1
|
Sailer V, von Amsberg G, Duensing S, Kirfel J, Lieb V, Metzger E, Offermann A, Pantel K, Schuele R, Taubert H, Wach S, Perner S, Werner S, Aigner A. Experimental in vitro, ex vivo and in vivo models in prostate cancer research. Nat Rev Urol 2023; 20:158-178. [PMID: 36451039 DOI: 10.1038/s41585-022-00677-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/02/2022]
Abstract
Androgen deprivation therapy has a central role in the treatment of advanced prostate cancer, often causing initial tumour remission before increasing independence from signal transduction mechanisms of the androgen receptor and then eventual disease progression. Novel treatment approaches are urgently needed, but only a fraction of promising drug candidates from the laboratory will eventually reach clinical approval, highlighting the demand for critical assessment of current preclinical models. Such models include standard, genetically modified and patient-derived cell lines, spheroid and organoid culture models, scaffold and hydrogel cultures, tissue slices, tumour xenograft models, patient-derived xenograft and circulating tumour cell eXplant models as well as transgenic and knockout mouse models. These models need to account for inter-patient and intra-patient heterogeneity, the acquisition of primary or secondary resistance, the interaction of tumour cells with their microenvironment, which make crucial contributions to tumour progression and resistance, as well as the effects of the 3D tissue network on drug penetration, bioavailability and efficacy.
Collapse
Affiliation(s)
- Verena Sailer
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Gunhild von Amsberg
- Department of Oncology and Hematology, University Cancer Center Hamburg Eppendorf and Martini-Klinik, Prostate Cancer Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Stefan Duensing
- Section of Molecular Urooncology, Department of Urology, University Hospital Heidelberg and National Center for Tumour Diseases, Heidelberg, Germany
| | - Jutta Kirfel
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Verena Lieb
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Eric Metzger
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Anne Offermann
- Institute for Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Klaus Pantel
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Roland Schuele
- Department of Urology, Center for Clinical Research, University of Freiburg Medical Center, Freiburg, Germany
| | - Helge Taubert
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Wach
- Research Division Molecular Urology, Department of Urology and Paediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Sven Perner
- University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Stefan Werner
- Institute for Tumour Biology, Center for Experimental Medicine, University Clinics Hamburg-Eppendorf, Hamburg, Germany
- Mildred-Scheel-Nachwuchszentrum HaTRiCs4, University Cancer Center Hamburg, Hamburg, Germany
| | - Achim Aigner
- Clinical Pharmacology, Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Medical Faculty, Leipzig, Germany.
| |
Collapse
|
2
|
Preclinical models of prostate cancer - modelling androgen dependency and castration resistance in vitro, ex vivo and in vivo. Nat Rev Urol 2023:10.1038/s41585-023-00726-1. [PMID: 36788359 DOI: 10.1038/s41585-023-00726-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 02/16/2023]
Abstract
Prostate cancer is well known to be dependent on the androgen receptor (AR) for growth and survival. Thus, AR is the main pharmacological target to treat this disease. However, after an initially positive response to AR-targeting therapies, prostate cancer will eventually evolve to castration-resistant prostate cancer, which is often lethal. Tumour growth was initially thought to become androgen-independent following treatments; however, results from molecular studies have shown that most resistance mechanisms involve the reactivation of AR. Consequently, tumour cells become resistant to castration - the blockade of testicular androgens - and not independent of AR per se. However, confusion still remains on how to properly define preclinical models of prostate cancer, including cell lines. Most cell lines were isolated from patients for cell culture after evolution of the tumour to castration-resistant prostate cancer, but not all of these cell lines are described as castration resistant. Moreover, castration refers to the blockade of testosterone production by the testes; thus, even the concept of "castration" in vitro is questionable. To ensure maximal transfer of knowledge from scientific research to the clinic, understanding the limitations and advantages of preclinical models, as well as how these models recapitulate cancer cell androgen dependency and can be used to study castration resistance mechanisms, is essential.
Collapse
|
3
|
Mukherjee A, Park A, Davies KP. PROL1 is essential for xenograft tumor development in mice injected with the human prostate cancer cell-line, LNCaP, and modulates cell migration and invasion. JOURNAL OF MEN'S HEALTH 2022; 18:044. [PMID: 35547856 PMCID: PMC9089447 DOI: 10.31083/jomh.2021.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background and objective A growing body of literature suggests modulated expression of members of the opiorphin family of genes (PROL1, SMR3A and SMR3B) is associated with cancer. Recently, overexpression of PROL1 was shown to be associated with prostate cancer, with evidence of a role in overcoming the hypoxic barrier that develops as tumors grow. The primary goal of the present studies was to support and expand evidence for a role of PROL1 in the development and progression of prostate cancer. Material and methods We engineered knock-out of the opiorphin gene, PROL1, in LNCaP, an androgen-sensitive, human prostate cancer derived, cell-line. Using xenograft assays, we compared the ability of injected LNCaP PROL1 knock-out cell-lines to develop tumors in both castrated and intact male mice with the parental LNCaP and LNCaP PROL1 overexpressing cell-lines. We used RNAseq to compare global gene expression between the parental and LNCaP PROL1 knock-out cell-lines. Wound closure and 3D spheroid invasion assays were used to compare cell motility and migration between parental LNCaP cells and LNCaP cells overexpressing of PROL1. Results The present studies demonstrate that LNCaP cell-lines with consisitutive knock-out of PROL1 fail to develop tumors when injected into both castrated and intact male mice. Using RNAseq to compare global gene expression between the parental and LNCaP PROL1 knock-out cell-lines, we confirmed a role for PROL1 in regulating molecular pathways associated with angiogenesis and tumor blood supply, and also identified a potential role in pathways related to cell motility and migration. Through the use of wound closure and 3D spheroid invasion assays, we confirmed that overexpression of PROL1 in LNCaP cells leads to greater cell motility and migration compared to parental cells, suggesting that PROL1 overexpression results in a more invasive phenotype. Conclusion Overall, our studies add to the growing body of evidence that opiorphin-encoding genes play a role in cancer development and progression. PROL1 is essential for establishment and growth of tumors in mice injected with LNCaP cells, and we provide evidence that PROL1 has a possible role in progression towards a more invasive, metastatic and castration resistant prostate cancer (PrCa).
