dose-dependent manner. CONCLUSION M701 showed significant inhibitory activity on tumor cells derived from ovarian cancer ascites, which had a promising application in immunotherapy for patients with ovarian cancer ascites.
Collapse
Affiliation(s)
- Yi-Nuo Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan-Yuan Li
- Department of Gynecology, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shi-Xuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiang-Yi Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Gogola S, Rejzer M, Bahmad HF, Alloush F, Omarzai Y, Poppiti R. Anti-Cancer Stem-Cell-Targeted Therapies in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15051621. [PMID: 36900412 PMCID: PMC10000420 DOI: 10.3390/cancers15051621] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Prostate cancer (PCa) is the second-most commonly diagnosed cancer in men around the world. It is treated using a risk stratification approach in accordance with the National Comprehensive Cancer Network (NCCN) in the United States. The main treatment options for early PCa include external beam radiation therapy (EBRT), brachytherapy, radical prostatectomy, active surveillance, or a combination approach. In those with advanced disease, androgen deprivation therapy (ADT) is considered as a first-line therapy. However, the majority of cases eventually progress while receiving ADT, leading to castration-resistant prostate cancer (CRPC). The near inevitable progression to CRPC has spurred the recent development of many novel medical treatments using targeted therapies. In this review, we outline the current landscape of stem-cell-targeted therapies for PCa, summarize their mechanisms of action, and discuss avenues of future development.
Collapse
Affiliation(s)
- Samantha Gogola
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Michael Rejzer
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Hisham F. Bahmad
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
- Correspondence: or ; Tel.: +1-305-674-2277
| | - Ferial Alloush
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Yumna Omarzai
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Robert Poppiti
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- The Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| |
Collapse
|
16
|
Castellón EA, Indo S, Contreras HR. Cancer Stemness/Epithelial-Mesenchymal Transition Axis Influences Metastasis and Castration Resistance in Prostate Cancer: Potential Therapeutic Target. Int J Mol Sci 2022; 23:ijms232314917. [PMID: 36499245 PMCID: PMC9736174 DOI: 10.3390/ijms232314917] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
Prostate cancer (PCa) is a leading cause of cancer death in men, worldwide. Mortality is highly related to metastasis and hormone resistance, but the molecular underlying mechanisms are poorly understood. We have studied the presence and role of cancer stem cells (CSCs) and the Epithelial-Mesenchymal transition (EMT) in PCa, using both in vitro and in vivo models, thereby providing evidence that the stemness-mesenchymal axis seems to be a critical process related to relapse, metastasis and resistance. These are complex and related processes that involve a cooperative action of different cancer cell subpopulations, in which CSCs and mesenchymal cancer cells (MCCs) would be responsible for invading, colonizing pre-metastatic niches, initiating metastasis and an evading treatments response. Manipulating the stemness-EMT axis genes on the androgen receptor (AR) may shed some light on the effect of this axis on metastasis and castration resistance in PCa. It is suggested that the EMT gene SNAI2/Slug up regulates the stemness gene Sox2, and vice versa, inducing AR expression, promoting metastasis and castration resistance. This approach will provide new sight about the role of the stemness-mesenchymal axis in the metastasis and resistance mechanisms in PCa and their potential control, contributing to develop new therapeutic strategies for patients with metastatic and castration-resistant PCa.
Collapse
Affiliation(s)
- Enrique A. Castellón
- Correspondence: (E.A.C.); (H.R.C.); Tel.: +56-229-786-863 (E.A.C.); +56-229-786-862 (H.R.C.)
| | | | - Héctor R. Contreras
- Correspondence: (E.A.C.); (H.R.C.); Tel.: +56-229-786-863 (E.A.C.); +56-229-786-862 (H.R.C.)
| |
Collapse
|
17
|
Zia S, Tehreem K, Batool S, Ishfaq M, Mirza SB, Khan S, Almashjary MN, Hazzazi MS, Qanash H, Shaikh A, Baty RS, Jafri I, Alsubhi NH, Alrefaei GI, Sami R, Shahid R. Epithelial Cell Adhesion Molecule ( EpCAM) Expression Can Be Modulated via NFκB. Biomedicines 2022; 10:biomedicines10112985. [PMID: 36428553 PMCID: PMC9687693 DOI: 10.3390/biomedicines10112985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) is considered an essential proliferation signature in cancer. In the current research study, qPCR induced expression of EpCAM was noted in acute lymphoblastic leukemia (ALL) cases. Costunolide, a sesquiterpene lactone found in crepe ginger and lettuce, is a medicinal herb with anticancer properties. Expression of EpCAM and its downstream target genes (Myc and TERT) wasdownregulated upon treatment with costunolide in Jurkat cells. A significant change in the telomere length of Jurkat cells was not noted at 72 h of costunolide treatment. An in silico study revealed hydrophobic interactions between EpCAM extracellular domain and Myc bHLH with costunolide. Reduced expression of NFκB, a transcription factor of EpCAM, Myc, and TERT in costunolide-treated Jurkat cells, suggested that costunolide inhibits gene expression by targeting NFκB and its downstream targets. Overall, the study proposes that costunolide could be a promising therapeutic biomolecule for leukemia.
Collapse
Affiliation(s)
- Saadiya Zia
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
- Department of Biochemistry, Faculty of Sciences, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Komal Tehreem
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Sidra Batool
- Research School of Chemistry, Australian National University, Canberra, ACT 2600, Australia
| | - Mehreen Ishfaq
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Shaher Bano Mirza
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Shahrukh Khan
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Majed N. Almashjary
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdul Aziz University, Jeddah 22254, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdul Aziz University, Jeddah 22254, Saudi Arabia
| | - Mohannad S. Hazzazi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdul Aziz University, Jeddah 22254, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdul Aziz University, Jeddah 22254, Saudi Arabia
| | - Husam Qanash
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail 55476, Saudi Arabia
- Molecular Diagnostics and Personalized Therapeutics Unit, University of Ha’il, Hail 55476, Saudi Arabia
| | - Ahmad Shaikh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia
| | - Roua S. Baty
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ibrahim Jafri
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Nouf H. Alsubhi
- Biological Sciences Department, College of Science and Arts, King Abdul Aziz University, Rabigh 21911, Saudi Arabia
| | - Ghadeer I. Alrefaei
- Department of Biology, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Saudi Arabia
| | - Rokayya Sami
- Department of Food Science and Nutrition, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ramla Shahid
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
- Correspondence:
| |
Collapse
|
18
|
Chandran E, Meininger L, Karzai F, Madan RA. Signaling new therapeutic opportunities: cytokines in prostate cancer. Expert Opin Biol Ther 2022; 22:1233-1243. [PMID: 35930001 DOI: 10.1080/14712598.2022.2108701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite FDA approval of sipuleucel-T in 2010, endeavors to use immune checkpoint inhibitors in unselected prostate cancer patients have not improved clinical outcomes. These efforts include studies with anti-PD1/PD-L1 and anti-CTLA-4 alone and in combination with existing standards of care. These strategies are generally T-cell centric and disregard the broader complex and pleiotropic components of the prostate cancer tumor microenvironment such as natural killer cells, myeloid-derived suppressor cells and tumor associated macrophages. AREAS COVERED We performed an online literature search and undertook a review of existing pre-clinical and clinical literature for cytokine-based therapy relating to prostate cancer, specifically on interleukin (IL)-2, IL-15, IL-12, IL-23, IL-8 and transforming growth factor (TGF)-β. EXPERT OPINION Cytokine-based therapies present an alternative immune strategy to target the pleiotropic prostate cancer tumor microenvironment beyond T-cells. Future immunotherapy strategies in prostate cancer should address these immune cell populations which may play more important roles in the prostate cancer tumor microenvironment.
Collapse
Affiliation(s)
- Elias Chandran
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luke Meininger
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fatima Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Kwizera EA, Ou W, Lee S, Stewart S, Shamul JG, Xu J, Tait N, Tkaczuk KHR, He X. Greatly Enhanced CTC Culture Enabled by Capturing CTC Heterogeneity Using a PEGylated PDMS-Titanium-Gold Electromicrofluidic Device with Glutathione-Controlled Gentle Cell Release. ACS NANO 2022; 16:11374-11391. [PMID: 35797466 PMCID: PMC9649890 DOI: 10.1021/acsnano.2c05195] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The circulating tumor cells (CTCs, the root cause of cancer metastasis and poor cancer prognosis) are very difficult to culture for scale-up in vitro, which has hampered their use in cancer research/prognosis and patient-specific therapeutic development. Herein, we report a robust electromicrofluidic chip for not only efficient capture of heterogeneous (EpCAM+ and CD44+) CTCs with high purity but also glutathione-controlled gentle release of the CTCs with high efficiency and viability. This is enabled by coating the polydimethylsiloxane (PDMS) surface in the device with a 10 nm gold layer through a 4 nm titanium coupling layer, for convenient PEGylation and linkage of capture antibodies via the thiol-gold chemistry. Surprisingly, the percentage of EpCAM+ mammary CTCs can be as low as ∼35% (∼70% on average), showing that the commonly used approach of capturing CTCs with EpCAM alone may miss many EpCAM- CTCs. Furthermore, the CD44+ CTCs can be cultured to form 3D spheroids efficiently for scale-up. In contrast, the CTCs captured with EpCAM alone are poor in proliferation in vitro, consistent with the literature. By capture of the CTC heterogeneity, the percentage of stage IV patients whose CTCs can be successfully cultured/scaled up is improved from 12.5% to 68.8%. These findings demonstrate that the common practice of CTC capture with EpCAM alone misses the CTC heterogeneity including the critical CD44+ CTCs. This study may be valuable to the procurement and scale-up of heterogeneous CTCs, to facilitate the understanding of cancer metastasis and the development of cancer metastasis-targeted personalized cancer therapies conveniently via the minimally invasive liquid/blood biopsy.
