1
|
Moghbeli M. MicroRNAs as the critical regulators of bone metastasis during prostate tumor progression. Int J Biol Macromol 2025; 309:142912. [PMID: 40203904 DOI: 10.1016/j.ijbiomac.2025.142912] [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: 03/01/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Prostate cancer (PCa) is the most prevalent cancer among men globally. Although, there are various therapeutic methods for the localized or advanced cancers, there is still a high rate of mortality among PCa patients that is mainly associated with bone metastasis in advanced tumors. There are few options available for treating bone metastasis in PCa, which only provide symptom relief without curing the disease. Therefore, it is crucial to evaluate the molecular mechanisms associated with bone metastasis of PCa cells to suggest the novel diagnostic and therapeutic approaches that could lower the morbidity and mortality rates in PCa patients. MicroRNAs (miRNAs) are involved in regulation of various pathophysiological processes such as tumor growth and osteoblasts/osteoclasts formation. Since, miRNA deregulation has been also frequently observed in PCa patients with bone metastasis, we discussed the role of miRNAs in bone metastasis during PCa progression. It has been reported that miRNAs mainly reduced the ability of PCa tumor cells for the bone metastasis through the regulation of WNT, NF-kB, PI3K/AKT, and TGF-β signaling pathways. They also affected the EMT process, transcription factors, and structural proteins to regulate the bone metastasis during PCa progression. This review paves the way to suggest the miRNAs as the reliable markers not only for the non-invasive early diagnosis, but also for the targeted therapy of PCa tumors with bone metastasis.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Trecarten S, Liss MA, Hamilton-Reeves J, DiGiovanni J. Obesity, dietary interventions and microbiome alterations in the development and progression of prostate cancer. Front Immunol 2025; 15:1448116. [PMID: 39840030 PMCID: PMC11747771 DOI: 10.3389/fimmu.2024.1448116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/06/2024] [Indexed: 01/23/2025] Open
Abstract
Purpose of review The role of the microbiome in prostate cancer is an emerging subject of research interest. Certain lifestyle factors, such as obesity and diet, can also impact the microbiome, which has been implicated in many diseases, such as heart disease and diabetes. However, this link has yet to be explored in detail in the context of prostate cancer. The purpose of this review is to explore the cross-talk between obesity, dietary interventions, and microbiome alterations in the development and progression of prostate cancer. Recent findings Many possible mechanisms exist linking obesity and dietary interventions to microbiome alterations and prostate cancer. The gut microbiome produces metabolites that could play a role in prostate cancer oncogenesis, including short-chain fatty acids, cholesterol derivatives, and folic acid. The microbiome also plays a pivotal role in the prostate tumor microenvironment (TME), contributing to inflammation, local tissue hypoxia, and epithelial-mesenchymal transition. A bidirectional relationship exists between obesity and the microbiome, and certain diets can enact changes to the microbiome, its associated metabolites, and prostate cancer outcomes. Summary Cross-talk exists between obesity, dietary interventions, and the role of the microbiome in the development and progression of prostate cancer. To further our understanding, future human studies in prostate cancer should investigate microbiome changes and incorporate an assessment of microbiome-derived metabolites and cellular/immune changes in the TME.
Collapse
Affiliation(s)
- Shaun Trecarten
- Department of Urology, The University of Texas Health Sciences Center San Antonio, San Antonio, TX, United States
| | - Michael A. Liss
- Department of Urology, University of San Diego, San Diego, CA, United States
| | - Jill Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, United States
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin and Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
3
|
Latorzeff I, Camps-Maléa A, Supiot S, de Crevoisier R, Farcy-Jacquet MP, Hannoun-Lévi JM, Riou O, Pommier P, Artignan X, Chapet O, Créhange G, Marchesi V, Pasquier D, Sargos P. Indication and perspectives of radiation therapy in the setting of de-novo metastatic prostate cancer. Cancer Radiother 2024; 28:49-55. [PMID: 37827959 DOI: 10.1016/j.canrad.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 10/14/2023]
Abstract
Prostate cancer is the most common cancer and the third leading cause of cancer mortality in men. Each year, approximately 10% of prostate cancers are diagnosed metastatic at initial presentation. The standard treatment option for de-novo metastatic prostate cancer is androgen deprivation therapy with novel hormonal agent or with chemotherapy. Recently, PEACE-1 trial highlighted the benefit of triplet therapy resulting in the combination of androgen deprivation therapy combined with docetaxel and abiraterone. Radiotherapy can be proposed in a curative intent or to treat local symptomatic disease. Nowadays, radiotherapy of the primary disease is only recommended for de novo low-burden/low-volume metastatic prostate cancer, as defined in the CHAARTED criteria. However, studies on stereotactic radiotherapy on oligometastases have shown that this therapeutic approach is feasible and well tolerated. Prospective research currently focuses on the benefit of intensification by combining treatment of the metastatic sites and the primary all together. The contribution of metabolic imaging to better define the target volumes and specify the oligometastatic character allows a better selection of patients. This article aims to define indications of radiotherapy and perspectives of this therapeutic option for de-novo metastatic prostate cancer.
Collapse
Affiliation(s)
- I Latorzeff
- Department of Radiation Oncology, clinique Pasteur, Toulouse, France.
| | - A Camps-Maléa
- Department of Radiation Oncology, hôpital Bretonneau, CHU de Tours, Tours, France
| | - S Supiot
- Department of Radiation Oncology, institut de cancérologie de l'Ouest centre René-Gauducheau, Saint-Herblain, France; CNRS, Nantes, France; Université de Nantes, Nantes, France
| | - R de Crevoisier
- Department of Radiation Oncology, centre Eugène-Marquis, Rennes, France
| | - M-P Farcy-Jacquet
- Fédération universitaire d'oncologie radiothérapie, institut de cancérologie du Gard, CHU de Nîmes, Nîmes, France
| | - J-M Hannoun-Lévi
- Department of Radiation Oncology, centre Antoine-Lacassagne, Nice, France
| | - O Riou
- Department of Radiation Oncology, institut du cancer de Montpellier, Montpellier, France; Fédération universitaire d'oncologie radiothérapie de Méditerranée Occitanie, université de Montpellier, Montpellier, France; U1194, Inserm, Montpellier, France; IRCM, Montpellier, France
| | - P Pommier
- Department of Radiation Oncology, institut de cancérologie de l'Ouest, Angers, France
| | - X Artignan
- Department of Radiation Oncology, centre hospitalier privé Saint-Grégoire, Rennes, France
| | - O Chapet
- Department of Radiation Oncology, centre hospitalier Lyon Sud, Pierre-Bénite, France
| | - G Créhange
- Department of Radiation Oncology, institut Curie, Saint-Cloud, France
| | - V Marchesi
- Department of Medical Physics, centre Alexis-Vautrin, Vandœuvre-lès-Nancy, France
| | - D Pasquier
- Academic Department of Radiation Oncology, centre Oscar-Lambret, Lille, France; UMR 9189 - CRIStAL, université de Lille, CNRS, école Centrale Lille, 59000 Lille, France
| | - P Sargos
- Department of Radiotherapy, institut Bergonié, Bordeaux, France
| |
Collapse
|
4
|
Maji S, Pradhan AK, Kumar A, Bhoopathi P, Mannangatti P, Guo C, Windle JJ, Subler MA, Wang XY, Semmes OJ, Nyalwidhe JO, Mukhopadhyay N, Paul AK, Hatfield B, Levit MM, Madan E, Sarkar D, Emdad L, Cohen DJ, Gogna R, Cavenee WK, Das SK, Fisher PB. MDA-9/Syntenin in the tumor and microenvironment defines prostate cancer bone metastasis. Proc Natl Acad Sci U S A 2023; 120:e2307094120. [PMID: 37922327 PMCID: PMC10636346 DOI: 10.1073/pnas.2307094120] [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: 04/29/2023] [Accepted: 09/25/2023] [Indexed: 11/05/2023] Open
Abstract
Bone metastasis is a frequent and incurable consequence of advanced prostate cancer (PC). An interplay between disseminated tumor cells and heterogeneous bone resident cells in the metastatic niche initiates this process. Melanoma differentiation associated gene-9 (mda-9/Syntenin/syndecan binding protein) is a prometastatic gene expressed in multiple organs, including bone marrow-derived mesenchymal stromal cells (BM-MSCs), under both physiological and pathological conditions. We demonstrate that PDGF-AA secreted by tumor cells induces CXCL5 expression in BM-MSCs by suppressing MDA-9-dependent YAP/MST signaling. CXCL5-derived tumor cell proliferation and immune suppression are consequences of the MDA-9/CXCL5 signaling axis, promoting PC disease progression. mda-9 knockout tumor cells express less PDGF-AA and do not develop bone metastases. Our data document a previously undefined role of MDA-9/Syntenin in the tumor and microenvironment in regulating PC bone metastasis. This study provides a framework for translational strategies to ameliorate health complications and morbidity associated with advanced PC.
Collapse
Affiliation(s)
- Santanu Maji
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Anjan K. Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Amit Kumar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Jolene J. Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Mark A. Subler
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Oliver J. Semmes
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA23507
| | - Julius O. Nyalwidhe
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA23507
| | - Nitai Mukhopadhyay
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- Department of Biostatistics, Virginia Commonwealth University, School of Medicine, Richmond, VA23238
| | - Asit Kr. Paul
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- Department of Internal Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23238
| | - Bryce Hatfield
- Department of Pathology, Virginia Commonwealth University, School of Medicine, Richmond, VA23238
| | - Michael M. Levit
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA23238
| | - Esha Madan
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- Department of Surgery, Virginia Commonwealth University, School of Medicine, Richmond, VA23238
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - David J. Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA23238
| | - Rajan Gogna
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Webster K. Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA92093
| | - Swadesh K. Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| | - Paul B. Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA23298
| |
Collapse
|
5
|
Gupta KR, Kyprianou N. Microbiota and the landscape of the prostate tumor microenvironment. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2023; 11:352-360. [PMID: 37941651 PMCID: PMC10628624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 08/07/2023] [Indexed: 11/10/2023]
Abstract
Prostate cancer remains one of the most common causes of cancer-related death in men globally. Progression of prostate cancer to lethal metastatic disease is mediated by multiple contributors. The role of prostate microbiota and their metabolites in metastasis, therapeutic resistance to castration resistant prostate cancer (CRPC), and tumor relapse has yet to be fully investigated. Characterization of microflora can provide new mechanistic insights into the functional significance in the emergence of therapeutic resistance, identification of novel effective targeted therapies, and development of biomarkers during prostate cancer progression. The tumor microenvironment (TME) and its components work concurrently with the prostate microbiota in promoting prostate cancer development and progression to metastasis. In this article, we discuss the growing evidence on the functional contribution of microbiota to the phenotypic landscape of the TME and its effect on prostate cancer therapeutic targeting and recurrent disease.
Collapse
Affiliation(s)
- Kasmira R Gupta
- Department of Urology, Icahn School of Medicine at Mount SinaiNew York, New York 10029, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount SinaiNew York, New York 10029, USA
- Oncological Sciences and Pathology & Cell Based Medicine, Icahn School of Medicine at Mount SinaiNew York, New York 10029, USA
| |
Collapse
|
6
|
Yin Z, Gong G, Liu X, Yin J. Mechanism of regulating macrophages/osteoclasts in attenuating wear particle-induced aseptic osteolysis. Front Immunol 2023; 14:1274679. [PMID: 37860014 PMCID: PMC10582964 DOI: 10.3389/fimmu.2023.1274679] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Joint replacement surgery is the most effective treatment for end-stage arthritis. Aseptic loosening caused by periprosthetic osteolysis is a common complication after joint replacement. Inflammation induced by wear particles derived from prosthetic biomaterials is a major cause of osteolysis. We emphasize that bone marrow-derived macrophages and their fusion-derived osteoclasts play a key role in this pathological process. Researchers have developed multiple intervention approaches to regulate macrophage/osteoclast activation. Aiming at wear particle-induced periprosthetic aseptic osteolysis, this review separately discusses the molecular mechanism of regulation of ROS formation and inflammatory response through intervention of macrophage/osteoclast RANKL-MAPKs-NF-κB pathway. These molecular mechanisms regulate osteoclast activation in different ways, but they are not isolated from each other. There is also a lot of crosstalk among the different mechanisms. In addition, other bone and joint diseases related to osteoclast activation are also briefly introduced. Therefore, we discuss these new findings in the context of existing work with a view to developing new strategies for wear particle-associated osteolysis based on the regulation of macrophages/osteoclasts.