Collapse
Affiliation(s)
- Amarnath Mukherjee
- Department of Urology, Albert Einstein College of Medicine,
Bronx, NY 10461, USA
| | - Augene Park
- Department of Urology, Albert Einstein College of Medicine,
Bronx, NY 10461, USA
| | - Kelvin Paul Davies
- Department of Urology, Albert Einstein College of Medicine,
Bronx, NY 10461, USA
- Department of Physiology and Biophysics, Albert Einstein
College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
4
|
Ban J, Fock V, Aryee DNT, Kovar H. Mechanisms, Diagnosis and Treatment of Bone Metastases. Cells 2021; 10:2944. [PMID: 34831167 PMCID: PMC8616226 DOI: 10.3390/cells10112944] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Bone and bone marrow are among the most frequent metastatic sites of cancer. The occurrence of bone metastasis is frequently associated with a dismal disease outcome. The prevention and therapy of bone metastases is a priority in the treatment of cancer patients. However, current therapeutic options for patients with bone metastatic disease are limited in efficacy and associated with increased morbidity. Therefore, most current therapies are mainly palliative in nature. A better understanding of the underlying molecular pathways of the bone metastatic process is warranted to develop novel, well-tolerated and more successful treatments for a significant improvement of patients' quality of life and disease outcome. In this review, we provide comparative mechanistic insights into the bone metastatic process of various solid tumors, including pediatric cancers. We also highlight current and innovative approaches to biologically targeted therapy and immunotherapy. In particular, we discuss the role of the bone marrow microenvironment in the attraction, homing, dormancy and outgrowth of metastatic tumor cells and the ensuing therapeutic implications. Multiple signaling pathways have been described to contribute to metastatic spread to the bone of specific cancer entities, with most knowledge derived from the study of breast and prostate cancer. However, it is likely that similar mechanisms are involved in different types of cancer, including multiple myeloma, primary bone sarcomas and neuroblastoma. The metastatic rate-limiting interaction of tumor cells with the various cellular and noncellular components of the bone-marrow niche provides attractive therapeutic targets, which are already partially exploited by novel promising immunotherapies.
Collapse
Affiliation(s)
- Jozef Ban
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Valerie Fock
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Dave N. T. Aryee
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| | - Heinrich Kovar
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
5
|
Nascimento-Gonçalves E, Seixas F, Ferreira R, Colaço B, Parada B, Oliveira PA. An overview of the latest in state-of-the-art murine models for prostate cancer. Expert Opin Drug Discov 2021; 16:1349-1364. [PMID: 34224283 DOI: 10.1080/17460441.2021.1943354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Prostate cancer (PCa) is a complex, heterogenous and multifocal disease, which is debilitating for patients and often fatal - due to bone metastasis and castration-resistant cancer. The use of murine models that mimic human disease has been crucial in the development of innovative therapies and for better understanding the mechanisms associated with initiation and progression of PCa. AREAS COVERED This review presents a critical analysis of murine models for the study of PCa, highlighting their strengths, weaknesses and applications. EXPERT OPINION In animal models, disease may not occur exactly as it does in humans, and sometimes the levels of efficacy that certain treatments obtain in animal models cannot be translated into clinical practice. To choose the most appropriate animal model for each research work, it is crucial to understand the anatomical and physiological differences between the mouse and the human prostate, while it is also important to identify biological similarities and differences between murine and human prostate tumors. Although significant progress has already been made, thanks to many years of research and study, the number of new challenges and obstacles to overcome mean there is a long and difficult road still to travel.
Collapse
Affiliation(s)
- Elisabete Nascimento-Gonçalves
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal.,Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, UTAD, Vila Real, Portugal.,Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (Laqv-requimte),department of Chemistry, University of Aveiro (UA), Portugal
| | - Fernanda Seixas
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal.,Animal and Veterinary Research Centre (CECAV), UTAD, Vila Real, Portugal
| | - Rita Ferreira
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (Laqv-requimte),department of Chemistry, University of Aveiro (UA), Portugal
| | - Bruno Colaço
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, UTAD, Vila Real, Portugal.,Department of Zootechnics, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Belmiro Parada
- Faculty of Medicine, University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (Icbr), Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,Urology and Renal Transplantation Department, Coimbra University Hospital Centre (CHUC), Coimbra, Portugal
| | - Paula A Oliveira
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal.,Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, UTAD, Vila Real, Portugal
| |
Collapse
|
6
|
Aubert N, Brunel S, Olive D, Marodon G. Blockade of HVEM for Prostate Cancer Immunotherapy in Humanized Mice. Cancers (Basel) 2021; 13:cancers13123009. [PMID: 34208480 PMCID: PMC8235544 DOI: 10.3390/cancers13123009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Current immune checkpoint inhibitors have shown limitations for immunotherapy of prostate cancer. Thus, it is crucial to investigate other immune checkpoints to prevent disease progression in patients with prostate cancer. Here, we first show that the HVEM/BTLA immune checkpoint is associated with disease progression in patients. We then show that immunotherapy aimed at targeting HVEM reduced tumor growth twofold in vivo in a humanized mouse model of the pathology. The mode of action of the therapy was dependent on CD8+ T cells and is associated with improved T cell activation and reduced exhaustion. Finally, we demonstrated that HVEM expressed by the tumor negatively regulated the anti-tumor immune response. Our results indicate that targeting HVEM might be an attractive option for patients with prostate cancer. Abstract The herpes virus entry mediator (HVEM) delivers a negative signal to T cells mainly through the B and T lymphocyte attenuator (BTLA) molecule. Thus, HVEM/BTLA may represent a novel immune checkpoint during an anti-tumor immune response. However, a formal demonstration that HVEM can represent a target for cancer immunotherapy is still lacking. Here, we first showed that HVEM and BTLA mRNA expression levels were associated with a worse progression-free interval in patients with prostate adenocarcinomas, indicating a detrimental role for the HVEM/BTLA immune checkpoint during prostate cancer progression. We then showed that administration of a monoclonal antibody to human HVEM resulted in a twofold reduction in the growth of a prostate cancer cell line in NOD.SCID.gc-null mice reconstituted with human T cells. Using CRISPR/Cas9, we showed that the therapeutic effect of the mAb depended on HVEM expression by the tumor, with no effect on graft vs. host disease or activation of human T cells in the spleen. In contrast, the proliferation and number of tumor-infiltrating leukocytes increased following treatment, and depletion of CD8+ T cells partly alleviated treatment’s efficacy. The expression of genes belonging to various T cell activation pathways was enriched in tumor-infiltrating leukocytes, whereas genes associated with immuno-suppressive pathways were decreased, possibly resulting in modifications of leukocyte adhesion and motility. Finally, we developed a simple in vivo assay in humanized mice to directly demonstrate that HVEM expressed by the tumor is an immune checkpoint for T cell-mediated tumor control. Our results show that targeting HVEM is a promising strategy for prostate cancer immunotherapy.
Collapse
Affiliation(s)
- Nicolas Aubert
- Centre d’Immunologie et Maladies Infectieuses-Paris, CIMI-PARIS, Sorbonne Université, INSERM, CNRS, 75013 Paris, France; (N.A.); (S.B.)
| | - Simon Brunel
- Centre d’Immunologie et Maladies Infectieuses-Paris, CIMI-PARIS, Sorbonne Université, INSERM, CNRS, 75013 Paris, France; (N.A.); (S.B.)
| | - Daniel Olive
- Institut Paoli-Calmettes, Aix-Marseille Université, INSERM, CNRS, CRCM, Tumor Immunity Team, IBISA Immunomonitoring Platform, 13009 Marseille, France;
| | - Gilles Marodon
- Centre d’Immunologie et Maladies Infectieuses-Paris, CIMI-PARIS, Sorbonne Université, INSERM, CNRS, 75013 Paris, France; (N.A.); (S.B.)