Collapse
Affiliation(s)
- Elyahb A Kwizera
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Sojeong Lee
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Jiangsheng Xu
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Nancy Tait
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| | - Katherine H R Tkaczuk
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland 21201, United States
| |
Collapse
|
20
|
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
|
21
|
Single-cell proteomics defines the cellular heterogeneity of localized prostate cancer. Cell Rep Med 2022; 3:100604. [PMID: 35492239 PMCID: PMC9044103 DOI: 10.1016/j.xcrm.2022.100604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/30/2021] [Accepted: 03/21/2022] [Indexed: 11/16/2022]
Abstract
Localized prostate cancer exhibits multiple genomic alterations and heterogeneity at the proteomic level. Single-cell technologies capture important cell-to-cell variability responsible for heterogeneity in biomarker expression that may be overlooked when molecular alterations are based on bulk tissue samples. This study aims to identify prognostic biomarkers and describe the heterogeneity of prostate cancer and the associated microenvironment by simultaneously quantifying 36 proteins using single-cell mass cytometry analysis of over 1.6 million cells from 58 men with localized prostate cancer. We perform this task, using a high-dimensional clustering pipeline named Franken to describe subpopulations of immune, stromal, and prostate cells, including changes occurring in tumor tissues and high-grade disease that provide insights into the coordinated progression of prostate cancer. Our results further indicate that men with localized disease already harbor rare subpopulations that typically occur in castration-resistant and metastatic disease.
Single-cell proteomics of localized prostate cancer defines disease heterogeneity Malignant and benign prostate tissues differ in rare cell-type proportional shifts T cells and proliferating macrophages are associated with high-grade PCa Rare CD15+ epithelial cells are amplified in high-grade PCa
Collapse
|
22
|
Xu T, Liu Y, Schulga A, Konovalova E, Deyev S, Tolmachev V, Vorobyeva A. Epithelial cell adhesion molecule‑targeting designed ankyrin repeat protein‑toxin fusion Ec1‑LoPE exhibits potent cytotoxic action in prostate cancer cells. Oncol Rep 2022; 47:94. [PMID: 35315504 PMCID: PMC8968790 DOI: 10.3892/or.2022.8305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Targeted anticancer therapeutics offer the advantage of reducing cytotoxic side effects to normal cells by directing the cytotoxic payload selectively to cancer cells. Designed ankyrin repeat proteins (DARPins) are promising non-immunoglobulin-based scaffold proteins for payload delivery to cancer-associated molecular targets. Epithelial cell adhesion molecule (EpCAM) is overexpressed in 40–60% of prostate cancers (PCs) and is associated with metastasis, increased risk of PC recurrence and resistance to treatment. Here, we investigated the use of DARPin Ec1 for targeted delivery of Pseudomonas exotoxin A variant (LoPE) with low immunogenicity and low non-specific toxicity to EpCAM-expressing prostate cancer cells. Ec1-LoPE fusion protein was radiolabeled with tricarbonyl technetium-99m and its binding specificity, binding kinetics, cellular processing, internalization and cytotoxicity were evaluated in PC-3 and DU145 cell lines. Ec1-LoPE showed EpCAM-specific binding to EpCAM-expressing prostate cancer cells. Rapid internalization mediated potent cytotoxic effect with picomolar IC50 values in both studied cell lines. Taken together, these data support further evaluation of Ec1-LoPE in a therapeutic setting in a prostate cancer model in vivo.
Collapse
Affiliation(s)
- Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin‑Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Sergey Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| |
Collapse
|
23
|
Bravo Vázquez LA, Moreno Becerril MY, Mora Hernández EO, de León Carmona GG, Aguirre Padilla ME, Chakraborty S, Bandyopadhyay A, Paul S. The Emerging Role of MicroRNAs in Bone Diseases and Their Therapeutic Potential. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010211. [PMID: 35011442 PMCID: PMC8746945 DOI: 10.3390/molecules27010211] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/16/2021] [Accepted: 12/24/2021] [Indexed: 01/24/2023]
Abstract
MicroRNAs (miRNAs) are a class of small (20-24 nucleotides), highly conserved, non-coding RNA molecules whose main function is the post-transcriptional regulation of gene expression through sequence-specific manners, such as mRNA degradation or translational repression. Since these key regulatory molecules are implicated in several biological processes, their altered expression affects the preservation of cellular homeostasis and leads to the development of a wide range of pathologies. Over the last few years, relevant investigations have elucidated that miRNAs participate in different stages of bone growth and development. Moreover, the abnormal expression of these RNA molecules in bone cells and tissues has been significantly associated with the progression of numerous bone diseases, including osteoporosis, osteosarcoma, osteonecrosis and bone metastasis, among others. In fact, miRNAs regulate multiple pathological mechanisms, including altering either osteogenic or osteoblast differentiation, metastasis, osteosarcoma cell proliferation, and bone loss. Therefore, in this present review, aiming to impulse the research arena of the biological implications of miRNA transcriptome in bone diseases and to explore their potentiality as a theragnostic target, we summarize the recent findings associated with the clinical significance of miRNAs in these ailments.
Collapse
Affiliation(s)
- Luis Alberto Bravo Vázquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico; (L.A.B.V.); (M.Y.M.B.); (G.G.d.L.C.); (M.E.A.P.)
| | - Mariana Yunuen Moreno Becerril
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico; (L.A.B.V.); (M.Y.M.B.); (G.G.d.L.C.); (M.E.A.P.)
| | - Erick Octavio Mora Hernández
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Mexico City, Calle del Puente, No. 222 Col. Ejidos de Huipulco, Tlalpan, Mexico City 14380, Mexico;
| | - Gabriela García de León Carmona
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico; (L.A.B.V.); (M.Y.M.B.); (G.G.d.L.C.); (M.E.A.P.)
| | - María Emilia Aguirre Padilla
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico; (L.A.B.V.); (M.Y.M.B.); (G.G.d.L.C.); (M.E.A.P.)
| | - Samik Chakraborty
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Anindya Bandyopadhyay
- International Rice Research Institute, Manila 4031, Philippines;
- Reliance Industries Ltd., Navi Mumbai 400701, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Querétaro, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico; (L.A.B.V.); (M.Y.M.B.); (G.G.d.L.C.); (M.E.A.P.)
- Correspondence:
| |
Collapse
|
24
|
Nastały P, Smentoch J, Popęda M, Martini E, Maiuri P, Żaczek AJ, Sowa M, Matuszewski M, Szade J, Kalinowski L, Niemira M, Brandt B, Eltze E, Semjonow A, Bednarz-Knoll N. Low Tumor-to-Stroma Ratio Reflects Protective Role of Stroma against Prostate Cancer Progression. J Pers Med 2021; 11:1088. [PMID: 34834440 PMCID: PMC8622253 DOI: 10.3390/jpm11111088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/09/2022] Open
Abstract
Tumor-to-stroma ratio (TSR) is a prognostic factor that expresses the relative amounts of tumor and intratumoral stroma. In this study, its clinical and molecular relevance was evaluated in prostate cancer (PCa). The feasibility of automated quantification was tested in digital scans of tissue microarrays containing 128 primary tumors from 72 PCa patients stained immunohistochemically for epithelial cell adhesion molecule (EpCAM), followed by validation in a cohort of 310 primary tumors from 209 PCa patients. In order to investigate the gene expression differences between tumors with low and high TSR, we applied multigene expression analysis (nCounter® PanCancer Progression Panel, NanoString) of 42 tissue samples. TSR scores were categorized into low (<1 TSR) and high (≥1 TSR). In the pilot cohort, 31 patients (43.1%) were categorized as low and 41 (56.9%) as high TSR score, whereas 48 (23.0%) patients from the validation cohort were classified as low TSR and 161 (77.0%) as high. In both cohorts, high TSR appeared to indicate the shorter time to biochemical recurrence in PCa patients (Log-rank test, p = 0.04 and p = 0.01 for the pilot and validation cohort, respectively). Additionally, in the multivariate analysis of the validation cohort, TSR predicted BR independent of other factors, i.e., pT, pN, and age (p = 0.04, HR 2.75, 95%CI 1.07-7.03). Our data revealed that tumors categorized into low and high TSR score show differential expression of various genes; the genes upregulated in tumors with low TSR score were mostly associated with extracellular matrix and cell adhesion regulation. Taken together, this study shows that high stroma content can play a protective role in PCa. Automatic EpCAM-based quantification of TSR might improve prognostication in personalized medicine for PCa.
Collapse
Affiliation(s)
- Paulina Nastały
- Laboratory of Translational Oncology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (P.N.); (J.S.); (M.P.); (A.J.Ż.)
- FIRC (Italian Foundation for Cancer Research), Institute of Molecular Oncology (IFOM), 20139 Milan, Italy; (E.M.); (P.M.)
| | - Julia Smentoch
- Laboratory of Translational Oncology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (P.N.); (J.S.); (M.P.); (A.J.Ż.)
| | - Marta Popęda
- Laboratory of Translational Oncology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (P.N.); (J.S.); (M.P.); (A.J.Ż.)
| | - Emanuele Martini
- FIRC (Italian Foundation for Cancer Research), Institute of Molecular Oncology (IFOM), 20139 Milan, Italy; (E.M.); (P.M.)
| | - Paolo Maiuri
- FIRC (Italian Foundation for Cancer Research), Institute of Molecular Oncology (IFOM), 20139 Milan, Italy; (E.M.); (P.M.)
| | - Anna J. Żaczek
- Laboratory of Translational Oncology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (P.N.); (J.S.); (M.P.); (A.J.Ż.)
| | - Marek Sowa
- Department of Urology, Medical University of Gdańsk, 80-214 Gdańsk, Poland; (M.S.); (M.M.)
| | - Marcin Matuszewski
- Department of Urology, Medical University of Gdańsk, 80-214 Gdańsk, Poland; (M.S.); (M.M.)
| | - Jolanta Szade
- Department of Pathomorphology, Medical University of Gdańsk, 80-214 Gdańsk, Poland;
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Biobank, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
- Biobanking and Biomolecular Resources Research Infrastructure (BBMRI.pl), 80-214 Gdańsk, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Burkhard Brandt
- Institute of Clinical Chemistry, University Medical Centre Schleswig-Holstein, 24105 Kiel, Germany;
| | - Elke Eltze
- Institute of Pathology Saarbruecken-Rastpfuhl, 66113 Saarbruecken, Germany;
| | - Axel Semjonow
- Department of Urology, Prostate Center, University Clinic Münster, 48149 Münster, Germany;
| | - Natalia Bednarz-Knoll
- Laboratory of Translational Oncology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (P.N.); (J.S.); (M.P.); (A.J.Ż.)