Collapse
Affiliation(s)
- Zhaoyang Yin
- Department of Orthopedics, The Affiliated Lianyungang Hospital of Xuzhou Medical University (The First People’s Hospital of Lianyungang), Lianyungang, China
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Liu X, Tan W, Wang W, Feng T, Wang C, Wang L, Zhou W. SEMA4A promotes prostate cancer invasion: involvement of tumor microenvironment. J Cancer 2023; 14:2633-2643. [PMID: 37779872 PMCID: PMC10539395 DOI: 10.7150/jca.86739] [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: 06/02/2023] [Accepted: 07/30/2023] [Indexed: 10/03/2023] Open
Abstract
Semaphorin 4A (SEMA4A) belonged to a family of membrane-bound proteins that were initially recognized as a kind of axon guidance factors in nervous system. It was preferentially expressed on immune cells and has been proven to play a prominent role in immune function and angiogenesis. In this study, we found that SEMA4A was highly expressed in prostate cancer (PCa) tissues and correlated with Gleason scores and distant metastasis. SEMA4A could induce Epithelial-mesenchymal transition (EMT) of PCa cells and consequently promote invasion by establishing a positive loop with IL-10 in stromal cells. In vivo experiments showed more dissemination in mice injected with SEMA4A-overexpressing cells in mouse models and both the number and size of lung metastases were significantly increased in SEMA4A-overexpressing tumors. SEMA4A depletion by genetic means prevents lung metastasis in PCa xenograft models. Our data suggest a crucial role of SEMA4A in PCa and blocking SEMA4A-IL-10 axis represents an attractive approach to improving therapeutic outcomes.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Oncology, Zibo Central Hospital, Zibo, China
| | - Weiwei Tan
- Department of Pathology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Weiqi Wang
- Institute of Radiation Medicine, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Tingting Feng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Chunni Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Zhou
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
8
|
Jin J, Cong J, Lei S, Zhang Q, Zhong X, Su Y, Lu M, Ma Y, Li Z, Wang L, Zhu N, Yang J. Cracking the code: Deciphering the role of the tumor microenvironment in osteosarcoma metastasis. Int Immunopharmacol 2023; 121:110422. [PMID: 37302370 DOI: 10.1016/j.intimp.2023.110422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. It is characterized by a rapid progression, poor prognosis, and early pulmonary metastasis. Over the past 30 years, approximately 85% of patients with osteosarcoma have experienced metastasis. The five-year survival of patients with lung metastasis during the early stages of treatment is less than 20%. The tumor microenvironment (TME) not only provides conditions for tumor cell growth but also releases a variety of substances that can promote the metastasis of tumor cells to other tissues and organs. Currently, there is limited research on the role of the TME in osteosarcoma metastasis. Therefore, to explore methods for regulating osteosarcoma metastasis, further investigations must be conducted from the perspective of the TME. This will help to identify new potential biomarkers for predicting osteosarcoma metastasis and assist in the discovery of new drugs that target regulatory mechanisms for clinical diagnosis and treatment. This paper reviews the research progress on the mechanism of osteosarcoma metastasis based on TME theory, which will provide guidance for the clinical treatment of osteosarcoma.
Collapse
Affiliation(s)
- Jiamin Jin
- Department of Gastroenterology, Affiliated Hospital of Guilin Medical University, Guangxi, Guilin 541001, China; Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Jiacheng Cong
- Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Shangbo Lei
- Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Qiujin Zhang
- Department of Immunology, Guilin Medical University, Guilin 541199, China
| | - Xinyi Zhong
- Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Yingying Su
- Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Mingchuan Lu
- Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Yifen Ma
- Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Zihe Li
- Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China
| | - Liyan Wang
- Department of Gastroenterology, Affiliated Hospital of Guilin Medical University, Guangxi, Guilin 541001, China
| | - Ningxia Zhu
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China.
| | - Jinfeng Yang
- Department of Gastroenterology, Affiliated Hospital of Guilin Medical University, Guangxi, Guilin 541001, China; Department of Immunology, Guilin Medical University, Guilin 541199, China; Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
9
|
Al-Thamiree Mezban S, Fox SW. Genistein and coumestrol reduce MCF-7 breast cancer cell viability and inhibit markers of preferential metastasis, bone matrix attachment and tumor-induced osteoclastogenesis. Arch Biochem Biophys 2023; 740:109583. [PMID: 36967033 DOI: 10.1016/j.abb.2023.109583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
The propensity of breast cancer to preferentially metastasize to the skeleton is well known. Once established in bone metastatic breast cancers have a poor prognosis due to their ability to promote extensive bone loss which augments tumor burden. Unfortunately, current anti-resorptive therapies for skeletal metastasis are typically prescribed after secondary tumors have formed and are palliative in nature. One group of compounds with the potential to reduce both tumor burden and osteolysis are phytoestrogens (PE), but the mechanisms mediating a beneficial effect are unclear. Therefore, the current study examined the effect of genistein and coumestrol alone or in combination on breast cancer cell number, expression of mediators of preferential skeletal metastasis, bone matrix attachment and tumor-induced osteoclast formation. Results showed that genistein and coumestrol significantly reduced viable cell number in an estrogen receptor dependent manner (p < 0.05), whereas combinations of PE had no effect. In addition, genistein and coumestrol significantly reduced expression of genes driving epithelial to mesenchymal transition (snail), bone attachment (CXCR4 and integrin αV) and osteolysis (PTHrP and TNF-α). In keeping with this genistein and coumestrol significantly suppressed attachment of breast cancer cells to bone matrix and inhibited tumor and RANKL-induced osteoclast formation. Our data suggests that phytoestrogens not only decrease breast cancer cell viability but also antagonize essential tumor bone interactions that establish and drive the progression of skeletal metastasis.
Collapse
|
10
|
Provera MD, Straign DM, Karimpour P, Ihle CL, Owens P. Bone morphogenetic protein pathway responses and alterations of osteogenesis in metastatic prostate cancers. Cancer Rep (Hoboken) 2023; 6:e1707. [PMID: 36054271 PMCID: PMC9940003 DOI: 10.1002/cnr2.1707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/07/2022] [Accepted: 07/27/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Prostate cancer is a common cancer in men that annually results in more than 33 000 US deaths. Mortality from prostate cancer is largely from metastatic disease, reflecting on the great strides in the last century of treatments in care for the localized disease. Metastatic castrate resistant prostate cancer (mCRPC) will commonly travel to the bone, creating unique bone pathology that requires nuanced treatments in those sites with surgical, radio and chemotherapeutic interventions. The bone morphogenetic protein (BMP) pathway has been historically studied in the capacity to regulate the osteogenic nature of new bone. New mineralized bone generation is a frequent and common observation in mCRPC and referred to as blastic bone lesions. Less common are bone destructive lesions that are termed lytic. METHODS We queried the cancer genome atlas (TCGA) prostate cancer databases for the expression of the BMP pathway and found that distinct gene expression of the ligands, soluble antagonists, receptors, and intracellular mediators were altered in localized versus metastatic disease. Human prostate cancer cell lines have an innate ability to promote blastic- or lytic-like bone lesions and we hypothesized that inhibiting BMP signaling in these cell lines would result in a distinct change in osteogenesis gene expression with BMP inhibition. RESULTS We found unique and common changes by comparing these cell lines response and unique BMP pathway alterations. We treated human PCa cell lines with distinct bone pathologic phenotypes with the BMP inhibitor DMH1 and found distinct osteogenesis responses. We analyzed distinct sites of metastatic PCa in the TCGA and found that BMP signaling was selectively altered in commons sites such as lymph node, bone and liver compared to primary tumors. CONCLUSIONS Overall we conclude that BMPs in metastatic prostate cancer are important signals and functional mediators of diverse processes that have potential for individualized precision oncology in mCRPC.
Collapse
Affiliation(s)
- Meredith D. Provera
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | - Desiree M. Straign
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | | | - Claire L. Ihle
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
| | - Philip Owens
- Department of PathologyUniversity of Colorado, Anschutz Medical CenterAuroraColoradoUSA
- Department of Veterans Affairs, Research Service, Eastern Colorado Health Care SystemAuroraColoradoUSA
| |
Collapse
|
11
|
Sadu L, Krishnan RH, Akshaya RL, Das UR, Satishkumar S, Selvamurugan N. Exosomes in bone remodeling and breast cancer bone metastasis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 175:120-130. [PMID: 36155749 DOI: 10.1016/j.pbiomolbio.2022.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/10/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Exosomes are endosome-derived microvesicles that carry cell-specific biological cargo, such as proteins, lipids, and noncoding RNAs (ncRNAs). They play a key role in bone remodeling by enabling the maintenance of a balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption. Recent evidence indicates that exosomes disrupt bone remodeling that occurs during breast cancer (BC) progression. The bone is a preferred site for BC metastasis owing to its abundant osseous reserves. In this review, we aimed to highlight the roles of exosomes derived from bone cells and breast tumor in bone remodeling and BC bone metastasis (BCBM). We also briefly outline the mechanisms of action of ncRNAs and proteins carried by exosomes secreted by bone and BCBM. Furthermore, this review highlights the potential of utilizing exosomes as biomarkers or delivery vehicles for the diagnosis and treatment of BCBM.
Collapse
Affiliation(s)
- Lakshana Sadu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India
| | - R Hari Krishnan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India
| | - R L Akshaya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India
| | - Udipt Ranjan Das
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India
| | - Sneha Satishkumar
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India.
| |
Collapse
|
12
|
Emam O, Wasfey EF, Hamdy NM. Notch-associated lncRNAs profiling circuiting epigenetic modification in colorectal cancer. Cancer Cell Int 2022; 22:316. [PMID: 36229883 PMCID: PMC9558410 DOI: 10.1186/s12935-022-02736-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/28/2022] [Indexed: 11/15/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most prevalent digestive cancers, ranking the 2nd cause of cancer-related fatality worldwide. The worldwide burden of CRC is predicted to rise by 60% by 2030. Environmental factors drive, first, inflammation and hence, cancer incidence increase. Main The Notch-signaling system is an evolutionarily conserved cascade, has role in the biological normal developmental processes as well as malignancies. Long non-coding RNAs (LncRNAs) have become major contributors in the advancement of cancer by serving as signal pathways regulators. They can control gene expression through post-translational changes, interactions with micro-RNAs or down-stream effector proteins. Recent emerging evidence has emphasized the role of lncRNAs in controlling Notch-signaling activity, regulating development of several cancers including CRC. Conclusion Notch-associated lncRNAs might be useful prognostic biomarkers or promising potential therapeutic targets for CRC treatment. Therefore, here-in we will focus on the role of “Notch-associated lncRNAs in CRC” highlighting “the impact of Notch-associated lncRNAs as player for cancer induction and/or progression.” Graphical Abstract ![]()
Collapse
Affiliation(s)
| | - Eman F Wasfey
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
13
|
Kang J, La Manna F, Bonollo F, Sampson N, Alberts IL, Mingels C, Afshar-Oromieh A, Thalmann GN, Karkampouna S. Tumor microenvironment mechanisms and bone metastatic disease progression of prostate cancer. Cancer Lett 2022; 530:156-169. [PMID: 35051532 DOI: 10.1016/j.canlet.2022.01.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 01/02/2022] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
During disease progression from primary towards metastatic prostate cancer (PCa), and in particular bone metastases, the tumor microenvironment (TME) evolves in parallel with the cancer clones, altering extracellular matrix composition (ECM), vasculature architecture, and recruiting specialized tumor-supporting cells that favor tumor spread and colonization at distant sites. We introduce the clinical profile of advanced metastatic PCa in terms of common genetic alterations. Findings from recently developed models of PCa metastatic spread are discussed, focusing mainly on the role of the TME (mainly matrix and fibroblast cell types), at distinct stages: premetastatic niche orchestrated by the primary tumor towards the metastatic site and bone metastasis. We report evidence of premetastatic niche formation, such as the mechanisms of distant site conditioning by extracellular vesicles, chemokines and other tumor-derived mechanisms, including altered cancer cell-ECM interactions. Furthermore, evidence supporting the similarities of stroma alterations among the primary PCa and bone metastasis, and contribution of TME to androgen deprivation therapy resistance are also discussed. We summarize the available bone metastasis transgenic mouse models of PCa from a perspective of pro-metastatic TME alterations during disease progression and give an update on the current diagnostic and therapeutic radiological strategies for bone metastasis clinical management.