- Correspondence:
| |
Collapse
|
7
|
Liu S, Zhang B, Rowan BG, Jazwinski SM, Abdel-Mageed AB, Steele C, Wang AR, Sartor O, Niu T, Zhang Q. A Novel Controlled PTEN-Knockout Mouse Model for Prostate Cancer Study. Front Mol Biosci 2021; 8:696537. [PMID: 34150854 PMCID: PMC8211560 DOI: 10.3389/fmolb.2021.696537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is associated with advanced age, but how age contributes to prostate carcinogenesis remains unknown. The prostate-specific Pten conditional knockout mouse model closely imitates human PCa initiation and progression. To better understand how age impacts PCa in an experimental model, we have generated a spatially and temporally controlled Pten-null PCa murine model at different ages (aged vs. non-aged) of adult mice. Here, we present a protocol to inject the Cre-expressing adenovirus with luciferin tag, intraductally, into the prostate anterior lobes of Pten-floxed mice; Pten-loss will be triggered post-Cre expression at different ages. In vivo imaging of luciferin signal following viral infection confirmed successful delivery of the virus and Cre activity. Immunohistochemical staining confirmed prostate epithelial-specific expression of Cre recombinase and the loss of Pten and activation of P-Akt, P-S6, and P-4E-BP1. The Cre-expression, Pten ablation, and activated PI3K/AKT/mTOR pathways were limited to the prostate epithelium. All mice developed prostatic epithelial hyperplasia within 4 weeks after Pten ablation and prostatic intraepithelial neoplasia (PIN) within 8 weeks post-Pten ablation. Some PINs had progressed to invasive adenocarcinoma at 8-16 weeks post-Pten ablation. Aged mice exhibited significantly accelerated PI3K/AKT/mTOR signaling and increased PCa onset and progression compared to young mice. The viral infection success rate is ∼80%. This model will be beneficial for investigations of cancer-related to aging.
Collapse
Affiliation(s)
- Sen Liu
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Bing Zhang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
- Medical Laboratory of ShenZhen LuoHu People’s Hospital, Shenzhen, China
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - S. Michal Jazwinski
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, United States
| | - Asim B. Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Chad Steele
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Alun R. Wang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Oliver Sartor
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, United States
| | - Tianhua Niu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Qiuyang Zhang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
8
|
Arrighetti N, Beretta GL. miRNAs as Therapeutic Tools and Biomarkers for Prostate Cancer. Pharmaceutics 2021; 13:380. [PMID: 33805590 PMCID: PMC7999286 DOI: 10.3390/pharmaceutics13030380] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PCa) is the fifth cause of tumor-related deaths in man worldwide. Despite the considerable improvement in the clinical management of PCa, several limitations emerged both in the screening for early diagnosis and in the medical treatment. The use of prostate-specific antigen (PSA)-based screening resulted in patients' overtreatment and the standard therapy of patients suffering from locally advanced/metastatic tumors (e.g., radical prostatectomy, radiotherapy, and androgen deprivation therapy) showed time-limited efficacy with patients undergoing progression toward the lethal metastatic castration-resistant PCa (mCRPC). Although valuable alternative therapeutic options have been recently proposed (e.g., docetaxel, cabazitaxel, abiraterone, enzalutamide, and sipuleucel-T), mCRPC remains incurable. Based on this background, there is an urgent need to identify new and more accurate prostate-specific biomarkers for PCa diagnosis and prognosis and to develop innovative medical approaches to counteract mCRPC. In this context, microRNA (miRNAs) emerged as potential biomarkers in prostate tissues and biological fluids and appeared to be promising therapeutic targets/tools for cancer therapy. Here we overview the recent literature and summarize the achievements of using miRNAs as biomarkers and therapeutic targets/tools for fighting PCa.
Collapse
Affiliation(s)
| | - Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| |
Collapse
|
9
|
Straign DM, Ihle CL, Provera MD, Owens P. Targeting the BMP Pathway in Prostate Cancer Induced Bone Disease. Front Endocrinol (Lausanne) 2021; 12:769316. [PMID: 34956082 PMCID: PMC8702552 DOI: 10.3389/fendo.2021.769316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
From the 33,000 men in the U.S. who die from prostate cancer each year, the majority of these patients exhibit metastatic disease with bone being the most common site of metastasis. Prostate cancer bone metastases are commonly blastic, exhibiting new growth of unhealthy sclerotic bone, which can cause painful skeletal related events. Patient's current care entails androgen deprivation therapy, anti-resorptive agents, radiation, and chemotherapy to help control the spread of the cancer but little intervention is available to treat blastic bone disease. The transforming growth factor beta (TGFβ) and bone morphogenetic protein (BMP) pathways are known to regulate bone growth and resorption of destructive lytic bone lesions, yet the role of TGFβ/BMP signaling in prostate cancer blastic vs lytic bone lesions are not fully understood. We hypothesized that to target the BMP/TGFβ pathway, a useful biomarker of bone lytic or blastic pathology would have superior response. We show distinct BMP vs. TGFβ signaling in clinical samples of human prostate cancer bone metastases with either lytic or blastic pathologies. BMPs exhibit distinct effects on bone homeostasis, so to examine the effect of BMP inhibition on healthy bone, we treated mice with the BMP receptor small molecule antagonist DMH1 and saw a modest temporary improvement in bone health, with increased trabecular bone. We next sought to use the BMP inhibitor DMH1 to treat bone metastasis engraftment seeded by a caudal artery injection of the lytic human prostate cell line PC3 in immunodeficient mice. The colonization by PC3 cells to the bone were restricted with DMH1 treatment and bone health was importantly preserved. We next proceeded to test BMP inhibition in an injury model of established bone metastasis via intratibial injection of the MYC-CaP mouse prostate cell line into FVBN syngeneic mice. DMH1 treated mice had a modest decrease in trabecular bone and reduced lymphocytes in circulation without affecting tumor growth. Taken together we show unique responses to BMP inhibition in metastatic prostate cancer in the bone. These studies suggest that profiling bone lesions in metastatic prostate cancer can help identify therapeutic targets that not only treat the metastatic tumor but also address the need to better treat the distinct tumor induced bone disease.