| |
Collapse
|
25
|
Deyev SM, Xu T, Liu Y, Schulga A, Konovalova E, Garousi J, Rinne SS, Larkina M, Ding H, Gräslund T, Orlova A, Tolmachev V, Vorobyeva A. Influence of the Position and Composition of Radiometals and Radioiodine Labels on Imaging of Epcam Expression in Prostate Cancer Model Using the DARPin Ec1. Cancers (Basel) 2021; 13:cancers13143589. [PMID: 34298801 PMCID: PMC8304184 DOI: 10.3390/cancers13143589] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Metastasis-targeting therapy might improve outcomes in oligometastatic prostate cancer. Epithelial cell adhesion molecule (EpCAM) is overexpressed in 40–60% of prostate cancer cases and might be used as a target for specific delivery of toxins and drugs. Radionuclide molecular imaging could enable non-invasive detection of EpCAM and stratification of patients for targeted therapy. Designed ankyrin repeat proteins (DARPins) are scaffold proteins, which can be selected for specific binding to different targets. The DARPin Ec1 binds strongly to EpCAM. To determine an optimal design of Ec1-based probes, we labeled Ec1 at two different positions with four different nuclides (68Ga, 111In, 57Co and 125I) and investigated the impact on Ec1 biodistribution. We found that the C-terminus is the best position for labeling and that 111In and 125I provide the best imaging contrast. This study might be helpful for scientists developing imaging probes based on scaffold proteins. Abstract The epithelial cell adhesion molecule (EpCAM) is intensively overexpressed in 40–60% of prostate cancer (PCa) cases and can be used as a target for the delivery of drugs and toxins. The designed ankyrin repeat protein (DARPin) Ec1 has a high affinity to EpCAM (68 pM) and a small size (18 kDa). Radiolabeled Ec1 might be used as a companion diagnostic for the selection of PCa patients for therapy. The study aimed to investigate the influence of radiolabel position (N- or C-terminal) and composition on the targeting and imaging properties of Ec1. Two variants, having an N- or C-terminal cysteine, were produced, site-specifically conjugated to a DOTA chelator and labeled with cobalt-57, gallium-68 or indium-111. Site-specific radioiodination was performed using ((4-hydroxyphenyl)-ethyl)maleimide (HPEM). Biodistribution of eight radiolabeled Ec1-probes was measured in nude mice bearing PCa DU145 xenografts. In all cases, positioning of a label at the C-terminus provided the best tumor-to-organ ratios. The non-residualizing [125I]I-HPEM label provided the highest tumor-to-muscle and tumor-to-bone ratios and is more suitable for EpCAM imaging in early-stage PCa. Among the radiometals, indium-111 provided the highest tumor-to-blood, tumor-to-lung and tumor-to-liver ratios and could be used at late-stage PCa. In conclusion, label position and composition are important for the DARPin Ec1.
Collapse
Affiliation(s)
- Sergey M. Deyev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
- Bio-Nanophotonic Lab., Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University “MEPhI”, 115409 Moscow, Russia
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
| | - Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
| | - Alexey Schulga
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Elena Konovalova
- Molecular Immunology Laboratory, Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
| | - Maria Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Pharmaceutical Analysis, Siberian State Medical University (SSMU), 2, Moscow Trakt, 634050 Tomsk, Russia
| | - Haozhong Ding
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Torbjörn Gräslund
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden; (H.D.); (T.G.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden;
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
- Correspondence:
| | - Anzhelika Vorobyeva
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (S.M.D.); (A.S.); (M.L.); (A.O.); (A.V.)
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.X.); (Y.L.); (J.G.)
| |
Collapse
|
26
|
Functional Implications of the Dynamic Regulation of EpCAM during Epithelial-to-Mesenchymal Transition. Biomolecules 2021; 11:biom11070956. [PMID: 34209658 PMCID: PMC8301972 DOI: 10.3390/biom11070956] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is a transmembrane glycoprotein expressed in epithelial tissues. EpCAM forms intercellular, homophilic adhesions, modulates epithelial junctional protein complex formation, and promotes epithelial tissue homeostasis. EpCAM is a target of molecular therapies and plays a prominent role in tumor biology. In this review, we focus on the dynamic regulation of EpCAM expression during epithelial-to-mesenchymal transition (EMT) and the functional implications of EpCAM expression on the regulation of EMT. EpCAM is frequently and highly expressed in epithelial cancers, while silenced in mesenchymal cancers. During EMT, EpCAM expression is downregulated by extracellular signal-regulated kinases (ERK) and EMT transcription factors, as well as by regulated intramembrane proteolysis (RIP). The functional impact of EpCAM expression on tumor biology is frequently dependent on the cancer type and predominant oncogenic signaling pathways, suggesting that the role of EpCAM in tumor biology and EMT is multifunctional. Membrane EpCAM is cleaved in cancers and its intracellular domain (EpICD) is transported into the nucleus and binds β-catenin, FHL2, and LEF1. This stimulates gene transcription that promotes growth, cancer stem cell properties, and EMT. EpCAM is also regulated by epidermal growth factor receptor (EGFR) signaling and the EpCAM ectoderm (EpEX) is an EGFR ligand that affects EMT. EpCAM is expressed on circulating tumor and cancer stem cells undergoing EMT and modulates metastases and cancer treatment responses. Future research exploring EpCAM’s role in EMT may reveal additional therapeutic opportunities.
Collapse
|
27
|
Zhang H, Lin X, Huang Y, Wang M, Cen C, Tang S, Dique MR, Cai L, Luis MA, Smollar J, Wan Y, Cai F. Detection Methods and Clinical Applications of Circulating Tumor Cells in Breast Cancer. Front Oncol 2021; 11:652253. [PMID: 34150621 PMCID: PMC8208079 DOI: 10.3389/fonc.2021.652253] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/29/2021] [Indexed: 12/18/2022] Open
Abstract
Circulating Tumor Cells (CTCs) are cancer cells that split away from the primary tumor and appear in the circulatory system as singular units or clusters, which was first reported by Dr. Thomas Ashworth in 1869. CTCs migrate and implantation occurs at a new site, in a process commonly known as tumor metastasis. In the case of breast cancer, the tumor cells often migrate into locations such as the lungs, brain, and bones, even during the early stages, and this is a notable characteristic of breast cancer. Survival rates have increased significantly over the past few decades because of progress made in radiology and tissue biopsy, making early detection and diagnosis of breast cancer possible. However, liquid biopsy, particularly that involving the collection of CTCs, is a non-invasive method to detect tumor cells in the circulatory system, which can be easily isolated from human plasma, serum, and other body fluids. Compared to traditional tissue biopsies, fluid sample collection has the advantages of being readily available and more acceptable to the patient. It can also detect tumor cells in blood earlier and in smaller numbers, possibly allowing for diagnosis prior to any tumor detection using imaging methods. Because of the scarcity of CTCs circulating in blood vessels (only a few CTCs among billions of erythrocytes and leukocytes), thorough but accurate detection methods are particularly important for further clinical applications.
Collapse
Affiliation(s)
- Hongyi Zhang
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Lin
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuan Huang
- Cellomics International Limited, Hong Kong, China
| | - Minghong Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Chunmei Cen
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shasha Tang
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Marcia R Dique
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Cai
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Manuel A Luis
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jillian Smollar
- Department of Biomedical Engineering, The City College of New York, New York, NY, United States
| | - Yuan Wan
- Department of Biomedical Engineering, The City College of New York, New York, NY, United States
| | - Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Agnoletto C, Caruso C, Garofalo C. Heterogeneous Circulating Tumor Cells in Sarcoma: Implication for Clinical Practice. Cancers (Basel) 2021; 13:cancers13092189. [PMID: 34063272 PMCID: PMC8124844 DOI: 10.3390/cancers13092189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The present review is aimed to discuss the relevance of assaying for the presence and isolation of circulating tumor cells (CTCs) in patients with sarcoma. Just a few studies have been performed to detect and enumerate viable CTCs in sarcoma and a majority of them still represent proof-of-concept studies, while more frequently tumor cells have been detected in the circulation by using the PCR-based method. Nevertheless, recent advances in technologies allowed detection of epithelial–mesenchymal transitioned CTCs from patients with mesenchymal malignancies, despite results being mostly preliminary. The possibility to identify CTCs holds a great promise for both applications of liquid biopsy in sarcoma for precision medicine, and for research purposes to pinpoint the mechanism of the metastatic process through the characterization of tumor mesenchymal cells. Coherently, clinical trials in sarcoma have been designed accordingly to detect CTCs, for diagnosis, identification of novel therapeutic targets and resistance mechanisms of systemic therapies, and patient stratification. Abstract Bone and soft tissue sarcomas (STSs) represent a group of heterogeneous rare malignant tumors of mesenchymal origin, with a poor prognosis. Due to their low incidence, only a few studies have been reported addressing circulating tumor cells (CTCs) in sarcoma, despite the well-documented relevance for applications of liquid biopsy in precision medicine. In the present review, the most recent data relative to the detection and isolation of viable and intact CTCs in these tumors will be reviewed, and the heterogeneity in CTCs will be discussed. The relevance of epithelial–mesenchymal plasticity and stemness in defining the phenotypic and functional properties of these rare cells in sarcoma will be highlighted. Of note, the existence of dynamic epithelial–mesenchymal transition (EMT)-related processes in sarcoma tumors has only recently been related to their clinical aggressiveness. Also, the presence of epithelial cell adhesion molecule (EpCAM)-positive CTC in sarcoma has been weakly correlated with poor outcome and disease progression, thus proving the existence of both epithelial and mesenchymal CTC in sarcoma. The advancement in technologies for capturing and enumerating all diverse CTCs phenotype originating from these mesenchymal tumors are presented, and results provide a promising basis for clinical application of CTC detection in sarcoma.
Collapse
|
29
|
Voss G, Haflidadóttir BS, Järemo H, Persson M, Catela Ivkovic T, Wikström P, Ceder Y. Regulation of cell-cell adhesion in prostate cancer cells by microRNA-96 through upregulation of E-Cadherin and EpCAM. Carcinogenesis 2021; 41:865-874. [PMID: 31738404 PMCID: PMC7359773 DOI: 10.1093/carcin/bgz191] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/24/2019] [Accepted: 11/15/2019] [Indexed: 11/15/2022] Open
Abstract
Prostate cancer is one of the most common cancers in men, yet the biology behind lethal disease progression and bone metastasis is poorly understood. In this study, we found elevated levels of microRNA-96 (miR-96) in prostate cancer bone metastasis samples. To determine the molecular mechanisms by which miR-96 deregulation contributes to metastatic progression, we performed an Argonaute2-immunoprecipitation assay, in which mRNAs associated with cell–cell interaction were enriched. The expression of two cell adhesion molecules, E-Cadherin and EpCAM, was upregulated by miR-96, and potential targets sites were identified in the coding sequences of their mRNAs. We further showed that miR-96 enhanced cell–cell adhesion between prostate cancer cells as well as their ability to bind to osteoblasts. Our findings suggest that increased levels of miR-96 give prostate cancer cells an advantage at forming metastases in the bone microenvironment due to increased cell–cell interaction. We propose that miR-96 promotes bone metastasis in prostate cancer patients by facilitating the outgrowth of macroscopic tumours in the bone.