Collapse
Affiliation(s)
- Juening Kang
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Federico La Manna
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Francesco Bonollo
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Natalie Sampson
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ian L Alberts
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - George N Thalmann
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland; Department of Urology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Sofia Karkampouna
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland.
| |
Collapse
|
14
|
Li X, Baek G, Carreira S, Yuan W, Ma S, Hofstad M, Lee S, Gao Y, Bertan C, Fenor de la Maza MDLD, Alluri PG, Burma S, Chen BP, Raj GV, de Bono J, Pommier Y, Mani RS. Targeting radioresistance and replication fork stability in prostate cancer. JCI Insight 2022; 7:152955. [PMID: 35349486 PMCID: PMC9090241 DOI: 10.1172/jci.insight.152955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
The bromodomain and extraterminal (BET) family of chromatin reader proteins bind to acetylated histones and regulate gene expression. The development of BET inhibitors (BETi) has expanded our knowledge of BET protein function beyond transcriptional regulation and has ushered several prostate cancer (PCa) clinical trials. However, BETi as a single agent is not associated with antitumor activity in patients with castration-resistant prostate cancer (CRPC). We hypothesized novel combinatorial strategies are likely to enhance the efficacy of BETi. By using PCa patient-derived explants and xenograft models, we show that BETi treatment enhanced the efficacy of radiation therapy (RT) and overcame radioresistance. Mechanistically, BETi potentiated the activity of RT by blocking DNA repair. We also report a synergistic relationship between BETi and topoisomerase I (TOP1) inhibitors (TOP1i). We show that the BETi OTX015 synergized with the new class of synthetic noncamptothecin TOP1i, LMP400 (indotecan), to block tumor growth in aggressive CRPC xenograft models. Mechanistically, BETi potentiated the antitumor activity of TOP1i by disrupting replication fork stability. Longitudinal analysis of patient tumors indicated that TOP1 transcript abundance increased as patients progressed from hormone-sensitive prostate cancer to CRPC. TOP1 was highly expressed in metastatic CRPC, and its expression correlated with the expression of BET family genes. These studies open new avenues for the rational combinatorial treatment of aggressive PCa.
Collapse
Affiliation(s)
- Xiangyi Li
- Department of Pathology, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - GuemHee Baek
- Department of Pathology, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - Suzanne Carreira
- Prostate Cancer Targeted Therapy and Cancer Biomarkers Group, The Institute of Cancer Research and The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Wei Yuan
- Prostate Cancer Targeted Therapy and Cancer Biomarkers Group, The Institute of Cancer Research and The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | | | | | - Sora Lee
- Department of Pathology, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - Yunpeng Gao
- Department of Pathology, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - Claudia Bertan
- Prostate Cancer Targeted Therapy and Cancer Biomarkers Group, The Institute of Cancer Research and The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Maria de los Dolores Fenor de la Maza
- Prostate Cancer Targeted Therapy and Cancer Biomarkers Group, The Institute of Cancer Research and The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Prasanna G. Alluri
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sandeep Burma
- Department of Biochemistry and Structural Biology and Department of Neurosurgery, UT Health Science Center, San Antonio, Texas, USA
| | - Benjamin P.C. Chen
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | - Johann de Bono
- Prostate Cancer Targeted Therapy and Cancer Biomarkers Group, The Institute of Cancer Research and The Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, United Kingdom
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Ram S. Mani
- Department of Pathology, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
- Department of Urology and
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Wang J, Du X, Wang X, Xiao H, Jing N, Xue W, Dong B, Gao WQ, Fang YX. Tumor-derived miR-378a-3p-containing extracellular vesicles promote osteolysis by activating the Dyrk1a/Nfatc1/Angptl2 axis for bone metastasis. Cancer Lett 2022; 526:76-90. [PMID: 34801597 DOI: 10.1016/j.canlet.2021.11.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 01/08/2023]
Abstract
Most prostate cancer (PCa)-related deaths are caused by progression to bone metastasis. Recently, the importance of extracellular vesicles (EVs) in pre-metastatic niche formation has been reported. However, whether and how tumor-derived EVs interact with bone marrow macrophages (BMMs) to release EV-delivered microRNAs to promote osteolysis and induce pre-metastatic niche formation for PCa bone metastasis remain unclear. Our in vitro and in vivo functional and mechanistic assays revealed that EV-mediated release of miR-378a-3p from tumor cells was upregulated in bone-metastatic PCa, maintaining low intracellular miR-378a-3p concentration to promote proliferation and MAOA-mediated epithelial-to-mesenchymal transition. Moreover, miR-378a-3p enrichment in tumor-derived EVs was induced by hnRNPA2B1 (a transfer chaperone) overexpression. After tumor-derived EVs were taken in by BMMs, enriched miR-378a-3p promoted osteolytic progression by inhibiting Dyrk1a to improve Nfatc1 (an osteolysis-related transcription factor) nuclear translocation, to activate the expression of downstream target gene Angptl2. As a feedback, increased Angptl2 secretion into the tumor environment promoted PCa progression. In conclusion, tumor-derived miR-378a-3p-containing EVs play a significant role in PCa bone metastasis by activating the Dyrk1a/Nfatc1/Angptl2 axis in BMMs to induce osteolytic progression, making miR-378a-3p a potential predictor of metastatic PCa. Reducing the release of miR-378a-3p-containing EVs or inhibiting the recruitment of miR-378a-3p into EVs can be a therapeutic strategy against PCa metastasis.
Collapse
Affiliation(s)
- Jialin Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiao Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huixiang Xiao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Nan Jing
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Yu-Xiang Fang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
16
|
Sun CY, Mi YY, Ge SY, Hu QF, Xu K, Guo YJ, Tan YF, Zhang Y, Zhong F, Xia GW. Tumor- and Osteoblast-Derived Periostin in Prostate Cancer bone Metastases. Front Oncol 2022; 11:795712. [PMID: 35087756 PMCID: PMC8787093 DOI: 10.3389/fonc.2021.795712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/13/2021] [Indexed: 11/24/2022] Open
Abstract
Exploring the biological function of periostin (POSTN) in prostate cancer (PCa) bone metastasis is of importance. It was observed that the expression of POSTN was high in PCa, especially highest in PCa metastasized to bone. In this study, we found that inhibiting POSTN in PCa cells could significantly alleviate PCa bone metastasis in vivo, suggesting POSTN is a promising therapeutic target. Since, due to the secreted expression of POSTN in osteoblasts and PCa, we hypothesized the positive feedback loop between osteoblasts and PCa mediated by POSTN in PCa bone metastasis. The in vitro experiments demonstrated that osteoblast-derived POSTN promoted PCa cell proliferation and invasion and PCa cell-derived POSTN promotes proliferation of osteoblasts. Furthermore, we found that POSTN regulated PCa and osteoblast function through integrin receptors. Finally, 18F-Alfatide II was used as the molecule probe of integrin αvβ3 in PET-CT, revealing high intake in metastatic lesions. Our findings together indicate that targeting POSTN in PCa cells as well as in the osteoblastic may be an effective treatment for PCa bone metastasis.
Collapse
Affiliation(s)
- Chuan-Yu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuan-Yuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Sheng-Yang Ge
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing-Feng Hu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Jun Guo
- Department of Urology, Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Yi-Fan Tan
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Zhang
- Department of Systems Biology for Medicine, Shanghai Medical College, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fan Zhong
- Department of Systems Biology for Medicine, Shanghai Medical College, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Guo-Wei Xia
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Malkov MI, Lee CT, Taylor CT. Regulation of the Hypoxia-Inducible Factor (HIF) by Pro-Inflammatory Cytokines. Cells 2021; 10:cells10092340. [PMID: 34571989 PMCID: PMC8466990 DOI: 10.3390/cells10092340] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 12/28/2022] Open
Abstract
Hypoxia and inflammation are frequently co-incidental features of the tissue microenvironment in a wide range of inflammatory diseases. While the impact of hypoxia on inflammatory pathways in immune cells has been well characterized, less is known about how inflammatory stimuli such as cytokines impact upon the canonical hypoxia-inducible factor (HIF) pathway, the master regulator of the cellular response to hypoxia. In this review, we discuss what is known about the impact of two major pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), on the regulation of HIF-dependent signaling at sites of inflammation. We report extensive evidence for these cytokines directly impacting upon HIF signaling through the regulation of HIF at transcriptional and post-translational levels. We conclude that multi-level crosstalk between inflammatory and hypoxic signaling pathways plays an important role in shaping the nature and degree of inflammation occurring at hypoxic sites.
Collapse
Affiliation(s)
- Mykyta I. Malkov
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Chee Teik Lee
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Cormac T. Taylor
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; (M.I.M.); (C.T.L.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Correspondence:
| |
Collapse
|
18
|
|
19
|
Patel CM, Wadas TJ, Shiozawa Y. Progress in Targeted Alpha-Particle-Emitting Radiopharmaceuticals as Treatments for Prostate Cancer Patients with Bone Metastases. Molecules 2021; 26:2162. [PMID: 33918705 PMCID: PMC8070008 DOI: 10.3390/molecules26082162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/03/2023] Open
Abstract
Bone metastasis remains a major cause of death in cancer patients, and current therapies for bone metastatic disease are mainly palliative. Bone metastases arise after cancer cells have colonized the bone and co-opted the normal bone remodeling process. In addition to bone-targeted therapies (e.g., bisphosphonate and denosumab), hormone therapy, chemotherapy, external beam radiation therapy, and surgical intervention, attempts have been made to use systemic radiotherapy as a means of delivering cytocidal radiation to every bone metastatic lesion. Initially, several bone-seeking beta-minus-particle-emitting radiopharmaceuticals were incorporated into the treatment for bone metastases, but they failed to extend the overall survival in patients. However, recent clinical trials indicate that radium-223 dichloride (223RaCl2), an alpha-particle-emitting radiopharmaceutical, improves the overall survival of prostate cancer patients with bone metastases. This success has renewed interest in targeted alpha-particle therapy development for visceral and bone metastasis. This review will discuss (i) the biology of bone metastasis, especially focusing on the vicious cycle of bone metastasis, (ii) how bone remodeling has been exploited to administer systemic radiotherapies, and (iii) targeted radiotherapy development and progress in the development of targeted alpha-particle therapy for the treatment of prostate cancer bone metastasis.
Collapse
Affiliation(s)
- Chirayu M. Patel
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| | - Thaddeus J. Wadas
- Department of Radiology, University of Iowa, Iowa City, IA 52242, USA;
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA;
| |
Collapse
|
20
|
Sweeney JD, Debeljak M, Riel S, Millena AC, Eshleman JR, Paller CJ, Odero-Marah V. Val16A SOD2 Polymorphism Promotes Epithelial-Mesenchymal Transition Antagonized by Muscadine Grape Skin Extract in Prostate Cancer Cells. Antioxidants (Basel) 2021; 10:antiox10020213. [PMID: 33535682 PMCID: PMC7912849 DOI: 10.3390/antiox10020213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 01/04/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT), a key event in cancer metastasis, allows polarized epithelial cells to assume mesenchymal morphologies, enhancing invasiveness and migration, and can be induced by reactive oxygen species (ROS). Val16A (Ala) SOD2 polymorphism has been associated with increased prostate cancer (PCa) risk. We hypothesized that SOD2 Ala single nucleotide polymorphism (SNP) may promote EMT. We analyzed SOD2 expression and genotype in various prostate cell lines. Stable overexpression of Ala-SOD2 or Val-SOD2 allele was performed in Lymph Node Carcinoma of the Prostate (LNCaP) cells followed by analysis of intracellular ROS and EMT marker protein expression. Treatments were performed with muscadine grape skin extract (MSKE) antioxidant, with or without addition of H2O2 to provide further oxidative stress. Furthermore, MTS cell proliferation, cell migration, and apoptosis assays were completed. The results showed that SOD2 expression did not correlate with tumor aggressiveness nor SOD2 genotype. We demonstrated that the Ala-SOD2 allele was associated with marked induction of EMT indicated by higher Snail and vimentin, lower E-cadherin, and increased cell migration, when compared to Val-SOD2 allele or Neo control cells. Ala-SOD2 SNP cells exhibited increased levels of total ROS and superoxide and were more sensitive to co-treatment with H2O2 and MSKE, which led to reduced cell growth and increased apoptosis. Additionally, MSKE inhibited Ala-SOD2 SNP-mediated EMT. Our data indicates that treatment with a combination of H2O2-generative drugs, such as certain chemotherapeutics and antioxidants such as MSKE that targets superoxide, hold promising therapeutic potential to halt PCa progression in the future.
Collapse
Affiliation(s)
- Janae D. Sweeney
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA; (J.D.S.); (A.C.M.)
| | - Marija Debeljak
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (M.D.); (S.R.); (J.R.E.)
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Stacy Riel
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (M.D.); (S.R.); (J.R.E.)
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ana Cecilia Millena
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA; (J.D.S.); (A.C.M.)
| | - James R. Eshleman
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (M.D.); (S.R.); (J.R.E.)
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Channing J. Paller
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Valerie Odero-Marah
- Center for Cancer Research and Therapeutic Development and Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA; (J.D.S.); (A.C.M.)
- Correspondence:
| |
Collapse
|
21
|
Blagoev KB, Iordanov R, Zhou M, Fojo T, Bates SE. Drug resistant cells with very large proliferative potential grow exponentially in metastatic prostate cancer. Oncotarget 2021; 12:15-21. [PMID: 33456710 PMCID: PMC7800777 DOI: 10.18632/oncotarget.27855] [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: 09/23/2020] [Accepted: 11/12/2020] [Indexed: 11/25/2022] Open
Abstract
Most metastatic cancers develop drug resistance during treatment and continue to grow, driven by a subpopulation of cancer cells unresponsive to the therapy being administered. There is evidence that metastases are formed by phenotypically plastic cancer cells with stem-cell like properties. Currently the population structure and growth dynamics of the resulting metastatic tumors is unknown. Here, using scaling analysis of clinical data of tumor burden in patients with metastatic prostate cancer, we show that the drug resistant, metastasis-causing cells (MCC) are capable of producing drug resistant, exponentially growing tumors, responsible for tumor growth as a patient receives different treatments.