Collapse
Affiliation(s)
- Desiree M. Straign
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Claire L. Ihle
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Cancer Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Meredith D. Provera
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Philip Owens
- Cancer Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Veterans Affairs, Research Service, Eastern Colorado Health Care System, Aurora, CO, United States
- *Correspondence: Philip Owens,
| |
Collapse
|
10
|
Xenograft-derived mRNA/miR and protein interaction networks of systemic dissemination in human prostate cancer. Eur J Cancer 2020; 137:93-107. [PMID: 32750503 DOI: 10.1016/j.ejca.2020.06.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/12/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Distant metastasis formation is the major clinical problem in prostate cancer (PCa) and the underlying mechanisms remain poorly understood. Our aim was to identify novel molecules that functionally contribute to human PCa systemic dissemination based on unbiased approaches. METHODS We compared mRNA, microRNA (miR) and protein expression levels in established human PCa xenograft tumours with high (PC-3), moderate (VCaP) or weak (DU-145) spontaneous micrometastatic potential. By focussing on those mRNAs, miRs and proteins that were differentially regulated among the xenograft groups and known to interact with each other we constructed dissemination-related mRNA/miR and protein/miR networks. Next, we clinically and functionally validated our findings. RESULTS Besides known determinants of PCa progression and/or metastasis, our interaction networks include several novel candidates. We observed a clear role of epithelial-to-mesenchymal transition (EMT) pathways for PCa dissemination, which was additionally confirmed by an independent human PCa model (ARCAP-E/-M). Two converging nodes, CD46 (decreasing with metastatic potential) and DDX21 (increasing with metastatic potential), were used to test the clinical relevance of the networks. Intriguingly, both network nodes consistently added prognostic information for patients with PCa whereas CD46 loss predicted poor outcome independent of established parameters. Accordingly, depletion of CD46 in weakly metastatic PCa cells induced EMT-like properties in vitro and spontaneous micrometastasis formation in vivo. CONCLUSIONS The clinical and functional relevance of the dissemination-related interaction networks shown here could be successfully validated by proof-of-principle experiments. Therefore, we suggest a direct pro-metastatic, clinically relevant role for the multiple novel candidates included in this study; these should be further exploited by future studies.
Collapse
|
11
|
Pistollato F, Bernasconi C, McCarthy J, Campia I, Desaintes C, Wittwehr C, Deceuninck P, Whelan M. Alzheimer's Disease, and Breast and Prostate Cancer Research: Translational Failures and the Importance to Monitor Outputs and Impact of Funded Research. Animals (Basel) 2020; 10:E1194. [PMID: 32674379 PMCID: PMC7401638 DOI: 10.3390/ani10071194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Dementia and cancer are becoming increasingly prevalent in Western countries. In the last two decades, research focused on Alzheimer's disease (AD) and cancer, in particular, breast cancer (BC) and prostate cancer (PC), has been substantially funded both in Europe and worldwide. While scientific research outcomes have contributed to increase our understanding of the disease etiopathology, still the prevalence of these chronic degenerative conditions remains very high across the globe. By definition, no model is perfect. In particular, animal models of AD, BC, and PC have been and still are traditionally used in basic/fundamental, translational, and preclinical research to study human disease mechanisms, identify new therapeutic targets, and develop new drugs. However, animals do not adequately model some essential features of human disease; therefore, they are often unable to pave the way to the development of drugs effective in human patients. The rise of new technological tools and models in life science, and the increasing need for multidisciplinary approaches have encouraged many interdisciplinary research initiatives. With considerable funds being invested in biomedical research, it is becoming pivotal to define and apply indicators to monitor the contribution to innovation and impact of funded research. Here, we discuss some of the issues underlying translational failure in AD, BC, and PC research, and describe how indicators could be applied to retrospectively measure outputs and impact of funded biomedical research.
Collapse
Affiliation(s)
- Francesca Pistollato
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Camilla Bernasconi
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Janine McCarthy
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
- Physicians Committee for Responsible Medicine (PCRM), Washington, DC 20016, USA;
| | - Ivana Campia
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Christian Desaintes
- European Commission, Directorate General for Research and Innovation (RTD), 1000 Brussels, Belgium;
| | - Clemens Wittwehr
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Pierre Deceuninck
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| |
Collapse
|
12
|
Cho SY. Patient-derived xenografts as compatible models for precision oncology. Lab Anim Res 2020; 36:14. [PMID: 32461927 PMCID: PMC7238616 DOI: 10.1186/s42826-020-00045-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/06/2020] [Indexed: 01/02/2023] Open
Abstract
Cancer is a very heterogeneous disease, displaying heterogeneity between patients (inter-tumoral heterogeneity) and heterogeneity within a patient (intra-tumoral heterogeneity). Precision oncology is a diagnostic and therapeutic approach for cancers based on the stratification of patients using genomic and molecular profiling of tumors. To develop diagnostic and therapeutic tools for the application of precision oncology, appropriate preclinical mouse models that reflect tumor heterogeneity are required. Patient-derived xenograft (PDX) models are generated by the engraftment of patient tumors into immunodeficient mice that retain several aspects of the patient’s tumor characteristics, including inter-tumoral heterogeneity and intra-tumoral heterogeneity. Therefore, PDX models can be applied in various developmental steps of cancer diagnostics and therapeutics, such as biomarker development, companion diagnostics, drug efficacy testing, overcoming drug resistance, and co-clinical trials. This review summarizes the diverse aspects of PDX models, addressing the factors considered for PDX generation, application of PDX models for cancer research, and future directions of PDX models.
Collapse
Affiliation(s)
- Sung-Yup Cho
- 1Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 South Korea.,2Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,3Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, South Korea
| |
Collapse
|
13
|
In vivo magnetic resonance imaging of orthotopic prostate cancer. Biotechniques 2020; 69:395-403. [PMID: 32363906 DOI: 10.2144/btn-2020-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Methods for imaging orthotopic prostate tumors within the prostate or small tumors with extension outside the prostate are needed to more closely model human prostate tumors, which are most commonly located within the gland or may extend just through the gland. By comparing MR sequences, we found that the T2-based Dixon 'water only' sequence best visualized tumors within the prostate of mouse models in both young and old mice and that tumor weight derived from this sequence correlated highly with ex vivo tumor weight (r2 = 0.98, p < 0.001, n = 12). This should aid tumor detection, margin delineation and evaluation of tumor burden to enable studies including, but not limited to, evaluating the natural history of the disease, the mechanisms of action and the efficacy of therapeutic interventions.
Collapse
|
14
|
Wu Y, Wang J, Zheng X, Chen Y, Huang M, Huang Q, Xiao W, Wei H, Tian Z, Sun R, Sun C. Establishment and Preclinical Therapy of Patient-derived Hepatocellular Carcinoma Xenograft Model. Immunol Lett 2020; 223:33-43. [PMID: 32335145 DOI: 10.1016/j.imlet.2020.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 12/06/2019] [Accepted: 04/18/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a world-wide health problem. Poor and delayed diagnoses as well as high recurrence rate resulting in high mortality rate. In this study, we established a patient-derived xenograft (PDX) model from HCC patient, and continuously maintained with subcutaneous passage more than 20 times. This HCC PDX tumor exhibited the same histological characteristics with the HCC patient and could be used to verify therapeutic effect of liver cancer. We further evaluated this PDX model by experimental chemotherapy, demonstrating that this HCC PDX model was sensitive to sorafenib treatment. Further, the potential of natural killer cell-based immunotherapy for HCC was tested using this model. We found that NK92 cells effectively suppressed the tumor growth in vivo and prolonged the survival time of HCC-bearing PDX mice. This study indicates that HCC PDX model is a good platform to testify the efficacy of preclinical chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yuwei Wu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Jinyu Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Xiaodong Zheng
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Yongyan Chen
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Mei Huang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China
| | - Qiang Huang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science & Technology of China, Hefei, China
| | - Weihua Xiao
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China.
| | - Cheng Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China; Transplant & Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
15
|
Elbadawy M, Abugomaa A, Yamawaki H, Usui T, Sasaki K. Development of Prostate Cancer Organoid Culture Models in Basic Medicine and Translational Research. Cancers (Basel) 2020; 12:777. [PMID: 32218271 PMCID: PMC7226333 DOI: 10.3390/cancers12040777] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PC) is the most prevalent cancer in men and the second main cause of cancer-related death in Western society. The lack of proper PC models that recapitulate the molecular and genomic landscape of clinical disease has hampered progress toward translational research to understand the disease initiation, progression, and therapeutic responses in each patient. Although several models have been developed, they hardly emulated the complicated PC microenvironment. Precision medicine is an emerging approach predicting appropriate therapies for individual cancer patients by means of various analyses of individual genomic profiling and targeting specific cancer pathways. In PC, precision medicine also has the potential to impose changes in clinical practices. Here, we describe the various PC models with special focus on PC organoids and their values in basic medicine, personalized therapy, and translational researches in vitro and in vivo, which could help to achieve the full transformative power of cancer precision medicine.