Collapse
Affiliation(s)
- Gjendine Voss
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Helena Järemo
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | | | - Tina Catela Ivkovic
- Department of Laboratory Medicine, Lund University, Lund, Sweden.,Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | | | - Yvonne Ceder
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
30
|
Krause J, von Felden J, Casar C, Fründt TW, Galaski J, Schmidt C, Jung C, Ittrich H, Weidemann SA, Krech T, Heumann A, Li J, Fischer L, Sauter G, Lohse AW, Wege H, Schulze K. Hepatocellular carcinoma: Intratumoral EpCAM-positive cancer stem cell heterogeneity identifies high-risk tumor subtype. BMC Cancer 2020; 20:1130. [PMID: 33225916 PMCID: PMC7682021 DOI: 10.1186/s12885-020-07580-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The translational interest in the intratumoral heterogeneity of hepatocellular carcinoma (HCC) has been increasing. The dismal prognosis of this pathology is linked to the features of the HCC harbouring cancer stem cells (CSC), represented by EpCAM-expression. However, the extent of the impact of intratumoral distribution of CSC-features, both on the recurrence after curative resection and on clinical outcome, remains unknown. To address this, we investigated the spatial heterogeneity of CSC-features with the aim of identifying the unique HCC patient subgroups amenable to adjuvant treatment. METHODS We designed a tissue microarray (TMA) from patients who had received liver resection between 2011 and 2017. Tumor specimens were sampled at multiple locations (n = 3-8). EpCAM-positivity was assessed for intensity and proportion by applying a score dividing three groups: (i) negative (E-/-); (ii) heterogeneous (E-/+); and (iii) homogeneous (E+/+). The groups were further analysed with regard to time-to-recurrence (TTR) and recurrence-free-survival (RFS). RESULTS We included 314 tumor spots from 69 patients (76.8% male, median age 66, liver cirrhosis/fibrosis 75.8%). The risk factors were alcohol abuse (26.2%), NASH (13.1%), HBV (15.5%), HCV (17.9%) and others (27.4%), representative of a typical Western cohort. E+/+ patients experienced significantly shorter TTR and RFS compared to E+/- and E-/- patients (TTR 5 vs. 19 months, p = 0.022; RFS 5 vs. 14 vs. 21 months, p = 0.016). Only homogeneous EpCAM-positivity correlated with higher AFP levels (> 400 ng/ml, p = 0.031). CONCLUSIONS Spatial heterogeneity of EpCAM-expression was markedly present in the cohort. Of note, only homogeneous EpCAM-expression correlated significantly with early recurrence, whereas heterogeneous EpCAM-expression was associated with clinical endpoints comparable to EpCAM-negativity. We identified a unique HCC subtype associated with a high risk of tumor recurrence.
Collapse
Affiliation(s)
- Jenny Krause
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Johann von Felden
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Casar
- Bioinformatics Core, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thorben W Fründt
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Johanna Galaski
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Constantin Schmidt
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Caroline Jung
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Harald Ittrich
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Sören A Weidemann
- Department of Pathology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Till Krech
- Department of Pathology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Asmus Heumann
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Jun Li
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Lutz Fischer
- Department of Visceral Transplant Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Department of Pathology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Henning Wege
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kornelius Schulze
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
- Mildred Scheel Cancer Career Centre HaTriCS4, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
31
|
Gires O, Pan M, Schinke H, Canis M, Baeuerle PA. Expression and function of epithelial cell adhesion molecule EpCAM: where are we after 40 years? Cancer Metastasis Rev 2020; 39:969-987. [PMID: 32507912 PMCID: PMC7497325 DOI: 10.1007/s10555-020-09898-3] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
EpCAM (epithelial cell adhesion molecule) was discovered four decades ago as a tumor antigen on colorectal carcinomas. Owing to its frequent and high expression on carcinomas and their metastases, EpCAM serves as a prognostic marker, a therapeutic target, and an anchor molecule on circulating and disseminated tumor cells (CTCs/DTCs), which are considered the major source for metastatic cancer cells. Today, EpCAM is reckoned as a multi-functional transmembrane protein involved in the regulation of cell adhesion, proliferation, migration, stemness, and epithelial-to-mesenchymal transition (EMT) of carcinoma cells. To fulfill these functions, EpCAM is instrumental in intra- and intercellular signaling as a full-length molecule and following regulated intramembrane proteolysis, generating functionally active extra- and intracellular fragments. Intact EpCAM and its proteolytic fragments interact with claudins, CD44, E-cadherin, epidermal growth factor receptor (EGFR), and intracellular signaling components of the WNT and Ras/Raf pathways, respectively. This plethora of functions contributes to shaping intratumor heterogeneity and partial EMT, which are major determinants of the clinical outcome of carcinoma patients. EpCAM represents a marker for the epithelial status of primary and systemic tumor cells and emerges as a measure for the metastatic capacity of CTCs. Consequentially, EpCAM has reclaimed potential as a prognostic marker and target on primary and systemic tumor cells.
Collapse
Affiliation(s)
- Olivier Gires
- Department of Otorhinolaryngology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Clinical Cooperation Group "Personalized Radiotherapy in Head and Neck Cancer", Helmholtz Zentrum, Neuherberg, Germany.
| | - Min Pan
- Department of Otorhinolaryngology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Henrik Schinke
- Department of Otorhinolaryngology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Patrick A Baeuerle
- Institute for Immunology, LMU Munich, Grosshadernerstr. 9, 82152 Planegg, Martinsried, Germany
- MPM Capital, Cambridge MA, 450 Kendall Street, Cambridge, MA, 02142, USA
| |
Collapse
|
32
|
Noman ASM, Parag RR, Rashid MI, Islam S, Rahman MZ, Chowdhury AA, Sultana A, Jerin C, Siddiqua A, Rahman L, Nayeem J, Akther S, Baidya S, Shil RK, Rahman M, Shirin A, Mahmud R, Hossain SMI, Sumi SA, Chowdhury A, Basher SB, Hasan A, Bithy S, Aklima J, Chowdhury N, Hasan MN, Banu T, Chowdhury S, Hossain MM, Yeger H, Farhat WA, Islam SS. Chemotherapeutic resistance of head and neck squamous cell carcinoma is mediated by EpCAM induction driven by IL-6/p62 associated Nrf2-antioxidant pathway activation. Cell Death Dis 2020; 11:663. [PMID: 32814771 PMCID: PMC7438524 DOI: 10.1038/s41419-020-02907-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/24/2022]
Abstract
Overexpression of epithelial cell adhesion molecule (EpCAM) has been associated with chemotherapeutic resistance, leads to aggressive tumor behavior, and results in an adverse clinical outcome. The molecular mechanism by which EpCAM enrichment is linked to therapeutic resistance via Nrf2, a key regulator of antioxidant genes is unknown. We have investigated the link between EpCAM and the Nrf2 pathway in light of therapeutic resistance using head and neck squamous cell carcinoma (HNSCC) patient tumor samples and cell lines. We report that EpCAM was highly expressed in Nrf2-positive and HPV-negative HNSCC cells. In addition, cisplatin-resistant tumor cells consisted of a higher proportion of EpCAMhigh cells compared to the cisplatin sensitive counterpart. EpCAMhigh populations exhibited resistance to cisplatin, a higher efficiency in colony formation, sphere growth and invasion capacity, and demonstrated reduced reactive oxygen species (ROS) activity. Furthermore, Nrf2 expression was significantly higher in EpCAMhigh populations. Mechanistically, expression of Nrf2 and its target genes were most prominently observed in EpCAMhigh populations. Silencing of EpCAM expression resulted in the attenuation of expressions of Nrf2 and SOD1 concomitant with a reduction of Sox2 expression. On the other hand, silencing of Nrf2 expression rendered EpCAMhigh populations sensitive to cisplatin treatment accompanied by the inhibition of colony formation, sphere formation, and invasion efficiency and increased ROS activity. The molecular mechanistic link between EpCAM expression and activation of Nrf2 was found to be a concerted interaction of interleukin-6 (IL-6) and p62. Silencing of p62 expression in EpCAMhigh populations resulted in the attenuation of Nrf2 pathway activation suggesting that Nrf2 pathway activation promoted resistance to cisplatin in EpCAMhigh populations. We propose that therapeutic targeting the Nrf2-EpCAM axis might be an excellent approach to modulate stress resistance and thereby survival of HNSCC patients enriched in EpCAMhigh populations.