Collapse
Affiliation(s)
- Krastan B Blagoev
- National Science Foundation, Alexandria, VA 22230, USA.,Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Roumen Iordanov
- Jackson Memorial Hospital, Department of Internal Medicine, Miami, FL 33136, USA
| | - Mengxi Zhou
- Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, NY 10032, USA.,James J. Peters Veterans Affairs Medical Center, Bronx, NY 10032, USA
| | - Tito Fojo
- Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, NY 10032, USA.,James J. Peters Veterans Affairs Medical Center, Bronx, NY 10032, USA
| | - Susan E Bates
- Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, NY 10032, USA.,James J. Peters Veterans Affairs Medical Center, Bronx, NY 10032, USA
| |
Collapse
|
22
|
Laville A, Coutte A, Blanchard P, Sun R, Deutsch E, Latorzeff I. [Treatment of primary disease for synchronous metastatic prostate cancer]. Cancer Radiother 2020; 24:547-553. [PMID: 32855028 DOI: 10.1016/j.canrad.2020.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 11/30/2022]
Abstract
The management of early metastatic prostate cancer is based on systemic treatment by androgen deprivation therapy with or without chemotherapy or next-generation anti-androgen therapies. Local treatment of the prostate was initially used only to alleviate local symptoms. However, local radiotherapy of the prostate has been the subject of retrospective and prospective studies in patients with better prognostic factors, particularly in oligometastatic status. The results of these studies support that prostate radiotherapy can prolong the survival of patients with a low metastatic burden. This article states the biological bases, the main published and future published studies aimed to embed this strategy to optimize therapeutic management.
Collapse
Affiliation(s)
- A Laville
- Département de radiothérapie, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94800 Villejuif, France; Département de radiothérapie, CHU d'Amiens, 1, rue du Professeur-Christian-Cabrol, 80050, France.
| | - A Coutte
- Département de radiothérapie, CHU d'Amiens, 1, rue du Professeur-Christian-Cabrol, 80050, France
| | - P Blanchard
- Département de radiothérapie, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94800 Villejuif, France
| | - R Sun
- Département de radiothérapie, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94800 Villejuif, France
| | - E Deutsch
- Département de radiothérapie, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94800 Villejuif, France; Inserm, UMR 1030 « radiosensibilité des tumeurs et tissus sains », 114, rue Édouard-Vaillant, 94805 Villejuif, France
| | - I Latorzeff
- Département de radiothérapie-oncologie, bâtiment Atrium, clinique Pasteur, 1, rue de la Petite-Vitesse, 31300 Toulouse, France
| |
Collapse
|
23
|
Ortiz-Otero N, Clinch AB, Hope J, Wang W, Reinhart-King CA, King MR. Cancer associated fibroblasts confer shear resistance to circulating tumor cells during prostate cancer metastatic progression. Oncotarget 2020; 11:1037-1050. [PMID: 32256977 PMCID: PMC7105166 DOI: 10.18632/oncotarget.27510] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/06/2020] [Indexed: 12/13/2022] Open
Abstract
Previous studies have demonstrated that CTCs do not travel in the bloodstream alone, but rather are accompanied by clusters of stromal cells such as cancer associated fibroblasts (CAFs). Our laboratory has confirmed the presence of CAFs in the peripheral blood of prostate cancer (PC) patients. The observation that CAFs disseminate with CTCs prompts the examination of the role of CAFs in CTC survival under physiological shear stress during the dissemination process using a clinically relevant, three-dimensional (3D) co-culture model. In this study, we found that "reactive CAFs" induce shear resistance to prostate tumor cells via intercellular contact and soluble derived factors. In addition, these reactive CAFs conserve the proliferative capability of tumor cells in the presence of high magnitude fluid shear stress (FSS). This reactive CAF phenotype emerges from normal fibroblasts (NF), which take on the CAF phenotype when co-cultured with tumor cells. The reactive CAFs showed higher expression of α-smooth muscle actin (α-SMA) and fibroblast activation protein (FAP) compared to differentiated CAFs, when co-cultured with PC cells at the same experimental conditions. Together, we found that the activation mechanism of NF to CAF comprises different stages that progress from a reactive to quiescent cellular state in which these two states are differentiated by the fluctuation of intensity in CAF markers. Here we determined that a reactive state of CAFs proved to be important for supporting tumor cell survival and proliferation. These findings suggest the use of CAFs as a marker for cancer progression and a potential target for novel cancer therapeutics to treat metastatic disease.
Collapse
Affiliation(s)
- Nerymar Ortiz-Otero
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Andrea B Clinch
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37202, USA
| | - Jacob Hope
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37202, USA
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37202, USA
| | | | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37202, USA
| |
Collapse
|
24
|
Cimadamore A, Montironi R, Serni S, Campi R. Seminal Vesicle Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1296:309-318. [PMID: 34185301 DOI: 10.1007/978-3-030-59038-3_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Primary diseases of the seminal vesicles (SV) are very rare entities.Nonneoplastic lesions of the seminal vesicles include amyloidosis, inflammation, calcification and calculi, radiation-induced changes, and basal cell proliferation.Seminal vesicles are frequently involved by tumors originating elsewhere, in particular by prostatic adenocarcinoma, urothelial carcinoma, and rectal adenocarcinoma. On the contrary, primary tumors of the seminal vesicles are rare. Among these, the most common is seminal vesicle adenocarcinoma. To date, less than 100 cases have been reported in literature. Morphologically, primary SV adenocarcinoma is described as a papillary or sheetlike growth architecture, with trabecular and glandular patterns, composed by hobnail tumor cells, frequently with mucinous differentiation. On the contrary, mesenchymal tumors include benign lesions such as leiomyoma, schwannoma, fibroma, paraganglioma, solitary fibrous tumor, cystadenoma, and mixed epithelial and stromal tumors (MEST).Cystadenoma is a rare benign tumor, while MESTs are biphasic tumors with stromal and benign epithelial components. Histological features such as stromal atypia, mitotic activity, nuclear pleomorphism, and tumor necrosis distinct MEST in low-, intermediate-, and high-grade tumors.In recent years, multiple studies reported a link between tumorigenesis and tumor microenvironment. In this regard, the molecular mechanisms connecting prostate cancer (PCa) progression and the host microenvironment have been described and include extracellular matrix (ECM), myofibroblasts, cancer-associated fibroblasts (CAFs), neuroendocrine cells, adipose tissue, and the immune-modulatory cells. Of note, only one study evaluated the influence of seminal vesicle's tumor microenvironment (SVME) on prostate cancer cells so far. Besides, in vivo experiments in NOD/SCID mice clarified the influence of SVME on PCa progression. As such, the injection of PC3 cells into the prostate or the SV resulted in different tumor aggressiveness, and the incidence of retroperitoneal lymph node metastases was significantly higher in mice models receiving SV injection. These findings demonstrated that SVs (rather than the prostate) offer a stimulating tumor microenvironment for growth and invasion of prostate cancer cells.
Collapse
Affiliation(s)
- Alessia Cimadamore
- Institute of Pathological Anatomy and Histopathology, Polytechnic University of the Marche Region (Ancona), Ancona, Italy
| | - Rodolfo Montironi
- Institute of Pathological Anatomy and Histopathology, Polytechnic University of the Marche Region (Ancona), Ancona, Italy
| | - Sergio Serni
- Department of Urologic Robotic Surgery and Renal Transplantation, University of Florence, Careggi Hospital, Florence, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Careggi Hospital, Florence, Italy
| | - Riccardo Campi
- Department of Urologic Robotic Surgery and Renal Transplantation, University of Florence, Careggi Hospital, Florence, Italy. .,Department of Experimental and Clinical Medicine, University of Florence, Careggi Hospital, Florence, Italy.
| |
Collapse
|
25
|
Landgraf M, Lahr CA, Sanchez-Herrero A, Meinert C, Shokoohmand A, Pollock PM, Hutmacher DW, Shafiee A, McGovern JA. Humanized bone facilitates prostate cancer metastasis and recapitulates therapeutic effects of zoledronic acid in vivo. Bone Res 2019; 7:31. [PMID: 31646018 PMCID: PMC6804745 DOI: 10.1038/s41413-019-0072-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/26/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
Advanced prostate cancer (PCa) is known for its high prevalence to metastasize to bone, at which point it is considered incurable. Despite significant effort, there is no animal model capable of recapitulating the complexity of PCa bone metastasis. The humanized mouse model for PCa bone metastasis used in this study aims to provide a platform for the assessment of new drugs by recapitulating the human-human cell interactions relevant for disease development and progression. The humanized tissue-engineered bone construct (hTEBC) was created within NOD-scid IL2rgnull (NSG) mice and was used for the study of experimental PC3-Luc bone metastases. It was confirmed that PC3-Luc cells preferentially grew in the hTEBC compared with murine bone. The translational potential of the humanized mouse model for PCa bone metastasis was evaluated with two clinically approved osteoprotective therapies, the non-species-specific bisphosphonate zoledronic acid (ZA) or the human-specific antibody Denosumab, both targeting Receptor Activator of Nuclear Factor Kappa-Β Ligand. ZA, but not Denosumab, significantly decreased metastases in hTEBCs, but not murine femora. These results highlight the importance of humanized models for the preclinical research on PCa bone metastasis and indicate the potential of the bioengineered mouse model to closely mimic the metastatic cascade of PCa cells to human bone. Eventually, it will enable the development of new effective antimetastatic treatments.
Collapse
Affiliation(s)
- Marietta Landgraf
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Christoph A. Lahr
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Alvaro Sanchez-Herrero
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Christoph Meinert
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Ali Shokoohmand
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Pamela M. Pollock
- School of Biomedical Science, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Dietmar W. Hutmacher
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, Queensland University of Technology, Brisbane, Australia
| | - Abbas Shafiee
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD Australia
| | - Jacqui A. McGovern
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
26
|
Primary Radical Prostatectomy or Ablative Radiotherapy as Protective Factors for Patients With mCRPC Treated With Radium-223 Dichloride: An Italian Multicenter Study. Clin Genitourin Cancer 2019; 18:185-191. [PMID: 32173355 DOI: 10.1016/j.clgc.2019.10.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/02/2019] [Accepted: 10/06/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND We investigated, in a real-life setting, the prognostic relevance of previous primary treatment (radical prostatectomy [RP] or external beam radiotherapy [EBRT]) on overall survival for patients with metastatic castration-resistant prostate cancer (mCRPC) treated with radium-223 (223Ra). MATERIALS AND METHODS In the present multicenter retrospective study, we enrolled 275 consecutive patients. The demographic and clinical data and mCRPC characteristics were recorded and evaluated at baseline and at the end of treatment or progression. 223Ra was administered according to the current label authorization until disease progression or unacceptable toxicity. We divided the whole cohort into 2 groups: those who had undergone primary radical prostatectomy or ablative radiotherapy (RP/EBRT) and those who had not received previous primary treatment (NO). RESULTS Of the 275 patients, 128 (46.5%) were alive and undergoing monitoring at the last follow-up examination, 103 (37.4%) had stopped treatment because of disease progression or the onset of comorbidities, and 147 (53.5%) had died during the study period. Of the 275 patients, 132 were in the RP/EBRT group (48%), of whom 93 had undergone RP and 76 had undergone ablative EBRT, and 143 patients were in the NO group (52%). The data showed a clear advantage for the patients in the RP/EBRT group compared with those in the NO group, with an estimated median survival of 18 versus 11 months, respectively (P < .001). The results from the multivariate analysis corroborated this trend, with a hazard ratio of 0.7 (P = .0443), confirming the better outcome for the RP/EBRT group. CONCLUSIONS Previous radical treatment provides a protective role for patients with mCRPC undergoing 223Ra treatment.
Collapse
|
27
|
Notch3 promotes prostate cancer-induced bone lesion development via MMP-3. Oncogene 2019; 39:204-218. [PMID: 31467432 PMCID: PMC6938550 DOI: 10.1038/s41388-019-0977-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 03/29/2019] [Accepted: 06/18/2019] [Indexed: 01/12/2023]
Abstract
Prostate cancer metastases primarily localize in the bone where they induce a unique osteoblastic response. Elevated Notch activity is associated with high-grade disease and metastasis. To address how Notch affects prostate cancer bone lesions, we manipulated Notch expression in mouse tibia xenografts and monitored tumor growth, lesion phenotype, and the bone microenvironment. Prostate cancer cell lines that induce mixed osteoblastic lesions in bone expressed 5–6 times more Notch3, than tumor cells that produce osteolytic lesions. Expression of active Notch3 (NICD3) in osteolytic tumors reduced osteolytic lesion area and enhanced osteoblastogenesis, while loss of Notch3 in osteoblastic tumors enhanced osteolytic lesion area and decreased osteoblastogensis. This was accompanied by a respective decrease and increase in the number of active osteoclasts and osteoblasts at the tumor-bone interface, without any effect on tumor proliferation. Conditioned medium from NICD3-expressing cells enhanced osteoblast differentiation and proliferation in vitro, while simultaneously inhibiting osteoclastogenesis. MMP-3 was specifically elevated and secreted by NICD3-expressing tumors, and inhibition of MMP-3 rescued the NICD3-induced osteoblastic phenotypes. Clinical osteoblastic bone metastasis samples had higher levels of Notch3 and MMP-3 compared to patient matched visceral metastases or osteolytic metastasis samples. We identified a Notch3-MMP-3 axis in human prostate cancer bone metastases that contributes to osteoblastic lesion formation by blocking osteoclast differentiation, while also contributing to osteoblastogenesis. These studies define a new role for Notch3 in manipulating the tumor microenvironment in bone metastases.