Collapse
Affiliation(s)
- Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (M.E.); (A.A.); (K.S.)
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (M.E.); (A.A.); (K.S.)
- Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Dakahliya, Egypt
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan;
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (M.E.); (A.A.); (K.S.)
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan; (M.E.); (A.A.); (K.S.)
| |
Collapse
|
16
|
Contreras HR, López-Moncada F, Castellón EA. Cancer stem cell and mesenchymal cell cooperative actions in metastasis progression and hormone resistance in prostate cancer: Potential role of androgen and gonadotropin‑releasing hormone receptors (Review). Int J Oncol 2020; 56:1075-1082. [PMID: 32319606 DOI: 10.3892/ijo.2020.5008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the leading cause of male cancer‑associated mortality worldwide. Mortality is associated with metastasis and hormone resistance. Cellular, genetic and molecular mechanisms underlying metastatic progression and hormone resistance are poorly understood. Studies have investigated the local effects of gonadotropin‑releasing hormone (GnRH) analogs (used for androgen deprivation treatments) and the presence of the GnRH receptor (GnRH‑R) on PCa cells. Furthermore, cell subpopulations with stem‑like properties, or cancer stem cells, have been isolated and characterized using a cell culture system derived from explants of human prostate tumors. In addition, the development of preclinical orthotopic models of human PCa in a nonobese diabetic/severe combined immunodeficiency mouse model of compromised immunity has enabled the establishment of a reproducible system of metastatic progression in vivo. There is increasing evidence that metastasis is a complex process involving the cooperative actions of different cancer cell subpopulations, in which cancer stem‑like cells would be responsible for the final step of colonizing premetastatic niches. It has been hypothesized that PCa cells with stemness and mesenchymal signatures act cooperatively in metastatic progression and the inhibition of stemness genes, and that overexpression of androgen receptor (AR) and GnRH‑R decreases the rate the metastasis and sensitizes tumors to hormone therapy. The aim of the present review is to analyze the evidence regarding this cooperative process and the possible influence of stem‑like cell phenotypes, AR and GnRH‑R in metastatic progression and hormone resistance. These aspects may represent an important contribution in the understanding of the mechanisms underlying metastasis and hormone resistance in PCa, and potential routes to blocking these processes, enabling the development of novel therapies that would be particularly relevant for patients with metastatic and castration‑resistant PCa.
Collapse
Affiliation(s)
- Héctor R Contreras
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Fernanda López-Moncada
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Enrique A Castellón
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
17
|
Regression of castration-resistant prostate cancer by a novel compound QW07 targeting androgen receptor N-terminal domain. Cell Biol Toxicol 2020; 36:399-416. [PMID: 32002708 DOI: 10.1007/s10565-020-09511-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/07/2020] [Indexed: 12/24/2022]
Abstract
Androgen deprivation therapy (ADT) via surgical or chemical castration frequently fails to halt lethal castration-resistant prostate cancer (CRPC), which is induced by multiple mechanisms involving constitutive androgen receptor (AR) splice variants, AR mutation, and/or de novo androgen synthesis. The AR N-terminal domain (NTD) possesses most transcriptional activity and is proposed as a potential target for CRPC drug development. We constructed a screening system targeting AR-NTD transcription activity to screening a compound library and identified a novel small molecule compound named QW07. The function evaluation and mechanism investigation of QW07 were carried out in vitro and in vivo. QW07 bound to AR-NTD directly, blocked the transactivation of AR-NTD, blocked interactions between co-regulatory proteins and androgen response elements (AREs), inhibited the expression of genes downstream of AR, and inhibited prostate cancer growth in vitro and in vivo. QW07 was demonstrated as an AR-NTD-specific antagonist with the potential to inhibit both canonical and variant-mediated AR signaling to regress the CRPC xenografts and is proposed as a lead compound for a specific antagonist targeting AR-NTD.
Collapse
|
18
|
Fiordelisi MF, Cavaliere C, Auletta L, Basso L, Salvatore M. Magnetic Resonance Imaging for Translational Research in Oncology. J Clin Med 2019; 8:jcm8111883. [PMID: 31698697 PMCID: PMC6912299 DOI: 10.3390/jcm8111883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The translation of results from the preclinical to the clinical setting is often anything other than straightforward. Indeed, ideas and even very intriguing results obtained at all levels of preclinical research, i.e., in vitro, on animal models, or even in clinical trials, often require much effort to validate, and sometimes, even useful data are lost or are demonstrated to be inapplicable in the clinic. In vivo, small-animal, preclinical imaging uses almost the same technologies in terms of hardware and software settings as for human patients, and hence, might result in a more rapid translation. In this perspective, magnetic resonance imaging might be the most translatable technique, since only in rare cases does it require the use of contrast agents, and when not, sequences developed in the lab can be readily applied to patients, thanks to their non-invasiveness. The wide range of sequences can give much useful information on the anatomy and pathophysiology of oncologic lesions in different body districts. This review aims to underline the versatility of this imaging technique and its various approaches, reporting the latest preclinical studies on thyroid, breast, and prostate cancers, both on small laboratory animals and on human patients, according to our previous and ongoing research lines.
Collapse
|
19
|
Shi C, Chen X, Tan D. Development of patient-derived xenograft models of prostate cancer for maintaining tumor heterogeneity. Transl Androl Urol 2019; 8:519-528. [PMID: 31807428 DOI: 10.21037/tau.2019.08.31] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Prostate cancer (Pca) is a heterogeneous disease with multiple morphological patterns. Thus, the establishment of a patient-derived xenograft (PDX) model that retains key features of the primary tumor is of great significance. This review demonstrates the characteristics and advantages of the Pca PDX model and summarizes the main factors affecting the establishment of the model. Because this model well recapitulates the diverse heterogeneity observed in the clinic, it was extensively utilized to discover new therapeutic targets, screen drugs, and explore metastatic mechanisms. In the future, clinical phenotype and different stages of the Pca patient might be faithfully reflected by PDX model, which provides tremendous potential for understanding Pca biology and achieving individualized treatment.