Collapse
Affiliation(s)
- Abu Shadat M Noman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh.,Department of Pathology, McGill University, Montreal, QC, Canada
| | - Rashed R Parag
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Muhammad I Rashid
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Shafiqul Islam
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Mohammad Z Rahman
- Department of Pathology, Chittagong Medical College Hospital, Chittagong, Bangladesh
| | - Ali A Chowdhury
- Department of Radiotherapy, Chittagong Medical College Hospital, Chittagong, Bangladesh
| | - Afrin Sultana
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Chandsultana Jerin
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Ayesha Siddiqua
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Lutfur Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Junayed Nayeem
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Sonam Akther
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Sunanda Baidya
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Rajib K Shil
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Mizanur Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh.,Department of Biochemistry, Rangamati Medical College, Rangamati, Bangladesh
| | - Afsana Shirin
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Reaz Mahmud
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - S M Ikram Hossain
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Sharmin A Sumi
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Arfina Chowdhury
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Shabnam B Basher
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Abul Hasan
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Shammy Bithy
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Jannatul Aklima
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Nabila Chowdhury
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Muhammad N Hasan
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Tahmina Banu
- Chittagong Research Institute for Children Surgery (CRICS), Chittagong, Bangladesh
| | - Srikanta Chowdhury
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Muhammad M Hossain
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Herman Yeger
- Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Walid A Farhat
- Division of Pediatric Urology, American Family Children's Hospital, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Syed S Islam
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia. .,School of Medicine, Al-Faisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
33
|
Tombolan L, Rossi E, Zin A, Santoro L, Bonvini P, Zamarchi R, Bisogno G. Pediatric sarcomas display a variable EpCAM expression in a histology-dependent manner. Transl Oncol 2020; 13:100846. [PMID: 32805674 PMCID: PMC7453064 DOI: 10.1016/j.tranon.2020.100846] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 01/06/2023] Open
Abstract
EpCAM is a transmembrane glycoprotein typically overexpressed in cancer of epithelial origin and mainly involved in the epithelial-to–mesenchymal transition (EMT) of tumor cells that spread and disseminate. Strategies for the targeting and capture of EpCAM-expressing tumor cells are showing promise in cancers prone to metastatize, both as diagnostic tools and potential therapies. Sarcomas are among the most aggressive tumors in children, with a common mesenchymal origin that comprises both soft tissue sarcomas (STS) and bone sarcomas. The aim of this study was to assess EpCAM expression in pediatric sarcomas and correlate its expression with disease progression. To do so, we analyzed a set of cell lines and primary tumor tissues from rhabdomyosarcoma (RMS), Ewing sarcoma (ES), synovial sarcoma (SS) and desmoplastic small round cell tumor (DSRCT) STS, or osteosarcoma (OS) bone cancer. We demonstrated that EpCAM was variably expressed in pediatric sarcomas, with DSRCT, a rare, aggressive and almost fatal tumor type, characterized by the highest EpCAM expression levels. Interestingly, although EpCAM expression was lower in RMS tumors, high levels at diagnosis correlated with reduced patients' overall survival (p < 0.05). Indeed, membrane-bound EpCAM was detected in circulating sarcoma tumor cells, revealing its potential to be used as dissemination biomarker in this type of childhood cancers. This reinforces the concept that pediatric sarcomas do express both epithelial and mesenchymal markers and reside in an intermediate condition that most likely contributes to their aggressive phenotype and low survival rate.
Collapse
Affiliation(s)
- Lucia Tombolan
- Institute of Pediatric Research, Fondazione Città della Speranza, Padua, Italy.
| | - Elisabetta Rossi
- Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padova, Padova, Italy; Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Angelica Zin
- Institute of Pediatric Research, Fondazione Città della Speranza, Padua, Italy
| | - Luisa Santoro
- University Hospital of Padova, Institute of Pathology, Padua, Italy
| | - Paolo Bonvini
- Institute of Pediatric Research, Fondazione Città della Speranza, Padua, Italy
| | - Rita Zamarchi
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Gianni Bisogno
- Department of Woman's and Children's Health, Hematology and Oncology Unit, University of Padua, Padua, Italy
| |
Collapse
|
34
|
León-Mateos L, Abalo A, Casas H, Anido U, Rapado-González Ó, Vieito M, Suárez-Cunqueiro M, Gómez-Tato A, Abal M, López-López R, Muinelo-Romay L. Global Gene Expression Characterization of Circulating Tumor Cells in Metastasic Castration-Resistant Prostate Cancer Patients. J Clin Med 2020; 9:jcm9072066. [PMID: 32630240 PMCID: PMC7408664 DOI: 10.3390/jcm9072066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 02/08/2023] Open
Abstract
Background: Current therapeutic options in the course of metastatic castration-resistant prostate cancers (mCRPC) reinforce the need for reliable tools to characterize the tumor in a dynamic way. Circulating tumor cells (CTCs) have emerged as a viable solution to the problem, whereby patients with a variety of solid tumors, including PC, often do not have recent tumor tissue available for analysis. The biomarker characterization in CTCs could provide insights into the current state of the disease and an overall picture of the intra-tumor heterogeneity. Methods: in the present study, we applied a global gene expression characterization of the CTC population from mCRPC (n = 9), with the goal to better understand the biology of these cells and identify the relevant molecules favoring this tumor progression. Results: This analysis allowed the identification of 50 genes specifically expressed in CTCs from patients. Six of these markers (HOXB13, QKI, MAOA, MOSPD1, SDK1, and FGD4), were validated in a cohort of 28 mCRPC, showing clinical interest for the management of these patients. Of note, the activity of this CTC signature was related to the regulation of MYC, a gene strongly implicated in the biology of mCRPC. Conclusions: Overall, our results represent new evidence on the great value of CTCs as a non-invasive biopsy to characterize PC.
Collapse
Affiliation(s)
- Luis León-Mateos
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (L.L.-M.); (U.A.); (M.S.-C.); (M.A.)
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Alicia Abalo
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (A.A.); (H.C.)
| | - Helena Casas
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (A.A.); (H.C.)
| | - Urbano Anido
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (L.L.-M.); (U.A.); (M.S.-C.); (M.A.)
| | - Óscar Rapado-González
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (A.A.); (H.C.)
- Department of Surgery and Medical Surgical Specialties, Medicine and Dentistry School, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - María Vieito
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, 08035 Barcelona, Spain;
| | - Mercedes Suárez-Cunqueiro
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (L.L.-M.); (U.A.); (M.S.-C.); (M.A.)
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
- Department of Surgery and Medical Surgical Specialties, Medicine and Dentistry School, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Antonio Gómez-Tato
- School of Mathematics, University of Santiago de Compostela (Campus Vida), 15782 Santiago de Compostela, Spain;
| | - Miguel Abal
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (L.L.-M.); (U.A.); (M.S.-C.); (M.A.)
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Rafael López-López
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain; (L.L.-M.); (U.A.); (M.S.-C.); (M.A.)
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
- Correspondence: (R.L.-L.); (L.M.-R.)
| | - Laura Muinelo-Romay
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
- Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain; (A.A.); (H.C.)
- Correspondence: (R.L.-L.); (L.M.-R.)
| |
Collapse
|
35
|
Wang Z, Li Y, Wang Y, Wu D, Lau AHY, Zhao P, Zou C, Dai Y, Chan FL. Targeting prostate cancer stem-like cells by an immunotherapeutic platform based on immunogenic peptide-sensitized dendritic cells-cytokine-induced killer cells. Stem Cell Res Ther 2020; 11:123. [PMID: 32183880 PMCID: PMC7079411 DOI: 10.1186/s13287-020-01634-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/10/2020] [Accepted: 03/04/2020] [Indexed: 12/29/2022] Open
Abstract
Background Autologous cellular immunotherapy or immune enhancement therapy has demonstrated some promising benefits for prostate cancer. T cell-based immunotherapy or sipuleucel-T therapy has yielded certain beneficial responses and a slight improvement on the overall survival of patients with metastatic castration-resistant prostate cancer (mCRPC) as shown in some clinical trials, suggesting that prostate cancer is immunoresponsive. Methods In this study, we developed an adaptive cytokine-induced killer cell (CIK)-based immunotherapeutic application targeting the prostate cancer stem-like cells (PCSCs). In this therapeutic platform, dendritic cells (DC) were isolated from the peripheral blood mononuclear cells (PBMCs) and preloaded or sensitized with immunogenic peptides derived from two PCSC-associated cell membrane molecules, CD44 and EpCAM, followed by co-culture with the expanded peripheral blood lymphocyte (PBL)-derived CIK cells. The in vitro cytotoxic activity of DC-activated CIK cells against PCSCs was determined by CCK8 and TUNEL assays, and the in vivo anti-tumor effect of DC-activated CIK cells on prostate cancer xenograft tumors was evaluated in subcutaneous and orthotopic xenograft models. Results Our results showed that the peptide-sensitized DC-CIK cell preparation manifested significant in vitro cytotoxic activity against the PCSC-enriched prostatospheroids and also in vivo anti-tumor effect against prostate cancer xenografts derived from the PCSC-enriched prostatospheroids. Conclusions Together, our established immunogenic peptide-sensitized DC-CIK-based cell preparation platform manifests its potential immunotherapeutic application in targeting the PCSCs and also prostate cancer.
Collapse
Affiliation(s)
- Zhu Wang
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China.,School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Youjia Li
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Dinglan Wu
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Alaster Hang Yung Lau
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Pan Zhao
- Clinical Medical Research Center, The Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
36
|
Tran PHL, Xiang D, Nguyen TNG, Tran TTD, Chen Q, Yin W, Zhang Y, Kong L, Duan A, Chen K, Sun M, Li Y, Hou Y, Zhu Y, Ma Y, Jiang G, Duan W. Aptamer-guided extracellular vesicle theranostics in oncology. Theranostics 2020; 10:3849-3866. [PMID: 32226524 PMCID: PMC7086349 DOI: 10.7150/thno.39706] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
In the past decade, the study of exosomes, nanosized vesicles (50-150 nm) released into the extracellular space via the fusion of multivesicular bodies with the plasma membrane, has burgeoned with impressive achievements in theranostics applications. These nanosized vesicles have emerged as key players in homeostasis and in the pathogenesis of diseases owing to the variety of the cargos they can carry, the nature of the molecules packaged inside the vesicles, and the robust interactions between exosomes and target cells or tissues. Accordingly, the development of exosome-based liquid biopsy techniques for early disease detection and for monitoring disease progression marks a new era of precision medicine in the 21st century. Moreover, exosomes possess intrinsic properties - a nanosized structure and unique "homing effects" - that make them outstanding drug delivery vehicles. In addition, targeted exosome-based drug delivery systems can be further optimized using active targeting ligands such as nucleic acid aptamers. Indeed, the aptamers themselves can function as therapeutic and/or diagnostic tools based on their attributes of unique target-binding and non-immunogenicity. This review aims to provide readers with a current picture of the research on exosomes and aptamers and their applications in cancer theranostics, highlighting recent advances in their transition from the bench to the clinic.