Collapse
|
28
|
Murray NP, Aedo S, Fuentealba C, Reyes E, Salazar A, Lopez MA, Minzer S, Orrego S, Guzman E. Subtypes of minimal residual disease, association with Gleason score, risk and time to biochemical failure in pT2 prostate cancer treated with radical prostatectomy. Ecancermedicalscience 2019; 13:934. [PMID: 31281431 PMCID: PMC6605630 DOI: 10.3332/ecancer.2019.934] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Indexed: 01/27/2023] Open
Abstract
INTRODUCTION The Gleason score is a strong prognostic factor for treatment failure in pathologically organ-confined prostate cancer (pT2) treated by radical prostatectomy (RP). However, within each Gleason score, there is clinical heterogeneity with respect to treatment outcome, even in patients with the same pathological stage and prostate-specific antigen (PSA) at diagnosis. This may be due to minimal residual disease (MRD) remaining after surgery. We hypothesise that the sub-type of MRD determines the risk of and timing of treatment failure, is a biological classification, and may explain in part clinical heterogeneity. We present a study of pT2 patients treated with RP, the subtypes of MRD for each Gleason score and clinical outcomes. PATIENTS AND METHODS Patients with Gleason ≤6 (G6) or Gleason 7 (G7) pT2 cancer participated in the study. One month after surgery, blood was taken for circulating prostate cell (CPCs); mononuclear cells were obtained by differential gel centrifugation and identified using immunocytochemistry with anti-PSA. The detection of one CPC/sample was defined as a positive test. Touch-preparations from bone-marrow biopsies were used to detect micro-metastasis using immunocytochemistry with anti-PSA. Biochemical failure was defined as a PSA >0.2 ng/mL. Patients were classified as: Group A MRD negative (CPC and micro-metastasis negative), Group B (only micro-metastasis positive) and Group C (CPC positive). Biochemical failure-free survival (BFFS) using Kaplan-Meier and time to failure using Restricted Mean Survival Time (RMST) after 10 years of follow-up were calculated for each group based on the Gleason score. RESULTS Of a cohort of 253 men, four were excluded for having Gleason 8 or 9 prostate cancer, leaving a study group of 249 men of whom 52 had G7 prostate cancer. G7 patients had a higher frequency of MRD (69% versus 36%) and worse prognosis. G6 and G7 patients negative for MRD had similar BBFS rates, 98% at 10 years, time to failure 9.9 years. Group C, G6 patients had a higher BFFS and longer time to failure compared to G7 patients (19% versus 5% and 7 versus 3 years). Group B showed similar results up to 5 years, thereafter G6 had a lower BFFS 63% versus 90%. CONCLUSIONS G7 and G6 pT2 patients have different patterns of MRD and relapse. Risk stratification using MRD sub-types may help to define the need for adjuvant therapy. This needs confirmation with large randomised long-term trials.
Collapse
Affiliation(s)
- Nigel P Murray
- Faculty of Medicine, University Finis Terrae, Pedro de Valdivia 1509, Providencia, Santiago 7501015, Chile
- Urology Service, Hospital de Carabineros, Simón Bolívar 2200, Ñuñoa, Santiago 7770199, Chile
| | - Socrates Aedo
- Faculty of Medicine, University Finis Terrae, Pedro de Valdivia 1509, Providencia, Santiago 7501015, Chile
| | - Cynthia Fuentealba
- Urology Service, Hospital de Carabineros, Simón Bolívar 2200, Ñuñoa, Santiago 7770199, Chile
| | - Eduardo Reyes
- Faculty of Medicine, University Diego Portales, Manuel Rodríguez Sur 415, Santiago 8370179, Chile
- Urology Service, Hospital DIPRECA, Vital Apoquindo 1200, Las Condes, Santiago 7601003, Chile
| | - Anibal Salazar
- Urology Service, Hospital de Carabineros, Simón Bolívar 2200, Ñuñoa, Santiago 7770199, Chile
| | - Marco Antonio Lopez
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| | - Simona Minzer
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| | - Shenda Orrego
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| | - Eghon Guzman
- Faculty of Medicine, University Mayor, San Pio X 2422, Providencia, Santiago 7510041, Chile
| |
Collapse
|
29
|
Frank S, Nelson P, Vasioukhin V. Recent advances in prostate cancer research: large-scale genomic analyses reveal novel driver mutations and DNA repair defects. F1000Res 2018; 7. [PMID: 30135717 PMCID: PMC6073096 DOI: 10.12688/f1000research.14499.1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is a disease of mutated and misregulated genes. However, primary prostate tumors have relatively few mutations, and only three genes (
ERG,
PTEN, and
SPOP) are recurrently mutated in more than 10% of primary tumors. On the other hand, metastatic castration-resistant tumors have more mutations, but, with the exception of the androgen receptor gene (
AR), no single gene is altered in more than half of tumors. Structural genomic rearrangements are common, including
ERG fusions, copy gains involving the
MYC locus, and copy losses containing
PTEN. Overall, instead of being associated with a single dominant driver event, prostate tumors display various combinations of modifications in oncogenes and tumor suppressors. This review takes a broad look at the recent advances in PCa research, including understanding the genetic alterations that drive the disease and how specific mutations can sensitize tumors to potential therapies. We begin with an overview of the genomic landscape of primary and metastatic PCa, enabled by recent large-scale sequencing efforts. Advances in three-dimensional cell culture techniques and mouse models for PCa are also discussed, and particular emphasis is placed on the benefits of patient-derived xenograft models. We also review research into understanding how ETS fusions (in particular,
TMPRSS2-ERG) and
SPOP mutations contribute to tumor initiation. Next, we examine the recent findings on the prevalence of germline DNA repair mutations in about 12% of patients with metastatic disease and their potential benefit from the use of poly(ADP-ribose) polymerase (PARP) inhibitors and immune modulation. Lastly, we discuss the recent increased prevalence of AR-negative tumors (neuroendocrine and double-negative) and the current state of immunotherapy in PCa. AR remains the primary clinical target for PCa therapies; however, it does not act alone, and better understanding of supporting mutations may help guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sander Frank
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Departments of Medicine and Urology, University of Washington, Seattle, WA 98195, USA.,Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Valeri Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
30
|
Tong HB, Zou CL, Qin SY, Meng J, Keller ET, Zhang J, Lu Y. Prostate cancer tends to metastasize in the bone-mimicking microenvironment via activating NF-κB signaling. J Biomed Res 2018; 32:343-353. [PMID: 30190448 PMCID: PMC6163113 DOI: 10.7555/jbr.32.20180035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Prostate cancer preferentially metastasizes to the bone. However, the underlying molecular mechanisms are still unclear. To explore the effects of a bone-mimicking microenvironment on PC3 prostate cancer cell growth and metastasis, we used osteoblast differentiation medium (ODM; minimal essential medium alpha supplemented with L-ascorbic acid) to mimic the bone microenvironment. PC3 cells grown in ODM underwent epithelial-mesenchymal transition and showed enhanced colony formation, migration, and invasion abilities compared to the cells grown in normal medium. PC3 cells grown in ODM showed enhanced metastasis when injected in mice. A screening of signaling pathways related to invasion and metastasis revealed that the NF-κB pathway was activated, which could be reversed by Bay 11-7082, a NF-κB pathway inhibitor. These results indicate that the cells in different culture conditions manifested significantly different biological behaviors and the NF-κB pathway is a potential therapeutic target for prostate cancer bone metastasis.
Collapse
Affiliation(s)
- Hai-Bo Tong
- Key Laboratory of Longevity and Aging-related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, China.,Southern University of Science and Technology School of Medicine, Shenzhen, Guangdong 518055, China
| | - Chun-Lin Zou
- Key Laboratory of Longevity and Aging-related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, China
| | - Si-Yuan Qin
- Key Laboratory of Longevity and Aging-related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, China.,Southern University of Science and Technology School of Medicine, Shenzhen, Guangdong 518055, China
| | - Jie Meng
- Key Laboratory of Longevity and Aging-related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, China.,Southern University of Science and Technology School of Medicine, Shenzhen, Guangdong 518055, China
| | - Evan T Keller
- Department of Pathology and Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jian Zhang
- Southern University of Science and Technology School of Medicine, Shenzhen, Guangdong 518055, China.,Department of Pathology and Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yi Lu
- Key Laboratory of Longevity and Aging-related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, China.,Southern University of Science and Technology School of Medicine, Shenzhen, Guangdong 518055, China
| |
Collapse
|
31
|
Flores-Morales A, Iglesias-Gato D. Quantitative Mass Spectrometry-Based Proteomic Profiling for Precision Medicine in Prostate Cancer. Front Oncol 2017; 7:267. [PMID: 29164064 PMCID: PMC5674010 DOI: 10.3389/fonc.2017.00267] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is one of the most frequently diagnosed cancer among men in the western societies. Many PCa patients bear tumors that will not threat their lives if left untreated or if treatment is delayed. Our inability for early identification of these patients has resulted in massive overtreatment. Therefore, there is a great need of finding biomarkers for patient stratification according to prognostic risk; as well as there is a need for novel targets that can allow the development of effective treatments for patients that progress to castration-resistant PCa. Most biomarkers in cancer are proteins, including the widely-used prostate-specific antigen (PSA). Recent developments in mass spectrometry allow the identification and quantification of thousands of proteins and posttranslational modifications from small amounts of biological material, including formalin-fixed paraffin-embedded tissues, and biological fluids. Novel diagnostic and prognostic biomarkers have been identified in tissue, blood, urine, and seminal plasma of PCa patients, and new insights in the ethology and progression of this disease have been achieved using this technology. In this review, we summarize these findings and discuss the potential of this technology to pave the way toward the clinical implementation of precision medicine in PCa.
Collapse
Affiliation(s)
- Amilcar Flores-Morales
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Diego Iglesias-Gato
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| |
Collapse
|
32
|
Runa F, Hamalian S, Meade K, Shisgal P, Gray PC, Kelber JA. Tumor microenvironment heterogeneity: challenges and opportunities. ACTA ACUST UNITED AC 2017; 3:218-229. [PMID: 29430386 DOI: 10.1007/s40610-017-0073-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment (TME) has been recognized as an integral component of malignancies in breast and prostate tissues, contributing in confounding ways to tumor progression, metastasis, therapy resistance and disease recurrence. Major components of the TME are immune cells, fibroblasts, pericytes, endothelial cells, mesenchymal stroma/stem cells (MSCs), and extracellular matrix (ECM) components. Herein, we discuss the molecular and cellular heterogeneity within the TME and how this presents unique challenges and opportunities for treating breast and prostate cancers.
Collapse
Affiliation(s)
- F Runa
- Department of Biology, California State University, Northridge, CA
| | - S Hamalian
- Department of Biology, California State University, Northridge, CA
| | - K Meade
- Department of Biology, California State University, Northridge, CA
| | - P Shisgal
- Department of Biology, California State University, Northridge, CA
| | - P C Gray
- The Salk Institute for Biological Studies, La Jolla, CA
| | - J A Kelber
- Department of Biology, California State University, Northridge, CA
| |
Collapse
|
33
|
Ye Y, Li SL, Ma YY, Diao YJ, Yang L, Su MQ, Li Z, Ji Y, Wang J, Lei L, Fan WX, Li LX, Xu Y, Hao XK. Exosomal miR-141-3p regulates osteoblast activity to promote the osteoblastic metastasis of prostate cancer. Oncotarget 2017; 8:94834-94849. [PMID: 29212270 PMCID: PMC5706916 DOI: 10.18632/oncotarget.22014] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 09/05/2017] [Indexed: 12/26/2022] Open
Abstract
Exosomes from cancer cells, which contain microRNA and reach metastasis loci prior to cancer cells, stimulate the formation of a metastatic microenvironment. Previous studies have shown that exosomal miR-141-3p is associated with metastatic prostate cancer (PCa). However, the role and regulatory mechanism of miR-141-3p in the microenvironment of bone metastases require further study. In this study, we performed a series of experiments in vivo and in vitro to determine whether exosomal miR-141-3p from MDA PCa 2b cells regulates osteoblast activity to promote osteoblastic metastasis. We demonstrate that extracts obtained from cell culture supernatants contained exosomes and that miR-141-3p levels were significantly higher in MDA PCa 2b cell exosomes. Via confocal imaging, numerous MDA PCa 2b exosomes were observed to enter osteoblasts, and miR-141-3p was transferred to osteoblasts through MDA PCa 2b exosomes in vitro. Exosomal miR-141-3p from MDA PCa 2b promoted osteoblast activity and increased osteoprotegerin OPG expression. miR-141-3p suppressed the protein levels of the target gene DLC1, indicating its functional significance in activating the p38MAPK pathway. In animal experiments, exosomal miR-141-3p had bone-target specificity and promoted osteoblast activity. Mice injected with miR-141-3p-mimics exosomes developed apparent osteoblastic bone metastasis. Exosomal miR-141-3p from MDA PCa 2b cells promoted osteoblast activity and regulated the microenvironment of bone metastases, which plays an important role in the formation of bone metastases and osteogenesis damage in PCa. Clarifying the specific mechanism of bone metastasis will help generate new possibilities for the treatment of PCa.