Collapse
Affiliation(s)
- Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.,School of Basic Medical Sciences, the Chengdu Medical University, Xindu 610500, China
| | - Xue Chen
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.,School of Basic Medical Sciences, the Chengdu Medical University, Xindu 610500, China
| | - Dengxu Tan
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
20
|
Zhan S, Li J, Ge W. Multifaceted Roles of Asporin in Cancer: Current Understanding. Front Oncol 2019; 9:948. [PMID: 31608236 PMCID: PMC6771297 DOI: 10.3389/fonc.2019.00948] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
The small leucine-rich proteoglycan (SLRP) family consists of 18 members categorized into five distinct classes, the traditional classes I–III, and the non-canonical classes IV–V. Unlike the other class I SLRPs (decorin and biglycan), asporin contains a unique and conserved stretch of aspartate (D) residues in its N terminus, and germline polymorphisms in the D-repeat-length are associated with osteoarthritis and prostate cancer progression. Since the first discovery of asporin in 2001, previous studies have focused mainly on its roles in bone and joint diseases, including osteoarthritis, intervertebral disc degeneration and periodontal ligament mineralization. Recently, asporin gene expression was also reported to be dysregulated in tumor tissues of different types of cancer, and to act as oncogene in pancreatic, colorectal, gastric, and prostate cancers, and some types of breast cancer, though it is also reported to function as a tumor suppressor gene in triple-negative breast cancer. Furthermore, asporin is also positively or negatively correlated with tumor proliferation, migration, invasion, and patient prognosis through its regulation of different signaling pathways, including the TGF-β, EGFR, and CD44 pathways. In this review, we seek to elucidate the signaling pathways and functions regulated by asporin in different types of cancer and to highlight some important issues that require investigation in future research.
Collapse
Affiliation(s)
- Shaohua Zhan
- National Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China.,National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China.,Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
21
|
Zhao M, Zhu N, Hao F, Song Y, Wang Z, Ni Y, Ding L. The Regulatory Role of Non-coding RNAs on Programmed Cell Death Four in Inflammation and Cancer. Front Oncol 2019; 9:919. [PMID: 31620370 PMCID: PMC6759660 DOI: 10.3389/fonc.2019.00919] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022] Open
Abstract
Programmed cell death 4 (PDCD4) is a tumor suppressor gene implicated in many cellular functions, including transcription, translation, apoptosis, and the modulation of different signal transduction pathways. The downstream mechanisms of PDCD4 have been well-discussed, but its upstream regulators have not been systematically summarized. Noncoding RNAs (ncRNAs) are gene transcripts with no protein-coding potential but play a pivotal role in the regulation of the pathogenesis of solid tumors, cardiac injury, and inflamed tissue. In recent studies, many ncRNAs, especially microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), were found to interact with PDCD4 to manipulate its expression through transcriptional regulation and function as oncogenes or tumor suppressors. For example, miR-21, as a classic oncogene, was identified as the key regulator of PDCD4 by targeting its 3′-untranslated region (UTR) to promote tumor proliferation, migration, and invasion in colon, breast, and bladder carcinoma. Therefore, we reviewed the recently emerging pleiotropic regulation of PDCD4 by ncRNAs in cancer and inflammatory disorders and aimed to shed light on the mechanisms of associated diseases, which could be conducive to the development of novel treatment strategies for PDCD4-induced diseases.
Collapse
Affiliation(s)
- Mengxiang Zhao
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Nisha Zhu
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fengyao Hao
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuxian Song
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhiyong Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Nanjing, China
| | - Yanhong Ni
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liang Ding
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
22
|
Thakur G, Prakash G, Murthy V, Sable N, Menon S, Alrokayan SH, Khan HA, Murugaiah V, Bakshi G, Kishore U, Madan T. Human SP-D Acts as an Innate Immune Surveillance Molecule Against Androgen-Responsive and Androgen-Resistant Prostate Cancer Cells. Front Oncol 2019; 9:565. [PMID: 31355132 PMCID: PMC6637921 DOI: 10.3389/fonc.2019.00565] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/10/2019] [Indexed: 02/05/2023] Open
Abstract
Surfactant Protein D (SP-D), a pattern recognition innate immune molecule, has been implicated in the immune surveillance against cancer. A recent report showed an association of decreased SP-D expression in human prostate adenocarcinoma with an increased Gleason score and severity. In the present study, the SP-D expression was evaluated in primary prostate epithelial cells (PrEC) and prostate cancer cell lines. LNCaP, an androgen dependent prostate cancer cell line, exhibited significantly lower mRNA and protein levels of SP-D than PrEC and the androgen independent cell lines (PC3 and DU145). A recombinant fragment of human SP-D, rfhSP-D, showed a dose and time dependent binding to prostate cancer cells via its carbohydrate recognition domain. This study, for the first time, provides evidence of significant and specific cell death of tumor cells in rfhSP-D treated explants as well as primary tumor cells isolated from tissue biopsies of metatstatic prostate cancer patients. Viability of PrEC was not altered by rfhSP-D. Treated LNCaP (p53+/+) and PC3 (p53 -/-) cells exhibited reduced cell viability in a dose and time dependent manner and were arrested in G2/M and G1/G0 phase of the cell cycle, respectively. rfhSP-D treated LNCaP cells showed a significant upregulation of p53 whereas a significant downregulation of pAkt was observed in both PC3 and LNCaP cell lines. The rfhSP-D-induced apoptosis signaling cascade involved upregulation of Bax:Bcl2 ratio, cytochrome c and cleaved products of caspase 7. The study concludes that rfhSP-D induces apoptosis in prostate tumor explants as well as in androgen dependent and independent prostate cancer cells via p53 and pAkt pathways.
Collapse
Affiliation(s)
- Gargi Thakur
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| | - Gagan Prakash
- Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Vedang Murthy
- Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Nilesh Sable
- Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Santosh Menon
- Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Salman H. Alrokayan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Haseeb A. Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Valarmathy Murugaiah
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Ganesh Bakshi
- Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Taruna Madan
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
23
|
Endepols H, Morgenroth A, Zlatopolskiy BD, Krapf P, Zischler J, Richarz R, Muñoz Vásquez S, Neumaier B, Mottaghy FM. Peripheral ganglia in healthy rats as target structures for the evaluation of PSMA imaging agents. BMC Cancer 2019; 19:633. [PMID: 31242896 PMCID: PMC6595687 DOI: 10.1186/s12885-019-5841-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/16/2019] [Indexed: 02/08/2023] Open
Abstract
Background The recent implementation of PET with prostate specific membrane antigen (PSMA)-specific radiotracers into the clinical practice has resulted in the significant improvement of accuracy in the detection of prostate carcinoma (PCa). PSMA-expression in ganglia has been regarded as an important pitfall in prostate carcinoma-PET diagnostics but has not found any practical use for diagnosis or therapy. Methods We explored this phenomenon and demonstrated the applicability of peripheral ganglia in healthy rats as surrogates for small PSMA positive lesions for the preclinical evaluation of diagnostic PCa PET probes. Healthy rats were measured with PET/CT using the tracers [18F]DCFPyL, [Al18F]PSMA-11 and [68Ga]PSMA-11. Sections of ganglia were stained with an anti-PSMA antibody. [18F]DCFPyL uptake in ganglia was compared to that in LNCaP tumor xenografts in mice. Results Whereas [18F]DCFPyL and [68Ga]PSMA-11 were stable in vivo and accumulated in peripheral ganglia, [Al18F]PSMA-11 suffered from fast in vivo deflourination resulting in high bone uptake. Ganglionic PSMA expression was confirmed by immunohistochemistry. [18F]DCFPyL uptake and signal-to-noise ratio in the superior cervical ganglion was not significantly different from LNCaP xenografts. Conclusions Our results demonstrated the non-inferiority of the novel model compared to conventionally used tumor xenografts in immune compromised rodents with regard to reproducibility and stability of the PSMA signal. Furthermore, the model involves less expense and efforts while it is permanently available and avoids tumor-growth associated animal morbidity and distress. To the best of our knowledge, this is the first tumor-free model suitable for the in vivo evaluation of tumor imaging agents.