Collapse
Affiliation(s)
- Phuong H-L Tran
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria, Australia
| | - Dongxi Xiang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, 77 Avenue Louise Pasteur, Boston, MA 02115, USA
| | - Tuong N-G Nguyen
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria, Australia
| | - Thao T-D Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Qian Chen
- Translational Medical Center, The Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian District, Beijing, China, 100853
| | - Wang Yin
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria, Australia
| | - Yumei Zhang
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria, Australia
| | - Lingxue Kong
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Andrew Duan
- School of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, 27 Rainforest Walk, Clayton VIC 3800, Australia
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, He'nan Key Laboratory of Tumor Pathology, Zhengzhou 450052, China
| | - Miomio Sun
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, He'nan Key Laboratory of Tumor Pathology, Zhengzhou 450052, China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, and St George and Sutherland Clinical School, University of New South Wales, Kensington, NSW, Australia
| | - Yingchun Hou
- Laboratory of Tumor Molecular and Cellular Biology, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi 710119, China
| | - Yimin Zhu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yongchao Ma
- Clinical School, Luohe Medical College, 148, Daxue Road, Luohe City, Henan Province, 462000, China
| | - Guoqin Jiang
- Department of General Surgery, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, P.R. China, 215004
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria, Australia
- GenePharma-Deakin Joint Laboratory of Aptamer Medicine, Suzhou 215123, China and Waurn Ponds, Victoria 3216, Australia
| |
Collapse
|
37
|
Park S, Song CS, Lin CL, Jiang S, Osmulski PA, Wang CM, Marck BT, Matsumoto AM, Morrissey C, Gaczynska ME, Chen Y, Mostaghel EA, Chatterjee B. Inhibitory Interplay of SULT2B1b Sulfotransferase with AKR1C3 Aldo-keto Reductase in Prostate Cancer. Endocrinology 2020; 161:bqz042. [PMID: 31894239 PMCID: PMC7341717 DOI: 10.1210/endocr/bqz042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/30/2019] [Indexed: 12/22/2022]
Abstract
SULT2B1b (SULT2B) is a prostate-expressed hydroxysteroid sulfotransferase, which may regulate intracrine androgen homeostasis by mediating 3β-sulfation of dehydroepiandrosterone (DHEA), the precursor for 5α-dihydrotestosterone (DHT) biosynthesis. The aldo-keto reductase (AKR)1C3 regulates androgen receptor (AR) activity in castration-resistant prostate cancer (CRPC) by promoting tumor tissue androgen biosynthesis from adrenal DHEA and also by functioning as an AR-selective coactivator. Herein we report that SULT2B-depleted CRPC cells, arising from stable RNA interference or gene knockout (KO), are markedly upregulated for AKR1C3, activated for ERK1/2 survival signal, and induced for epithelial-to-mesenchymal (EMT)-like changes. EMT was evident from increased mesenchymal proteins and elevated EMT-inducing transcription factors SNAI1 and TWIST1 in immunoblot and single-cell mass cytometry analyses. SULT2B KO cells showed greater motility and invasion in vitro; growth escalation in xenograft study; and enhanced metastatic potential predicted on the basis of decreased cell stiffness and adhesion revealed from atomic force microscopy analysis. While AR and androgen levels were unchanged, AR activity was elevated, since PSA and FKBP5 mRNA induction by DHT-activated AR was several-fold higher in SULT2B-silenced cells. AKR1C3 silencing prevented ERK1/2 activation and SNAI1 induction in SULT2B-depleted cells. SULT2B was undetectable in nearly all CRPC metastases from 50 autopsy cases. Primary tumors showed variable and Gleason score (GS)-independent SULT2B levels. CRPC metastases lacking SULT2B expressed AKR1C3. Since AKR1C3 is frequently elevated in advanced prostate cancer, the inhibitory influence of SULT2B on AKR1C3 upregulation, ERK1/2 activation, EMT-like induction, and on cell motility and invasiveness may be clinically significant. Pathways regulating the inhibitory SULT2B-AKR1C3 axis may inform new avenue(s) for targeting SULT2B-deficient prostate cancer.
Collapse
Affiliation(s)
- Sulgi Park
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
- Department of Microbiology & Immunology, Pusan National University School of Medicine, South Korea
- South Texas Veterans Health Care System, San Antonio, Texas
| | - Chung-Seog Song
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
- South Texas Veterans Health Care System, San Antonio, Texas
| | - Chun-Lin Lin
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Shoulei Jiang
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
- South Texas Veterans Health Care System, San Antonio, Texas
| | - Pawel A Osmulski
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Chiou-Miin Wang
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Brett T Marck
- Geriatric Research, Education & Clinical Center, VA Puget Sound Health Care System, Seattle, WA
| | - Alvin M Matsumoto
- Geriatric Research, Education & Clinical Center, VA Puget Sound Health Care System, Seattle, WA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA
| | - Maria E Gaczynska
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Yidong Chen
- Department of Epidemiology & Biostatistics, University of Texas Health San Antonio, San Antonio, Texas
- Greehy Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas
| | - Elahe A Mostaghel
- Geriatric Research, Education & Clinical Center, VA Puget Sound Health Care System, Seattle, WA
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Bandana Chatterjee
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
- South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
38
|
A comparative study on EpCAM antibody immobilization on gold surfaces and microfluidic channels for the detection of circulating tumor cells. Colloids Surf B Biointerfaces 2020; 188:110808. [PMID: 31991289 DOI: 10.1016/j.colsurfb.2020.110808] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 01/09/2023]
Abstract
Detection of circulating tumor cells (CTCs) from the bloodstream holds great importance to diagnose cancer at early stages. However, CTCs being extremely rare in blood makes them difficult to reach. In this paper, we introduced different surface modification techniques for the enrichment and detection of MCF-7 in microfluidic biosensor applications using gold surface and EpCAM antibody. Mainly, two different mechanisms were employed to immobilize the antibodies; covalent bonding and bioaffinity interaction. Self-assembled monolayers (SAMs) formed on the gold surfaces were treated further for the immobilization of the antibody. The bioaffinity-based studies were performed with streptavidin and biotinylated EpCAM over the SAM coated surfaces. The cell attachment events were monitored using fluorescent microscope. Comparisons were made considering the length and functional end of alkanethiols and the positioning of the antibody. Then, these methods were integrated into a microfluidic channel system. Surface characterizations were performed with X-ray Photoelectron Spectroscopy, Atomic Force Microscopy, and contact angle measurements. The selectivity studies were carried out with EpCAM negative K562 leukaemia cell lines and the experiments were repeated for different types of surfaces, such as glass and polymer. Studies showed that long (n>10) and aromatic ring containing alkanethiols lead to better cell capture events compared to shorter ones. Results obtained from the comparisons are of importance for the gold surface-based microfluidic biosensor designs aimed for CTC detection.
Collapse
|
39
|
Deliorman M, Janahi FK, Sukumar P, Glia A, Alnemari R, Fadl S, Chen W, Qasaimeh MA. AFM-compatible microfluidic platform for affinity-based capture and nanomechanical characterization of circulating tumor cells. MICROSYSTEMS & NANOENGINEERING 2020; 6:20. [PMID: 34567635 PMCID: PMC8433216 DOI: 10.1038/s41378-020-0131-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/30/2019] [Accepted: 12/22/2019] [Indexed: 05/05/2023]
Abstract
Circulating tumor cells (CTCs) carried by the patient's bloodstream are known to lead to the metastatic spread of cancer. It is becoming increasingly clear that an understanding of the nanomechanical characteristics of CTCs, such as elasticity and adhesiveness, represents advancements in tracking and monitoring cancer progression and metastasis. In the present work, we describe a combined microfluidic-atomic force microscopy (AFM) platform that uses antibody-antigen capture to routinely isolate and nanomechanically characterize CTCs present in blood samples from prostate cancer patients. We introduce the reversible assembly of a microfluidic device and apply refined and robust chemistry to covalently bond antibodies onto its glass substrate with high density and the desired orientation. As a result, we show that the device can efficiently capture CTCs from patients with localized and metastatic prostate cancer through anti-EpCAM, anti-PSA, and anti-PSMA antibodies, and it is suitable for AFM measurements of captured intact CTCs. When nanomechanically characterized, CTCs originating from metastatic cancer demonstrate decreased elasticity and increased deformability compared to those originating from localized cancer. While the average adhesion of CTCs to the AFM tip surface remained the same in both the groups, there were fewer multiple adhesion events in metastatic CTCs than there were in their counterparts. The developed platform is simple, robust, and reliable and can be useful in the diagnosis and prognosis of prostate cancer as well as other forms of cancer.
Collapse
Affiliation(s)
- Muhammedin Deliorman
- Division of Engineering, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Farhad K. Janahi
- Mohammed Bin Rashid University of Medicine and Health Sciences, P.O. Box 505055, Dubai, UAE
| | - Pavithra Sukumar
- Division of Engineering, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Ayoub Glia
- Division of Engineering, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
- Department of Mechanical and Aerospace Engineering, New York University, New York, NY 10003 USA
| | - Roaa Alnemari
- Division of Engineering, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Samar Fadl
- Division of Science, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, New York, NY 10003 USA
- Department of Biomedical Engineering, New York University, New York, NY 10003 USA
| | - Mohammad A. Qasaimeh
- Division of Engineering, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
- Department of Mechanical and Aerospace Engineering, New York University, New York, NY 10003 USA
| |
Collapse
|
40
|
Lin HP, Ho HM, Chang CW, Yeh SD, Su YW, Tan TH, Lin WJ. DUSP22 suppresses prostate cancer proliferation by targeting the EGFR-AR axis. FASEB J 2019; 33:14653-14667. [PMID: 31693867 DOI: 10.1096/fj.201802558rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dual-specificity phosphatases (DUSPs) regulate the activity of various downstream kinases through serine or threonine or tyrosine dephosphorylation. Loss of function and aberrant expression of DUSPs has been implicated in cancer progression and poor survival, yet the function of DUSP22 in prostate cancer (PCa) cells is not clear. Gene Expression Omnibus and cBioPortal microarray database analyses showed that DUSP22 expression was lower in PCa tissues than normal prostate tissues, and altered DUSP22 expression was associated with shorter progression-free and disease-free survival of patients with PCa. Exogenous DUSP22 expression in LNCaP, PC3, and C4-2B PCa cells inhibited cellular proliferation and colony formation, supporting a growth inhibitory role for DUSP22 in PCa cells. DUSP22 expression significantly attenuated epidermal growth factor (EGF) receptor (EGFR) and its downstream ERK1/2 signaling by dephosphorylation. However, DUSP22 failed to suppress the growth of CWR22Rv1 and DU145 cells with elevated phosphorylated (p-)ERK1/2 levels. A serine-to-alanine mutation at position 58, a potential ERK1/2-targeted phosphorylation site in DUSP22, was sufficient to suppress growth of CWR22Rv1 cells with elevated p-ERK1/2 levels, suggesting a mutually antagonistic relationship between DUSP22 and ERK1/2 dependent on phosphorylation status. We showed that DUSP22 can suppress prostate-specific antigen gene expression through phosphatase-dependent pathways, suggesting that DUSP22 is an important regulator of the androgen receptor (AR) in PCa cells. Mechanistically, DUSP22 can interact with AR as a regulatory partner and interfere with EGF-induced AR phosphorylation at Tyr534, suggesting that DUSP22 serves as a crucial suppressor of both EGFR and AR-dependent signaling in PCa cells via dephosphorylation. Our findings indicate that loss of function of DUSP22 in PCa cells leads to aberrant activation of both EGFR-ERKs and AR signaling and ultimately progression of PCa, supporting the potential for novel therapeutic design of harnessing DUSP22 in the treatment of PCa.-Lin, H.-P., Ho, H.-M., Chang, C.-W., Yeh, S.-D., Su, Y.-W., Tan, T.-H., Lin, W.-J. DUSP22 suppresses prostate cancer proliferation by targeting the EGFR-AR axis.