Collapse
Affiliation(s)
- Yun Ye
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Su-Liang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Yue-Yun Ma
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yan-Jun Diao
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Liu Yang
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming-Quan Su
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhuo Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Yang Ji
- Department of Radiology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, China
| | - Juan Wang
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Lin Lei
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wei-Xiao Fan
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - La-Xiu Li
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yi Xu
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao-Ke Hao
- Department of Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
34
|
Canesin G, Evans-Axelsson S, Hellsten R, Krzyzanowska A, Prasad CP, Bjartell A, Andersson T. Treatment with the WNT5A-mimicking peptide Foxy-5 effectively reduces the metastatic spread of WNT5A-low prostate cancer cells in an orthotopic mouse model. PLoS One 2017; 12:e0184418. [PMID: 28886116 PMCID: PMC5590932 DOI: 10.1371/journal.pone.0184418] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/23/2017] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer patients with high WNT5A expression in their tumors have been shown to have more favorable prognosis than those with low WNT5A expression. This suggests that reconstitution of Wnt5a in low WNT5A-expressing tumors might be an attractive therapeutic approach. To explore this idea, we have in the present study used Foxy-5, a WNT5A mimicking peptide, to investigate its impact on primary tumor and metastasis in vivo and on prostate cancer cell viability, apoptosis and invasion in vitro. We used an in vivo orthotopic xenograft mouse model with metastatic luciferase-labeled WNT5A-low DU145 cells and metastatic luciferase-labeled WNT5A-high PC3prostate cancer cells. We provide here the first evidence that Foxy-5 significantly inhibits the initial metastatic dissemination of tumor cells to regional and distal lymph nodes by 90% and 75%, respectively. Importantly, this effect was seen only with the WNT5A-low DU145 cells and not with the WNT5A-high PC3 cells. The inhibiting effect in the DU145-based model occurred despite the fact that no effects were observed on primary tumor growth, apoptosis or proliferation. These findings are consistent with and supported by the in vitro data, where Foxy-5 specifically targets invasion without affecting apoptosis or viability of WNT5A-low prostate cancer cells. To conclude, our data indicate that the WNT5A-mimicking peptide Foxy-5, which has been recently used in a phase 1 clinical trial, is an attractive candidate for complimentary anti-metastatic treatment of prostate cancer patients with tumors exhibiting absent or low WNT5A expression.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Translational Medicine, Division of Cell and Experimental Pathology, Lund University, Clinical Research Centre, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Susan Evans-Axelsson
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Rebecka Hellsten
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Agnieszka Krzyzanowska
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Chandra P. Prasad
- Department of Translational Medicine, Division of Cell and Experimental Pathology, Lund University, Clinical Research Centre, Skåne University Hospital Malmö, Malmö, Sweden
| | - Anders Bjartell
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Tommy Andersson
- Department of Translational Medicine, Division of Cell and Experimental Pathology, Lund University, Clinical Research Centre, Skåne University Hospital Malmö, Malmö, Sweden
- * E-mail:
| |
Collapse
|
35
|
Sun Y, Fan X, Zhang Q, Shi X, Xu G, Zou C. Cancer-associated fibroblasts secrete FGF-1 to promote ovarian proliferation, migration, and invasion through the activation of FGF-1/FGFR4 signaling. Tumour Biol 2017; 39:1010428317712592. [PMID: 28718374 DOI: 10.1177/1010428317712592] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy, due to its high propensity for metastasis. Cancer-associated fibroblasts, as the dominant component of tumor microenvironment, are crucial for tumor progression. However, the mechanisms underlying the regulation of ovarian cancer cells by cancer-associated fibroblasts remain little known. Here, we first isolated cancer-associated fibroblasts from patients' ovarian tissues and found that cancer-associated fibroblasts promoted SKOV3 cells' proliferation, migration, and invasion. Fibroblast growth factor-1 was identified as a highly increased factor in cancer-associated fibroblasts compared with normal fibroblasts by quantitative reverse transcription polymerase chain reaction (~4.6-fold, p < 0.01) and ELISA assays (~4-fold, p < 0.01). High expression of fibroblast growth factor-1 in cancer-associated fibroblasts either naturally or through gene recombination led to phosphorylation of fibroblast growth factor receptor 4 in SKOV3 cells, which is followed by the activation of mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathway and epithelial-to-mesenchymal transition-associated gene Snail1 and MMP3 expression. Moreover, treatment of SKOV3 cell with fibroblast growth factor receptor inhibitor PD173074 terminated cellular proliferation, migration, and invasion, reduced the phosphorylation level of fibroblast growth factor receptor 4, and suppressed the activation of mitogen-activated protein kinase/extracellular signal-regulated protein kinase pathway. In addition, the expression level of Snail1 and MMP3 was reduced, while the expression level of E-cadherin increased. These observations suggest a crucial role for cancer-associated fibroblasts and fibroblast growth factor-1/fibroblast growth factor receptor 4 signaling in the progression of ovarian cancer. Therefore, this fibroblast growth factor-1/fibroblast growth factor receptor 4 axis may become a potential target for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yuanzhen Sun
- 1 Department of Laboratory, Laiwu Maternal and Child Health Care Hospital, Laiwu, China
| | - Xiaoli Fan
- 2 Department of Occupational Poisoning, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, China
| | - Qing Zhang
- 3 Department of Laboratory, Shandong Provincial Hospital, Jinan, China
| | - Xiaoyu Shi
- 1 Department of Laboratory, Laiwu Maternal and Child Health Care Hospital, Laiwu, China
| | - Guangwei Xu
- 4 Department of Laboratory, Weihaiwei People's Hospital, Weihai, China
| | - Cuimin Zou
- 4 Department of Laboratory, Weihaiwei People's Hospital, Weihai, China
| |
Collapse
|
36
|
Chen PC, Tang CH, Lin LW, Tsai CH, Chu CY, Lin TH, Huang YL. Thrombospondin-2 promotes prostate cancer bone metastasis by the up-regulation of matrix metalloproteinase-2 through down-regulating miR-376c expression. J Hematol Oncol 2017; 10:33. [PMID: 28122633 PMCID: PMC5264454 DOI: 10.1186/s13045-017-0390-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/03/2017] [Indexed: 11/24/2022] Open
Abstract
Background Thrombospondin-2 (TSP-2) is a secreted matricellular glycoprotein that is found to mediate cell-to-extracellular matrix attachment and participates in many physiological and pathological processes. The expression profile of TSP-2 on tumors is controversial, and it up-regulates in some cancers, whereas it down-regulates in others, suggesting that the functional role of TSP-2 on tumors is still uncertain. Methods The expression of TSP-2 on prostate cancer progression was determined in the tissue array by the immunohistochemistry. The molecular mechanism of TSP-2 on prostate cancer (PCa) metastasis was investigated through pharmaceutical inhibitors, siRNAs, and miRNAs analyses. The role of TSP-2 on PCa metastasis in vivo was verified through xenograft in vivo imaging system. Results Based on the gene expression omnibus database and immunohistochemistry, we found that TSP-2 increased with the progression of PCa, especially in metastatic PCa and is correlated with the matrix metalloproteinase-2 (MMP-2) expression. Additionally, through binding to CD36 and integrin ανβ3, TSP-2 increased cell migration and MMP-2 expression. With inhibition of p38, ERK, and JNK, the TSP-2-induced cell migration and MMP-2 expression were abolished, indicating that the TSP-2’s effect on PCa is MAPK dependent. Moreover, the microRNA-376c (miR-376c) was significantly decreased by the TSP-2 treatment. Furthermore, the TSP-2-induced MMP-2 expression and the subsequent cell motility were suppressed upon miR-376c mimic stimulation. On the other hand, the animal studies revealed that the bone metastasis was abolished when TSP-2 was stably knocked down in PCa cells. Conclusions Taken together, our results indicate that TSP-2 enhances the migration of PCa cells by increasing MMP-2 expression through down-regulation of miR-376c expression. Therefore, TSP-2 may represent a promising new target for treating PCa. Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0390-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Po-Chun Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Liang-Wei Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Cheng-Ying Chu
- The Ph.D. Program for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Tien-Huang Lin
- Department of Urology, Buddhist Tzu Chi General Hospital Taichung Branch, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
37
|
Aprile C, Persico MG, Lodola L, Buroni FE. Radium-223 and metastatic castration-resistant prostate cancer: All that glitters is not gold. World J Radiol 2016; 8:816-818. [PMID: 27843540 PMCID: PMC5084059 DOI: 10.4329/wjr.v8.i10.816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 08/04/2016] [Accepted: 08/29/2016] [Indexed: 02/06/2023] Open
Abstract
After being approved by the National Drug Agency in several countries, Radium-223 (Ra-223) is gaining wide acceptance in the treatment of bone metastatic castration resistant prostate cancer. The exact mechanism of action remain unclear: The established model of direct alpha-particle irradiation from the remodelling bone surface, where Ra-223 accumulates, surrounding the tumor foci can explain a lethal effect only on metastatic microdeposits, but not on higher tumor burden. According to the “pre-metastatic niche model”, it is likely that Ra-223 targets several non-tumoral cell types of the tumor microenvironment involved in the complex mechanism of cancer bone homing and colonization. A deeper insight into this hypothetical mechanism will lead to a more accurate dosimetric approach and to find optimal sequencing and/or combination with the other therapeutic options.
Collapse
|
38
|
Human Bone Xenografts: from Preclinical Testing for Regenerative Medicine to Modeling of Diseases. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s40610-016-0044-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
39
|
Sternberg C, Armstrong A, Pili R, Ng S, Huddart R, Agarwal N, Khvorostenko D, Lyulko O, Brize A, Vogelzang N, Delva R, Harza M, Thanos A, James N, Werbrouck P, Bögemann M, Hutson T, Milecki P, Chowdhury S, Gallardo E, Schwartsmann G, Pouget JC, Baton F, Nederman T, Tuvesson H, Carducci M. Randomized, Double-Blind, Placebo-Controlled Phase III Study of Tasquinimod in Men With Metastatic Castration-Resistant Prostate Cancer. J Clin Oncol 2016; 34:2636-43. [PMID: 27298414 DOI: 10.1200/jco.2016.66.9697] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Tasquinimod, a novel oral therapy targeting the tumor microenvironment, significantly improved progression-free survival (PFS) in a randomized, placebo-controlled phase II trial in men with metastatic castration-resistant prostate cancer (mCRPC). This phase III study was conducted to confirm the phase II results and to detect an overall survival (OS) benefit. PATIENTS AND METHODS Men with chemotherapy-naïve mCRPC and evidence of bone metastases were assigned (2:1) to receive tasquinimod once per day or placebo until progression or toxicity. The primary end point was radiographic PFS (rPFS; time from random assignment to radiologic progression or death) per Prostate Cancer Working Group 2 criteria and RECIST 1.1. The study had 99.9% power to detect an rPFS hazard ratio (HR) of 0.6 with a two-sided alpha error of .05 and 80% power to detect a target HR of 0.8 for OS, the key secondary end point. RESULTS In all, 1,245 patients were randomly assigned to either tasquinimod (n = 832) or placebo (n = 413) between March 2011 and December 2012 at 241 sites in 37 countries. Baseline characteristics were balanced between groups: median age, 71 years; Karnofsky performance status ≥ 90%, 77.3%; and visceral metastases, 21.1%. Estimated median rPFS by central review was 7.0 months (95% CI, 5.8 to 8.2 months) with tasquinimod and 4.4 months (95% CI, 3.5 to 5.5 months) with placebo (HR, 0.64; 95% CI, 0.54 to 0.75; P < .001). Median OS was 21.3 months (95% CI, 19.5 to 23.0 months) with tasquinimod and 24.0 months (95% CI, 21.4 to 26.9 months) with placebo (HR, 1.10; 95% CI, 0.94 to 1.28; P = .25). Grade ≥ 3 adverse events were more frequent with tasquinimod (42.8% v 33.6%), the most common being anemia, fatigue, and cancer pain. CONCLUSION In chemotherapy-naïve men with mCRPC, tasquinimod significantly improved rPFS compared with placebo. However, no OS benefit was observed.