Collapse
Affiliation(s)
- Heike Endepols
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany.,Department of Nuclear Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany.,Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital, RWTH Aachen, Pauwelsstraße 30, Aachen, 52074, Germany
| | - Boris D Zlatopolskiy
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany.,Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Philipp Krapf
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany.,Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Johannes Zischler
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany.,Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Raphael Richarz
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany
| | - Sergio Muñoz Vásquez
- Department of Nuclear Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937, Cologne, Germany. .,Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428, Jülich, Germany.
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital, RWTH Aachen, Pauwelsstraße 30, Aachen, 52074, Germany. .,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center X, Maastricht, the Netherlands.
| |
Collapse
|
24
|
[ 89Zr]A2cDb Immuno-PET of Prostate Cancer in a Human Prostate Stem Cell Antigen Knock-in (hPSCA KI) Syngeneic Model. Mol Imaging Biol 2019; 22:367-376. [PMID: 31209779 DOI: 10.1007/s11307-019-01386-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE A great challenge in the diagnosis and treatment of prostate cancer is distinguishing between indolent or local disease and aggressive or metastatic disease. Antibody-based positron emission tomography (immuno-PET) as a cancer-specific imaging modality could improve diagnosis of primary disease, aid the detection of metastases to regional lymph nodes as well as to distant sites (e.g., bone), and monitor response to therapy. PROCEDURE In search for a more physiologically relevant disease model, a human prostate stem cell antigen knock-in (hPSCA KI) mouse model was generated. The use of a syngeneic prostate cancer cell line transduced to express human PSCA (RM-9-hPSCA) enabled the evaluation of anti-PSCA immuno-PET in immunocompetent mice and in the context of normal tissue expression of PSCA. Two PSCA-specific humanized antibody fragments, A11 minibody and A2 cys-diabody, were radiolabeled with positron emitters iodine-124 and zirconium-89, respectively ([124I]A11 Mb and [89Zr]A2cDb), and used for immuno-PET in wild-type, hPSCA KI and tumor-bearing mice. RESULTS The hPSCA KI mice express PSCA at low levels in the normal prostate, bladder and stomach, reproducing the expression pattern seen in humans. [124I]A11 Mb immuno-PET detected increased levels of PSCA expression in the stomach, and because I-124 is non-residualizing, very little activity was seen in organs of clearance (liver, kidney, spleen). However, due to the longer half-life of the 80 kDa protein, blood activity (and thus urine activity) at 20 h postinjection remains high. The smaller 50 kDa [89Zr]A2cDb cleared faster, resulting in lower blood and background activity, despite the use of a residualizing radiometal. Importantly, [89Zr]A2cDb immuno-PET showed antigen-specific targeting of PSCA-expressing tumors and minimal nonspecific uptake in PSCA-negative controls. CONCLUSION Tracer biodistribution was not significantly impacted by normal tissue expression of PSCA. [89Zr]A2cDb immuno-PET yielded high tumor-to-blood ratio at early time points. Rapid renal clearance of the 50 kDa tracer resulted in an unobstructed view of the pelvic region at 20 h postinjection that would allow the detection of cancer in the prostate.
Collapse
|
25
|
Zhu G, Foletti D, Liu X, Ding S, Melton Witt J, Hasa-Moreno A, Rickert M, Holz C, Aschenbrenner L, Yang AH, Kraynov E, Evering W, Obert L, Lee C, Sai T, Mistry T, Lindquist KC, Van Blarcom T, Strop P, Chaparro-Riggers J, Liu SH. Targeting CLDN18.2 by CD3 Bispecific and ADC Modalities for the Treatments of Gastric and Pancreatic Cancer. Sci Rep 2019; 9:8420. [PMID: 31182754 PMCID: PMC6557842 DOI: 10.1038/s41598-019-44874-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
Human CLDN18.2 is highly expressed in a significant proportion of gastric and pancreatic adenocarcinomas, while normal tissue expression is limited to the epithelium of the stomach. The restricted expression makes it a potential drug target for the treatment of gastric and pancreatic adenocarcinoma, as evidenced by efforts to target CLDN18.2 via naked antibody and CAR-T modalities. Herein we describe CLDN18.2-targeting via a CD3-bispecific and an antibody drug conjugate and the characterization of these potential therapeutic molecules in efficacy and preliminary toxicity studies. Anti-hCLDN18.2 ADC, CD3-bispecific and diabody, targeting a protein sequence conserved in rat, mouse and monkey, exhibited in vitro cytotoxicity in BxPC3/hCLDN18.2 (IC50 = 1.52, 2.03, and 0.86 nM) and KATO-III/hCLDN18.2 (IC50 = 1.60, 0.71, and 0.07 nM) respectively and inhibited tumor growth of pancreatic and gastric patient-derived xenograft tumors. In a rat exploratory toxicity study, the ADC was tolerated up to 10 mg/kg. In a preliminary assessment of tolerability, the anti-CLDN18.2 diabody (0.34 mg/kg) did not produce obvious signs of toxicity in the stomach of NSG mice 4 weeks after dosing. Taken together, our data indicate that targeting CLDN18.2 with an ADC or bispecific modality could be a valid therapeutic approach for the treatment of gastric and pancreatic cancer.
Collapse
Affiliation(s)
- Guoyun Zhu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.