Collapse
Affiliation(s)
- Hsiu-Ping Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Hui-Min Ho
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Cheng-Wei Chang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Shauh-Der Yeh
- Department of Urology, Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Wen-Jye Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| |
Collapse
|
41
|
Razdan A, de Souza P, Roberts TL. Role of MicroRNAs in Treatment Response in Prostate Cancer. Curr Cancer Drug Targets 2019; 18:929-944. [PMID: 29644941 PMCID: PMC6463399 DOI: 10.2174/1568009618666180315160125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 12/16/2022]
Abstract
Prostate cancer (PCa) is the most common non-skin cancer in men worldwide, resulting in significant mortality and morbidity. Depending on the grade and stage of the cancer, patients may be given radiation therapy, hormonal therapy, or chemotherapy. However, more than half of these patients develop resistance to treatment, leading to disease progression and metastases, often with lethal consequences. MicroRNAs (miRNAs) are short, non-coding RNAs, which regulate numerous physiological as well as pathological processes, including cancer. miRNAs mediate their regulatory effect predominately by binding to the 3'-untranslated region (UTR) of their target mRNAs. In this review, we will describe the mechanisms by which miRNAs mediate resistance to radiation and drug therapy (i.e. hormone therapy and chemotherapy) in PCa, including control of apoptosis, cell growth and proliferation, autophagy, epithelial-to-mesenchymal transition (EMT), invasion and metastasis, and cancer stem cells (CSCs). Furthermore, we will discuss the utility of circulating miRNAs isolated from different body fluids of prostate cancer patients as non-invasive biomarkers of cancer detection, disease progression, and therapy response. Finally, we will shortlist the candidate miRNAs, which may have a role in drug and radioresistance, that could potentially be used as predictive biomarkers of treatment response.
Collapse
Affiliation(s)
- Anshuli Razdan
- Medical Oncology Group, Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Sydney, New South Wales, Australia.,Centre for Oncology Education and Research Translation (CONCERT), Liverpool, New South Wales, Australia
| | - Paul de Souza
- Medical Oncology Group, Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Sydney, New South Wales, Australia.,Centre for Oncology Education and Research Translation (CONCERT), Liverpool, New South Wales, Australia.,School of Medicine, The University of New South Wales, Sydney, New South Wales, Australia.,Department of Medical Oncology, Liverpool Hospital, Liverpool, New South Wales, Australia
| | - Tara Laurine Roberts
- Medical Oncology Group, Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Sydney, New South Wales, Australia.,Centre for Oncology Education and Research Translation (CONCERT), Liverpool, New South Wales, Australia.,School of Medicine, The University of New South Wales, Sydney, New South Wales, Australia.,The University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| |
Collapse
|
42
|
Radiosensitive core/satellite ternary heteronanostructure for multimodal imaging-guided synergistic cancer radiotherapy. Biomaterials 2019; 226:119545. [PMID: 31648136 DOI: 10.1016/j.biomaterials.2019.119545] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/12/2019] [Accepted: 10/13/2019] [Indexed: 01/06/2023]
Abstract
Developing safe, effective and targeting radiosensitizers with clear action mechanisms to achieve synergistic localized cancer treatment is an important strategy for radiotherapy. Herein, we design and synthesize a ternary heteronanostructure radiosensitizer (SeAuFe-EpC) with core/satellite morphology by a simple method to realize multimodal imaging-guided cancer radiotherapy. The mechanistic studies reveal that Se incorporation could drastically improve the electrical conductivity and lower the energy barrier between the three components, resulting in more electrons transfer between Se-Au interface and migration over the heterogeneous junction of Au-Fe3O4 NPs interface. This synergistic interaction enhanced the energy transfer and facilitated more excited excitons generated by SeAuFe-EpC NPs, thus promoting the transformation of 3O2 to 1O2via resonance energy transfer, finally resulting in irreversible cancer cell apoptosis. Additionally, based on the X-ray attenuation ability and high NIR absorption of AuNPs and the superparamagnetism of Fe3O4, in vivo computer tomography, photoacoustic and magnetic resonance tri-modal imaging have been employed to visualize the tracking and targeting ability of the NPs. As expected, the NPs specifically accumulated in orthotopic breast tumor area and achieved synergistic anticancer efficacy, but showed no toxic side effects on main organs. Collectively, this study sheds light on the potential roles of core/satellite heteronanostructure in imaging-guided cancer radiotherapy.
Collapse
|
43
|
He Z, Duan X, Zeng G. Identification of potential biomarkers and pivotal biological pathways for prostate cancer using bioinformatics analysis methods. PeerJ 2019; 7:e7872. [PMID: 31598425 PMCID: PMC6779116 DOI: 10.7717/peerj.7872] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022] Open
Abstract
Background Prostate cancer (PCa) is a common urinary malignancy, whose molecular mechanism has not been fully elucidated. We aimed to screen for key genes and biological pathways related to PCa using bioinformatics method. Methods Differentially expressed genes (DEGs) were filtered out from the GSE103512 dataset and subjected to the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The protein–protein interactions (PPI) network was constructed, following by the identification of hub genes. The results of former studies were compared with ours. The relative expression levels of hub genes were examined in The Cancer Genome Atlas (TCGA) and Oncomine public databases. The University of California Santa Cruz Xena online tools were used to study whether the expression of hub genes was correlated with the survival of PCa patients from TCGA cohorts. Results Totally, 252 (186 upregulated and 66 downregulated) DEGs were identified. GO analysis enriched mainly in “oxidation-reduction process” and “positive regulation of transcription from RNA polymerase II promoter”; KEGG pathway analysis enriched mostly in “metabolic pathways” and “protein digestion and absorption.” Kallikrein-related peptidase 3, cadherin 1 (CDH1), Kallikrein-related peptidase 2 (KLK2), forkhead box A1 (FOXA1), and epithelial cell adhesion molecule (EPCAM) were identified as hub genes from the PPI network. CDH1, FOXA1, and EPCAM were validated by other relevant gene expression omnibus datasets. All hub genes were validated by both TCGA and Oncomine except KLK2. Two additional top DEGs (ABCC4 and SLPI) were found to be associated with the prognosis of PCa patients. Conclusions This study excavated the key genes and pathways in PCa, which might be biomarkers for diagnosis, prognosis, and potential therapeutic targets.
Collapse
Affiliation(s)
- Zihao He
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Urology, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Xiaolu Duan
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Urology, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Guohua Zeng
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Urology, Guangzhou, China.,Guangdong Key Laboratory of Urology, Guangzhou, China
| |
Collapse
|
44
|
Cho S, Yang HC, Rhee WJ. Simultaneous multiplexed detection of exosomal microRNAs and surface proteins for prostate cancer diagnosis. Biosens Bioelectron 2019; 146:111749. [PMID: 31600625 DOI: 10.1016/j.bios.2019.111749] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/23/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022]
Abstract
Since the tumor is extremely heterogeneous, a single biomarker cannot reflect the exact symptoms of the disease or its stage. Exosomes are biomarker reservoirs that provide disease information with a high accuracy, especially when specific markers, including microRNAs (miRNAs) and proteins, are combined. However, currently available exosomal miRNA and protein detection methods are time consuming, expensive, and laborious. Meanwhile, simultaneous detection of an exosomal miRNA and protein in a single reaction is even more challenging. Thus, development of an efficient method for detecting multiple miRNAs and proteins in a single exosomal reaction is highly needed. Herein, to increase the value of using exosomes over other circulating biomarkers for prostate cancer (PCa) liquid biopsy, a method for simultaneous multiplexed in situ detection of exosomal miRNAs and proteins was developed. Exosomal miRNAs and surface proteins were simultaneously detected in captured exosomes with a high specificity, using nano-sized molecular beacons and fluorescent dye-conjugated antibodies. The method allowed the quantitative analysis of various disease-specific miRNAs and surface proteins in PCa cell-derived exosomes in a single exosomal reaction. Overall, simultaneous multiplexed in situ detection of exosomal miRNAs and surface proteins can be developed as a simple, cost-effective, non-invasive liquid biopsy method for diagnosing PCa.
Collapse
Affiliation(s)
- Seongcheol Cho
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Hee Cheol Yang
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Won Jong Rhee
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea; Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea.
| |
Collapse
|
45
|
Islam MK, Syed P, Lehtinen L, Leivo J, Gidwani K, Wittfooth S, Pettersson K, Lamminmäki U. A Nanoparticle-Based Approach for the Detection of Extracellular Vesicles. Sci Rep 2019; 9:10038. [PMID: 31296879 PMCID: PMC6624270 DOI: 10.1038/s41598-019-46395-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/18/2019] [Indexed: 01/17/2023] Open
Abstract
The analysis of extracellular vesicles (EVs) typically requires tedious and time-consuming isolation process from bio-fluids. We developed a nanoparticle-based time resolved fluorescence immunoassay (NP-TRFIA) that uses biotinylated antibodies against the proteins of tetraspanin family and tumor-associated antigens for capturing EVs from urine samples and cell culture supernatants without the need for isolation. The captured-EVs were detected either with Eu3+-chelate or Eu3+-doped nanoparticle-based labels conjugated either to antibodies against the tetraspanins or lectins targeting the glycan moieties on EVs surface. The NP-TRFIA demonstrated specific capturing and detection of EVs by antibodies and lectins. Lectin-nanoparticle based assays showed 2–10 fold higher signal-to-background ratio compared with lectin-chelate assays. The nanoparticle assay concept allowed surface glycosylation profiling of the urine derived-EVs with lectins. It was also applied to establish an assay showing differential expression of tumor-associated proteins on more aggressive (higher ITGA3 on DU145- and PC3-EVs) compared to less aggressive (higher EpCAM on LNCaP-EVs) PCa- cell lines derived-EVs. This NP-TRFIA can be used as a simple tool for analysis and characterization of EVs in urine and cell culture supernatants. Such approach could be useful in identification of disease-specific markers on the surface of patient-derived urinary EVs.