Collapse
Affiliation(s)
- Cora Sternberg
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Andrew Armstrong
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Roberto Pili
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Siobhan Ng
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Robert Huddart
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Neeraj Agarwal
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Denis Khvorostenko
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Olexiy Lyulko
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Arija Brize
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Nicholas Vogelzang
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Rémy Delva
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Mihai Harza
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Anastasios Thanos
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Nicholas James
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Patrick Werbrouck
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Martin Bögemann
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Thomas Hutson
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Piotr Milecki
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Simon Chowdhury
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Enrique Gallardo
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Gilberto Schwartsmann
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Jean-Christophe Pouget
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Frédérique Baton
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Thore Nederman
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Helen Tuvesson
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Michael Carducci
- Cora Sternberg, San Camillo Forlanini Hospitals, Rome, Italy; Andrew Armstrong, Duke Cancer Institute, Duke University, Durham, NC; Roberto Pili, Indiana University School of Medicine, Indianapolis, IN; Siobhan Ng, St John of God Medical Centre, Subiaco, Western Australia, Australia; Robert Huddart, Royal Marsden Hospital, Sutton; Nicholas James, Queen Elizabeth Hospital, Birmingham; Simon Chowdhury, Guy's Hospital and Sarah Cannon Research UK, London, United Kingdom; Neeraj Agarwal, University of Utah, Salt Lake City, UT; Denis Khvorostenko, Leningrad Regional Oncology Dispensary, St Petersburg, Russia; Olexiy Lyulko, Zaporizhzhya Regional Clinical Hospital, Zaporizhzhya, Ukraine; Arija Brize, Riga Eastern Clinical University Hospital, Riga, Latvia; Nicholas Vogelzang, Comprehensive Cancer Centers of Nevada, Las Vegas, NV; Rémy Delva, Centre Régional de Lutte Contre le Cancer Paul Papin, Angers; Jean-Christophe Pouget and Frédérique Baton, Ipsen Innovation, Les Ulis, France; Mihai Harza, Fundeni Clinical Institute, Bucharest, Romania; Anastasios Thanos, Agios Savas Anticancer Oncology Hospital of Athens, Athens, Greece; Patrick Werbrouck, Algemeen Ziekenhuis Groeninge, Kortrijk, Belgium; Martin Bögemann, Universitätsklinikum Münster, Münster, Germany; Thomas Hutson, Texas Oncology, Dallas, TX; Piotr Milecki, Poznan University of Medical Sciences, Poznan, Poland; Enrique Gallardo, Corporació Sanitaria Parc Taulí, Sabadell, Spain; Gilberto Schwartsmann, Hospital De Clinicas De Porto Alegre, Porto Alegre, Brazil; Thore Nederman and Helen Tuvesson, Active Biotech, Lund, Sweden; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
40
|
Novío S, Cartea ME, Soengas P, Freire-Garabal M, Núñez-Iglesias MJ. Effects of Brassicaceae Isothiocyanates on Prostate Cancer. Molecules 2016; 21:E626. [PMID: 27187332 PMCID: PMC6272898 DOI: 10.3390/molecules21050626] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/13/2016] [Accepted: 05/03/2016] [Indexed: 12/21/2022] Open
Abstract
Despite the major progress made in the field of cancer biology, cancer is still one of the leading causes of mortality, and prostate cancer (PCa) is one of the most encountered malignancies among men. The effective management of this disease requires developing better anticancer agents with greater efficacy and fewer side effects. Nature is a large source for the development of chemotherapeutic agents, with more than 50% of current anticancer drugs being of natural origin. Isothiocyanates (ITCs) are degradation products from glucosinolates that are present in members of the family Brassicaceae. Although they are known for a variety of therapeutic effects, including antioxidant, immunostimulatory, anti-inflammatory, antiviral and antibacterial properties, nowadays, cell line and animal studies have additionally indicated the chemopreventive action without causing toxic side effects of ITCs. In this way, they can induce cell cycle arrest, activate apoptosis pathways, increase the sensitivity of resistant PCa to available chemodrugs, modulate epigenetic changes and downregulate activated signaling pathways, resulting in the inhibition of cell proliferation, progression and invasion-metastasis. The present review summarizes the chemopreventive role of ITCs with a particular emphasis on specific molecular targets and epigenetic alterations in in vitro and in vivo cancer animal models.
Collapse
Affiliation(s)
- Silvia Novío
- Lennart Levi Stress and Neuroimmunology Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, 15782 Santiago de Compostela, A Coruña, Spain.
| | - María Elena Cartea
- Group of Genetics, Breeding and Biochemistry of Brassicas, Misión Biológica de Galicia (CSIC) Aptdo. 28, 36080 Pontevedra, Spain.
| | - Pilar Soengas
- Group of Genetics, Breeding and Biochemistry of Brassicas, Misión Biológica de Galicia (CSIC) Aptdo. 28, 36080 Pontevedra, Spain.
| | - Manuel Freire-Garabal
- Lennart Levi Stress and Neuroimmunology Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, 15782 Santiago de Compostela, A Coruña, Spain.
| | - María Jesús Núñez-Iglesias
- Lennart Levi Stress and Neuroimmunology Laboratory, School of Medicine and Dentistry, University of Santiago de Compostela, c/San Francisco, s/n, 15782 Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
41
|
Wu AC, He Y, Broomfield A, Paatan NJ, Harrington BS, Tseng HW, Beaven EA, Kiernan DM, Swindle P, Clubb AB, Levesque JP, Winkler IG, Ling MT, Srinivasan B, Hooper JD, Pettit AR. CD169(+) macrophages mediate pathological formation of woven bone in skeletal lesions of prostate cancer. J Pathol 2016; 239:218-30. [PMID: 27174786 DOI: 10.1002/path.4718] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/04/2016] [Accepted: 03/08/2016] [Indexed: 12/31/2022]
Abstract
Skeletal metastases present a major clinical challenge for prostate cancer patient care, inflicting distinctive mixed osteoblastic and osteolytic lesions that cause morbidity and refractory skeletal complications. Macrophages are abundant in bone and bone marrow and can influence both osteoblast and osteoclast function in physiology and pathology. Herein, we examined the role of macrophages in prostate cancer bone lesions, particularly the osteoblastic response. First, macrophage and lymphocyte distributions were qualitatively assessed in patient's prostate cancer skeletal lesions by immunohistochemistry. Second, macrophage functional contributions to prostate tumour growth in bone were explored using an immune-competent mouse model combined with two independent approaches to achieve in vivo macrophage depletion: liposome encapsulated clodronate that depletes phagocytic cells (including macrophages and osteoclasts); and targeted depletion of CD169(+) macrophages using a suicide gene knock-in model. Immunohistochemistry and histomorphometric analysis were performed to quantitatively assess cancer-induced bone changes. In human bone metastasis specimens, CD68(+) macrophages were consistently located within the tumour mass. Osteal macrophages (osteomacs) were associated with pathological woven bone within the metastatic lesions. In contrast, lymphocytes were inconsistently present in prostate cancer skeletal lesions and when detected, had varied distributions. In the immune-competent mouse model, CD169(+) macrophage ablation significantly inhibited prostate cancer-induced woven bone formation, suggesting that CD169(+) macrophages within pathological woven bone are integral to tumour-induced bone formation. In contrast, pan-phagocytic cell, but not targeted CD169(+) macrophage depletion resulted in increased tumour mass, indicating that CD169(-) macrophage subset(s) and/or osteoclasts influenced tumour growth. In summary, these observations indicate a prominent role for macrophages in prostate cancer bone metastasis that may be therapeutically targetable to reduce the negative skeletal impacts of this malignancy, including tumour-induced bone modelling. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Andy C Wu
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Yaowu He
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Amy Broomfield
- Department of Anatomical Pathology, Mater Misericordiae Ltd., South Brisbane, Australia
| | - Nicoll J Paatan
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, Australia
| | - Brittney S Harrington
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Hsu-Wen Tseng
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Elizabeth A Beaven
- Department of Anatomical Pathology, Mater Misericordiae Ltd., South Brisbane, Australia
| | - Deirdre M Kiernan
- Department of Urology, Mater Health Services, South Brisbane, Australia
| | - Peter Swindle
- Department of Urology, Mater Health Services, South Brisbane, Australia
| | - Adrian B Clubb
- Department of Urology, Mater Health Services, South Brisbane, Australia
| | - Jean-Pierre Levesque
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Ingrid G Winkler
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Ming-Tat Ling
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, Australia.,Institute for Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolloongabba, Australia
| | - Bhuvana Srinivasan
- Department of Anatomical Pathology, Mater Misericordiae Ltd., South Brisbane, Australia
| | - John D Hooper
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Allison R Pettit
- Faculty of Medicine and Biomedical Sciences, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| |
Collapse
|
42
|
Meshcheryakova A, Svoboda M, Tahir A, Köfeler HC, Triebl A, Mungenast F, Heinze G, Gerner C, Zimmermann P, Jaritz M, Mechtcheriakova D. Exploring the role of sphingolipid machinery during the epithelial to mesenchymal transition program using an integrative approach. Oncotarget 2016; 7:22295-323. [PMID: 26967245 PMCID: PMC5008362 DOI: 10.18632/oncotarget.7947] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/20/2016] [Indexed: 12/30/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) program is activated in epithelial cancer cells and facilitates their ability to metastasize based on enhanced migratory, proliferative, anti-apoptotic, and pluripotent capacities. Given the fundamental impact of sphingolipid machinery to each individual process, the sphingolipid-related mechanisms might be considered among the most prominent drivers/players of EMT; yet, there is still limited knowledge. Given the complexity of the interconnected sphingolipid system, which includes distinct sphingolipid mediators, their synthesizing enzymes, receptors and transporters, we herein apply an integrative approach for assessment of the sphingolipid-associated mechanisms underlying EMT program. We created the sphingolipid-/EMT-relevant 41-gene/23-gene signatures which were applied to denote transcriptional events in a lung cancer cell-based EMT model. Based on defined 35-gene sphingolipid/EMT-attributed signature of regulated genes, we show close associations between EMT markers, genes comprising the sphingolipid network at multiple levels and encoding sphingosine 1-phosphate (S1P)-/ceramide-metabolizing enzymes, S1P and lysophosphatidic acid (LPA) receptors and S1P transporters, pluripotency genes and inflammation-related molecules, and demonstrate the underlying biological pathways and regulators. Mass spectrometry-based sphingolipid analysis revealed an EMT-attributed shift towards increased S1P and LPA accompanied by reduced ceramide levels. Notably, using transcriptomics data across various cell-based perturbations and neoplastic tissues (24193 arrays), we identified the sphingolipid/EMT signature primarily in lung adenocarcinoma tissues; besides, bladder, colorectal and prostate cancers were among the top-ranked. The findings also highlight novel regulatory associations between influenza virus and the sphingolipid/EMT-associated mechanisms. In sum, data propose the multidimensional contribution of sphingolipid machinery to pathological EMT and may yield new biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Anastasia Meshcheryakova
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Martin Svoboda
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Ammar Tahir
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria
- Mass Spectrometry Center, University of Vienna, Vienna, Austria
| | - Harald C. Köfeler
- Core Facility for Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Alexander Triebl
- Core Facility for Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Felicitas Mungenast
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Georg Heinze
- Section for Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University Vienna, Vienna, Austria
| | - Christopher Gerner
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria
- Mass Spectrometry Center, University of Vienna, Vienna, Austria
| | | | - Markus Jaritz
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Diana Mechtcheriakova
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
Huang Y, Cheng C, Zhang C, Zhang Y, Chen M, Strand DW, Jiang M. Advances in prostate cancer research models: From transgenic mice to tumor xenografting models. Asian J Urol 2016; 3:64-74. [PMID: 29264167 PMCID: PMC5730804 DOI: 10.1016/j.ajur.2016.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/01/2016] [Accepted: 02/02/2016] [Indexed: 12/17/2022] Open
Abstract
The identification of the origin and molecular characteristics of prostate cancer (PCa) has crucial implications for personalized treatment. The development of effective treatments for PCa has been limited; however, the recent establishment of several transgenic mouse lines and/or xenografting models is better reflecting the disease in vivo. With appropriate models, valuable tools for elucidating the functions of specific genes have gone deep into prostate development and carcinogenesis. In the present review, we summarize a number of important PCa research models established in our laboratories (PSA-Cre-ERT2/PTEN transgenic mouse models, AP-OX model, tissue recombination-xenografting models and PDX models), which represent advances of translational models from transgenic mouse lines to human tumor xenografting. Better understanding of the developments of these models will offer new insights into tumor progression and may help explain the functional significance of genetic variations in PCa. Additionally, this understanding could lead to new modes for curing PCa based on their particular biological phenotypes.