Collapse
Affiliation(s)
- Md Khirul Islam
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland.
| | - Parvez Syed
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Laura Lehtinen
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Janne Leivo
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland.,Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kamlesh Gidwani
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Saara Wittfooth
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Kim Pettersson
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Urpo Lamminmäki
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| |
Collapse
|
46
|
Abstract
Since the introduction of the cancer stem cell (CSC) hypothesis, accumulating evidence shows that most cancers present stem-like niches. However, therapies aimed at targeting this niche have not been as successful as expected. New evidence regarding CSCs hierarchy, similarities with normal tissue stem cells and cell plasticity might be key in understanding their role in cancer biology and how to efficiently eliminate them. In this Chapter, we discuss what is known in breast and prostate CSCs from their initial discoveries to the current therapeutic efforts in the field. Future challenges towards better CSC identification and isolation strategies will be key to shed light into how CSCs could accurately be targeted in combination to traditional therapies to ultimately prolong patient survival.
Collapse
Affiliation(s)
- Rocío G Sampayo
- Department of Chemical and Biomolecular Engineering, University of California at Berkeley, Berkeley, CA, United States
| | - Mina J Bissell
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
47
|
Campos-Silva C, Suárez H, Jara-Acevedo R, Linares-Espinós E, Martinez-Piñeiro L, Yáñez-Mó M, Valés-Gómez M. High sensitivity detection of extracellular vesicles immune-captured from urine by conventional flow cytometry. Sci Rep 2019; 9:2042. [PMID: 30765839 PMCID: PMC6376115 DOI: 10.1038/s41598-019-38516-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) provide an invaluable tool to analyse physiological processes because they transport, in biological fluids, biomolecules secreted from diverse tissues of an individual. EV biomarker detection requires highly sensitive techniques able to identify individual molecules. However, the lack of widespread, affordable methodologies for high-throughput EV analyses means that studies on biomarkers have not been done in large patient cohorts. To develop tools for EV analysis in biological samples, we evaluated here the critical parameters to optimise an assay based on immunocapture of EVs followed by flow cytometry. We describe a straightforward method for EV detection using general EV markers like the tetraspanins CD9, CD63 and CD81, that allowed highly sensitive detection of urinary EVs without prior enrichment. In proof-of-concept experiments, an epithelial marker enriched in carcinoma cells, EpCAM, was identified in EVs from cell lines and directly in urine samples. However, whereas EVs isolated from 5–10 ml of urine were required for western blot detection of EpCAM, only 500 μl of urine were sufficient to visualise EpCAM expression by flow cytometry. This method has the potential to allow any laboratory with access to conventional flow cytometry to identify surface markers on EVs, even non-abundant proteins, using minimally processed biological samples.
Collapse
Affiliation(s)
- Carmen Campos-Silva
- Department of Immunology and Oncology, National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - Henar Suárez
- Department of Molecular Biology, UAM, Centro de Biología Molecular Severo Ochoa (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | | | | | - Luis Martinez-Piñeiro
- Servicio de Urología and Instituto Sanitario (Idipaz), Hospital Universitario La Paz, Madrid, Spain
| | - María Yáñez-Mó
- Department of Molecular Biology, UAM, Centro de Biología Molecular Severo Ochoa (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Mar Valés-Gómez
- Department of Immunology and Oncology, National Centre for Biotechnology, CNB-CSIC, Madrid, Spain.
| |
Collapse
|
48
|
Kapka-Skrzypczak L, Popek S, Sawicki K, Drop B, Czajka M, Jodłowska-Jędrych B, Matysiak-Kucharek M, Furman-Toczek D, Zagórska-Dziok M, Kruszewski M. IL‑6 prevents CXCL8‑induced stimulation of EpCAM expression in ovarian cancer cells. Mol Med Rep 2019; 19:2317-2322. [PMID: 30747214 DOI: 10.3892/mmr.2019.9890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/19/2018] [Indexed: 11/06/2022] Open
Abstract
Epithelial cell adhesion molecule (EpCAM), which is expressed in the majority of epithelial tissues, exhibits tumor growth promoting abilities and is overexpressed in human epithelial ovarian cancer. Therefore, EpCAM is considered to be a promising target for specific immune‑based therapies. The present study evaluated the role of IL‑6 and IL‑8 in the expression of EpCAM in the A2780 human ovarian cancer cell line. Furthermore, the cellular localization of the EpCAM protein in A2780 cells was determined and the effect of EpCAM inhibition on the proliferation of the A2780 cells was investigated. An MTT assay demonstrated that blocking EpCAM with anti‑EPCAM antibodies had no effect on cellular metabolic activity (proliferation). Gene expression analysis revealed that IL‑8 increased EpCAM expression, whereas IL‑6 and the combination of IL‑6/IL‑8 had no effect on EpCAM expression. Immunofluorescence analysis confirmed that EpCAM is expressed on A2780 cell membranes. The present results demonstrated that IL‑8 increased EpCAM expression at the mRNA level in ovarian cancer cells and suggested a potential role of IL‑6 as an inhibitor of IL‑8‑stimulated EpCAM expression.
Collapse
Affiliation(s)
- Lucyna Kapka-Skrzypczak
- Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, 35‑225 Rzeszow, Poland
| | - Sylwia Popek
- Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, 20‑080 Lublin, Poland
| | - Krzysztof Sawicki
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 0‑090 Lublin, Poland
| | - Bartłomiej Drop
- Department of Informatics and Medical Statistics, Faculty of Health Sciences, Medical University, 20‑090 Lublin, Poland
| | - Magdalena Czajka
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 0‑090 Lublin, Poland
| | - Barbara Jodłowska-Jędrych
- Department of Histology and Embryology with Experimental Cytology Unit, Medical University of Lublin, 20‑080 Lublin, Poland
| | | | - Dominika Furman-Toczek
- Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, 35‑225 Rzeszow, Poland
| | - Martyna Zagórska-Dziok
- Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, 35‑225 Rzeszow, Poland
| | - Marcin Kruszewski
- Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, 35‑225 Rzeszow, Poland
| |
Collapse
|
49
|
Spontaneous formation of tumor spheroid on a hydrophilic filter paper for cancer stem cell enrichment. Colloids Surf B Biointerfaces 2018; 174:426-434. [PMID: 30481703 DOI: 10.1016/j.colsurfb.2018.11.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/16/2018] [Accepted: 11/17/2018] [Indexed: 01/30/2023]
Abstract
Emerging evidence has demonstrated that cancer stem cells (CSCs) play critical roles in tumor invasion, metastasis and recurrence. The specific targeting capability on CSCs is of high importance for the development of effective anti-tumor therapeutics. However, isolation, enrichment and cultivation of these special and rare groups of tumor cells for in vitro analyses is a nontrivial job and requires particular culture medium and environmental control. Herein, we established a low-cost and efficient method for CSC enrichment by culturing prostate cancer cells on a hydrophilic filter paper. We found that tumor spheroids could form spontaneously on a pristine filter paper solely with regular cell culture medium. The paper-grown cells had elevated expression of putative CSC markers, indicating increased stemness of the cancer cells. Moreover, increased resistance of the chemotherapeutic drug doxorubicin was observed on the formed CSC spheroids compared to regular culture. The properties of the filter paper were characterized to investigate the underlying mechanism behind the promoted tumor spheroid formation. The obtained results suggested that the excellent hydrophilicity of the cellulose fibers retarded the hydrophobic interaction-mediated cell anchoring on the cellulose fibers, while the limited space/niche between fibers promoted the aggregation of cells. In addition, biocompatible paper-based materials are able to realize convenient assembly of tissue-like structures for developing in vitro disease models or organs-on-paper applications. Therefore, hydrophilic filter papers could be a low-cost material for construction of various assay platforms for isolating and enriching CSCs, screening anti-tumor drugs, and constructing tumor models in vitro.
Collapse
|
50
|
Ni J, Cozzi P, Beretov J, Duan W, Bucci J, Graham P, Li Y. Epithelial cell adhesion molecule (EpCAM) is involved in prostate cancer chemotherapy/radiotherapy response in vivo. BMC Cancer 2018; 18:1092. [PMID: 30419852 PMCID: PMC6233586 DOI: 10.1186/s12885-018-5010-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022] Open
Abstract
Background Development of chemo−/radioresistance is a major challenge for the current prostate cancer (CaP) therapy. We have previously demonstrated that epithelial cell adhesion molecule (EpCAM) is associated with CaP growth and therapeutic resistance in vitro, however, the role of EpCAM in CaP in vivo is not fully elucidated. Here, we aimed to investigate how expression of EpCAM is involved in CaP growth and chemo−/radiotherapy response in NOD/SCID mouse models in vivo and to validate its role as a therapeutic target for CaP therapy. Methods EpCAM was knocked down in PC-3 CaP cell line using short hairpin RNA (shRNA). The effect of EpCAM-knockdown (KD) on tumour growth, chemo−/radiotherapy response and animal survival was evaluated on subcutaneous (s.c) and orthotopic mouse models. Results We found that KD of EpCAM significantly inhibited tumour growth, increased xenograft sensitivity to chemotherapy/radiotherapy, and prolonged the survival of tumour-bearing mice. In addition, we demonstrated that KD of EpCAM is associated with downregulation of the PI3K/Akt/mTOR pathway. Conclusions In conclusion, our data confirms that CaP growth and chemo−/radioresistance in vivo is associated with over-expression of EpCAM, which serves both a functional biomarker and promising therapeutic target. Electronic supplementary material The online version of this article (10.1186/s12885-018-5010-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Paul Cozzi
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Department of Surgery, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Julia Beretov
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Anatomical Pathology, NSW Health Pathology, St George Hospital, Gray St, Kogarah, NSW, 2217, Australia
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - Joseph Bucci
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Peter Graham
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia. .,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia. .,School of Basic Medical Sciences, Zhengzhou University, Henan, 450001, China.
| |
Collapse
|