Collapse
Affiliation(s)
- Yuejiao Huang
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, China
| | - Chun Cheng
- Department of Immunology, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Chong Zhang
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Yonghui Zhang
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Miaomiao Chen
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Douglas W Strand
- Department of Urology, UT Southernwestern Medical Center, Dallas, TX, USA
| | - Ming Jiang
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China.,Institute of Medicine and Public Health, Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
44
|
Spheroid culture of LuCaP 136 patient-derived xenograft enables versatile preclinical models of prostate cancer. Clin Exp Metastasis 2016; 33:325-37. [PMID: 26873136 DOI: 10.1007/s10585-016-9781-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/10/2016] [Indexed: 12/22/2022]
Abstract
LuCaP serially transplantable patient-derived xenografts (PDXs) are valuable preclinical models of locally advanced or metastatic prostate cancer. Using spheroid culture methodology, we recently established cell lines from several LuCaP PDXs. Here, we characterized in depth the features of xenografts derived from LuCaP 136 spheroid cultures and found faithful retention of the phenotype of the original PDX. In vitro culture enabled luciferase transfection into LuCaP 136 spheroids, facilitating in vivo imaging. We showed that LuCaP 136 spheroids formed intratibial, orthotopic, and subcutaneous tumors when re-introduced into mice. Intratibial tumors responded to castration and were highly osteosclerotic. LuCaP 136 is a realistic in vitro-in vivo preclinical model of a subtype of bone metastatic prostate cancer.
Collapse
|
45
|
Hussain M, Le Moulec S, Gimmi C, Bruns R, Straub J, Miller K. Differential Effect on Bone Lesions of Targeting Integrins: Randomized Phase II Trial of Abituzumab in Patients with Metastatic Castration-Resistant Prostate Cancer. Clin Cancer Res 2016; 22:3192-200. [PMID: 26839144 DOI: 10.1158/1078-0432.ccr-15-2512] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/05/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Integrins play a critical role in the progression of prostate cancer and its bone metastases. We investigated the use of the pan-αv integrin inhibitor abituzumab in chemotherapy-naïve patients with asymptomatic or mildly symptomatic metastatic castration-resistant prostate cancer. EXPERIMENTAL DESIGN PERSEUS (NCT01360840) was a randomized, double-blind phase II study. Men with pathologically confirmed prostate cancer and radiologic progression of bone lesions in the 28 days prior to randomization were assigned to receive abituzumab 750 mg or 1,500 mg or placebo (1:1:1) every 3 weeks in combination with luteinizing hormone-releasing hormone agonist/antagonist therapy. The primary endpoint was progression-free survival (PFS). RESULTS The intent-to-treat population comprised 180 patients, 60 in each arm. The primary endpoint of PFS was not significantly different with abituzumab-based therapy compared with placebo [abituzumab 750 mg, 3.4 months, HR = 0.89; 95% confidence interval (CI), 0.57-1.39; abituzumab 1,500 mg, 4.3 months, HR = 0.81; 95% CI, 0.52-1.26; placebo, 3.3 months], but the cumulative incidence of bone lesion progression was lower with abituzumab than with placebo for up to 24 months (cumulative incidence 23.6% vs. 41.1% at 6 months, 26.1% vs. 45.4% at 12 months). Two partial tumor responses were observed (1 abituzumab 1,500 mg and 1 placebo). Approximately 85% to 90% of patients experienced at least one treatment-emergent adverse event (TEAE) in the different arms, but the incidences of serious TEAEs and TEAEs with fatal outcome were similar in the three arms. CONCLUSIONS Although PFS was not significantly extended, abituzumab appears to have specific activity in prostate cancer-associated bone lesions that warrants further investigation. Clin Cancer Res; 22(13); 3192-200. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | - Kurt Miller
- Department of Urology, Charité, Berlin, Germany
| | | |
Collapse
|
46
|
Abstract
In the context of breast cancer, the importance of the skeleton in the regulation of primary tumour development and as a site for subsequent metastasis is well characterized. Our understanding of the contributions made by the host bone and bone marrow cells increasingly demonstrates the extent of the interaction between tumour cells and normal host cells. As a result, the need to develop and utilize therapies that can impede the growth and/or function of tumour cells while sparing normal host bone and bone marrow cells is immense and expanding. The need for these new treatments is, however, superimposed on the orthopaedic management of patients' quality of life, where pain control and continued locomotion are paramount. Indeed, the majority of the anticancer therapies used to date often result in direct or indirect damage to bone. Thus, although the bone microenvironment regulates tumour cell growth in bone, cells within the bone marrow niche also mediate many of the orthopaedic consequences of tumour progression as well as resistance to the antitumour effects of existing therapies. In this Review, we highlight the effects of existing cancer treatments on bone and the bone marrow microenvironment as well as the mechanisms mediating these effects and the current utility of modern orthopaedic interventions.
Collapse
Affiliation(s)
- Issam Makhoul
- Department of Medicine, Division of Haematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Corey O Montgomery
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Dana Gaddy
- Department of Physiology and Biophysics, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Larry J Suva
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| |
Collapse
|
47
|
Moreno-Sánchez R, Saavedra E, Gallardo-Pérez JC, Rumjanek FD, Rodríguez-Enríquez S. Understanding the cancer cell phenotype beyond the limitations of current omics analyses. FEBS J 2015; 283:54-73. [DOI: 10.1111/febs.13535] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/24/2015] [Accepted: 09/25/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Rafael Moreno-Sánchez
- Departamento de Bioquímica; Instituto Nacional de Cardiología Ignacio Chávez; Tlalpan Mexico
| | - Emma Saavedra
- Departamento de Bioquímica; Instituto Nacional de Cardiología Ignacio Chávez; Tlalpan Mexico
| | | | | | - Sara Rodríguez-Enríquez
- Departamento de Bioquímica; Instituto Nacional de Cardiología Ignacio Chávez; Tlalpan Mexico
| |
Collapse
|
48
|
Xie JJ, Guo JC, Wu ZY, Xu XE, Wu JY, Chen B, Ran LQ, Liao LD, Li EM, Xu LY. Integrin α5 promotes tumor progression and is an independent unfavorable prognostic factor in esophageal squamous cell carcinoma. Hum Pathol 2015; 48:69-75. [PMID: 26772401 DOI: 10.1016/j.humpath.2015.09.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
The integrin family plays a major role in complex biological events such as differentiation, development, wound healing, and the altered adhesive and invasive properties of tumor cells. The expression and function of integrin α5 in esophageal squamous cell carcinoma (ESCC) are not clear. Here, by using tissue microarrays and immunohistochemical method, integrin α5 expression was retrospectively evaluated in 147 samples of human ESCC. Results showed that expression of integrin α5 was heterogeneous and varied from negative to intense expression in a membrane and cytoplasmic distribution manner. High expression of integrin α5 was significantly correlated with lymph node metastasis (P = .042) and tumor size (P = .042). Kaplan-Meier analysis revealed that high expression of integrin α5 was related to poor overall survival of ESCC patients (P = .018). Multivariate analysis suggested that integrin α5 expression status was an independent prognostic factor for ESCC (P = .003). Moreover, integrin α5 expression was associated with the survival of patients with lymph node metastasis (P = .020), but did not influence the survival of patients without lymph node metastasis. Finally, we found that RNAi-mediated knockdown of integrin α5 led to decreased growth, migration, and invasion of ESCC cells. Combined, integrin α5 might play important roles in the progression of ESCC. Integrin α5 is a novel biomarker to predict the prognosis of ESCC patients.
Collapse
Affiliation(s)
- Jian-Jun Xie
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Medical College of Shantou University, Shantou 515041, PR China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 515041, PR China.
| | - Jin-Cheng Guo
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Medical College of Shantou University, Shantou 515041, PR China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 515041, PR China
| | - Zhi-Yong Wu
- Department of Oncologic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou 515041, PR China
| | - Xiu-E Xu
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou 515041, PR China
| | - Jian-Yi Wu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Medical College of Shantou University, Shantou 515041, PR China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 515041, PR China
| | - Bo Chen
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou 515041, PR China
| | - Li-Qiang Ran
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Medical College of Shantou University, Shantou 515041, PR China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 515041, PR China
| | - Lian-Di Liao
- Institute of Oncologic Pathology, Medical College of Shantou University, Shantou 515041, PR China
| | - En-Min Li
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Medical College of Shantou University, Shantou 515041, PR China; Department of Biochemistry and Molecular Biology, Medical College of Shantou University, Shantou 515041, PR China
| | - Li-Yan Xu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Medical College of Shantou University, Shantou 515041, PR China; Institute of Oncologic Pathology, Medical College of Shantou University, Shantou 515041, PR China.
| |
Collapse
|
49
|
Cancer as a Proinflammatory Environment: Metastasis and Cachexia. Mediators Inflamm 2015; 2015:791060. [PMID: 26508818 PMCID: PMC4609868 DOI: 10.1155/2015/791060] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/17/2015] [Accepted: 05/11/2015] [Indexed: 01/08/2023] Open
Abstract
The development of the syndrome of cancer cachexia and that of metastasis are related with a poor prognostic for cancer patients. They are considered multifactorial processes associated with a proinflammatory environment, to which tumour microenvironment and other tissues from the tumour bearing individuals contribute. The aim of the present review is to address the role of ghrelin, myostatin, leptin, HIF, IL-6, TNF-α, and ANGPTL-4 in the regulation of energy balance, tumour development, and tumoural cell invasion. Hypoxia induced factor plays a prominent role in tumour macro- and microenvironment, by modulating the release of proinflammatory cytokines.
Collapse
|
50
|
Li J, Xiang S, Zhang Q, Wu J, Tang Q, Zhou J, Yang L, Chen Z, Hann SS. Combination of curcumin and bicalutamide enhanced the growth inhibition of androgen-independent prostate cancer cells through SAPK/JNK and MEK/ERK1/2-mediated targeting NF-κB/p65 and MUC1-C. J Exp Clin Cancer Res 2015; 34:46. [PMID: 25971429 PMCID: PMC4446835 DOI: 10.1186/s13046-015-0168-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/08/2015] [Indexed: 11/17/2022] Open
Abstract
Background Prostate cancer is one of the most common malignancies in men. The mucin 1 (MUC1) heterodimeric oncoprotein is overexpressed in human prostate cancers with aggressive pathologic and clinical features, resulting in a poor outcome. However, the functional role for MUC1 C-terminal domain (MUC1-C) in androgen-independent prostate cancer occurrence and development has remained unclear. Methods Cell viability was measured by MTT assays. Western blot analysis was performed to measure the phosphorylation and protein expression of SAPK/JNK and ERK1/2, and MUC1-C, NF-κB subunit p65 and p50. Exogenous expression of MUC1-C, NF-κB subunit p65 was carried out by transient and electroporated transfection assays. Results We showed that curcumin inhibited the growth of androgen-independent prostate cancer cells and a synergy was observed in the presence of curcumin and bicalutamide, the androgen receptor antagonist. To further explore the potential mechanism underlining this, we found that curcumin increased the phosphorylation of ERK1/2 and SAPK/JNK, which was enhanced by bicalutamide. In addition, curcumin reduced the protein expression of MUC1-C and NF-κB subunit p65, which were abrogated in the presence of the inhibitors of MEK/ERK1/2 (PD98059) and SAPK/JNK (SP60015). A further reduction was observed in the combination of curcumin with bicalutamide. Moreover, while exogenous expression of MUC1-C had little effect on curcumin-reduced p65, the overexpression of p65 reversed the effect of curcumin on MUC1-C protein expression suggesting that p65 is upstream of MUC1-C. Intriguingly, we showed that exogenous expression of MUC1-C feedback diminished the effect of curcumin on phosphorylation of ERK1/2 and SAPK/JNK, and antagonized the effect of curcumin on cell growth. Conclusion Our results show that curcumin inhibits the growth of androgen-independent prostate cancer cells through ERK1/2- and SAPK/JNK-mediated inhibition of p65, followed by reducing expression of MUC1-C protein. More importantly, there are synergistic effects of curcumin and bicalutamide. The negative feedback regulatory loop of MUC1-C to ERK1/2 and SAPK/JNK further demonstrates the role of MUC1-C that contributes to the overall responses of curcumin. This study unveils the potential molecular mechanism by which combination of curcumin with bicalutamide enhances the growth inhibition of androgen-independent prostate cancer cells.
Collapse
Affiliation(s)
- Jing Li
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China. .,Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - SongTao Xiang
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - QiouHong Zhang
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - JingJing Wu
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Qing Tang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - JianFu Zhou
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - LiJun Yang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - ZhiQiang Chen
- Department of Urology Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Swei Sunny Hann
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China. .,Higher Education Mega Center, Panyu District, Guangdong Provincial Hospital of Chinese Medicine, No. 55, Neihuan West Road, Guangzhou, Guangdong Province, 510006, People's Republic of China.
| |
Collapse
|