| | - Davide Foletti
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,23 and Me, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Xiaohui Liu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Sheng Ding
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Gilead Sciences, 333 Lakeside Drive, Foster City, CA, 94404, USA
| | - Jody Melton Witt
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Grifols Diagnostic Solutions, 6455 Christie Ave B-334C, Emeryville, CA, 94608, USA
| | - Adela Hasa-Moreno
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Kodiak Sciences Inc., 2631 Hanover St, Palo Alto, CA, 94304, USA
| | - Mathias Rickert
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Applied Molecular Transport, 1 Tower Place, Suite 850, South San Francisco, CA, 94080, USA
| | - Charles Holz
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Grifols Diagnostic Solutions, 6455 Christie Ave B-334C, Emeryville, CA, 94608, USA
| | - Laura Aschenbrenner
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA.,Covance Inc. Early Phase Development Solutions, 3301 Kinsman Blvd, Madison, WI, 53704, USA
| | - Amy H Yang
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Eugenia Kraynov
- BioMedicine Design, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Winston Evering
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Leslie Obert
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 280 Shennecossett Rd, Groton, CT, 06340, USA.,GSK, 1250 South Collegeville Road, Collegeville, PA, 19426, USA
| | - Chenyu Lee
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Alector, 151 Oyster Point Blvd #300, South San Francisco, CA, 94080, USA
| | - Tao Sai
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Tina Mistry
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Kevin C Lindquist
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Thomas Van Blarcom
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Allogene Therapeutics, 210 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Pavel Strop
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Bristol-Myers Squibb, 700 Bay Rd suite A, Redwood City, CA, 94063, USA
| | - Javier Chaparro-Riggers
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Shu-Hui Liu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Multitude Therapeutics, Abmart, 3698 Haven Avenue Suite A, Redwood City, CA, 94063, USA
| |
Collapse
|
26
|
Logothetis C, Morris MJ, Den R, Coleman RE. Current perspectives on bone metastases in castrate-resistant prostate cancer. Cancer Metastasis Rev 2019; 37:189-196. [PMID: 29380085 PMCID: PMC5801387 DOI: 10.1007/s10555-017-9719-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent noncutaneous cancer occurring in men. On average, men with localized prostate cancer have a high 10-year survival rate, and many can be cured. However, men with metastatic castrate-resistant prostate cancer have incurable disease with poor survival despite intensive therapy. This unmet need has led to recent advances in therapy aimed at treating bone metastases resulting from prostate cancer. The bone microenvironment lends itself to metastases in castrate-resistant prostate cancer, as a result of complex interactions between the microenvironment and tumor cells. The development of 223radium dichloride (Ra-223) to treat symptomatic bone metastases has improved survival in men with metastatic castrate-resistant prostate cancer. Moreover, Ra-223 may have effects on the tumor microenvironment that enhance its activity. Ra-223 treatment has been shown to prolong survival, and its effects on the immune system are under investigation. Because prostate cancer affects a sizable portion of the adult male population, understanding how it metastasizes to bone is an important step in advancing therapy. Clinical trials that are underway should yield new information on whether Ra-223 synergizes effectively with immunotherapy agents and whether Ra-223 has enhancing effects on the immune system in patients with prostate cancer.
Collapse
Affiliation(s)
| | - Michael J Morris
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Robert Den
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | |
Collapse
|
27
|
Engineered bone for probing organotypic growth and therapy response of prostate cancer tumoroids in vitro. Biomaterials 2019; 197:296-304. [PMID: 30682644 DOI: 10.1016/j.biomaterials.2019.01.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/24/2018] [Accepted: 01/20/2019] [Indexed: 01/23/2023]
Abstract
Mechanistic analysis of metastatic prostate cancer (PCa) biology and therapy response critically depends upon clinically relevant three-dimensional (3D) bone-like, organotypic culture. We here combine an engineered bone-mimetic environment (BME) with longitudinal microscopy to test the growth and therapy response of 3D PCa tumoroids. Besides promoting both tumor-cell autonomous and microenvironment-dependent growth in PCa cell lines and patient-derived xenograft cells, the BME enables in vivo-like tumor cell response to therapy, and reveals bone stroma dependent resistance to chemotherapy and BME-targeted localization and induction of cytoxicity by Radium-223. The BME platform will allow the propagation, compound screening and mechanistic dissection of patient-derived bone tumor isolates and applications toward personalized medicine.
Collapse
|
28
|
Wells A, Clark A, Bradshaw A, Ma B, Edington H. The great escape: How metastases of melanoma, and other carcinomas, avoid elimination. Exp Biol Med (Maywood) 2019; 243:1245-1255. [PMID: 30764707 DOI: 10.1177/1535370218820287] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT Cancers kill mainly because metastatic disease is resistant to systemic therapies. It was hoped that newer targeted and immunomodulatory interventions could overcome these issues. However, recent findings point to a generalized resistance to elimination imparted by both cancer-intrinsic and -extrinsic changes to provide survival advantages to the disseminated tumor cells. Here, we present a novel conceptual framework for the microenvironmental inputs and changes that contribute to this generalized therapeutic resistance. In addition we address the issues of experimental systems in terms of studying this phenomenon with their advantages and limitations. This is meant to spur studies into this critical aspect of tumor progression that directly leads to cancer mortality.
Collapse
Affiliation(s)
- Alan Wells
- 1 Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,2 Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA.,3 Pittsburgh VA Medical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA.,4 McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.,5 Hillman Cancer Centers of UPMC, Pittsburgh, PA 15232, USA
| | - Amanda Clark
- 1 Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Andrew Bradshaw
- 1 Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,3 Pittsburgh VA Medical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Bo Ma
- 1 Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,3 Pittsburgh VA Medical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA.,5 Hillman Cancer Centers of UPMC, Pittsburgh, PA 15232, USA
| | - Howard Edington
- 6 Department of Surgery, Allegheny Health Network, Pittsburgh, PA 15224, USA
| |
Collapse
|
29
|
Cabello M, Ge H, Aracil C, Moschou D, Estrela P, Manuel Quero J, I Pascu S, R F Rocha P. Extracellular Electrophysiology in the Prostate Cancer Cell Model PC-3. SENSORS (BASEL, SWITZERLAND) 2019; 19:E139. [PMID: 30609788 PMCID: PMC6339143 DOI: 10.3390/s19010139] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Although prostate cancer is one of the most common cancers in the male population, its basic biological function at a cellular level remains to be fully understood. This lack of in depth understanding of its physiology significantly hinders the development of new, targeted and more effective treatment strategies. Whilst electrophysiological studies can provide in depth analysis, the possibility of recording electrical activity in large populations of non-neuronal cells remains a significant challenge, even harder to address in the picoAmpere-range, which is typical of cellular level electrical activities. In this paper, we present the measurement and characterization of electrical activity of populations of prostate cancer cells PC-3, demonstrating for the first time a meaningful electrical pattern. The low noise system used comprises a multi-electrode array (MEA) with circular gold electrodes on silicon oxide substrates. The extracellular capacitive currents present two standard patterns: an asynchronous sporadic pattern and a synchronous quasi-periodic biphasic spike pattern. An amplitude of ±150 pA, a width between 50⁻300 ms and an inter-spike interval around 0.5 Hz characterize the quasi-periodic spikes. Our experiments using treatment of cells with Gd³⁺, known as an inhibitor for the Ca²⁺ exchanges, suggest that the quasi-periodic signals originate from Ca²⁺ channels. After adding the Gd³⁺ to a population of living PC-3 cells, their electrical activity considerably decreased; once the culture was washed, thus eliminating the Gd³⁺ containing medium and addition of fresh cellular growth medium, the PC-3 cells recovered their normal electrical activity. Cellular viability plots have been carried out, demonstrating that the PC-3 cells remain viable after the use of Gd³⁺, on the timescale of this experiment. Hence, this experimental work suggests that Ca²⁺ is significantly affecting the electrophysiological communication pattern among PC-3 cell populations. Our measuring platform opens up new avenues for real time and highly sensitive investigations of prostate cancer signalling pathways.
Collapse
Affiliation(s)
- Miguel Cabello
- Department of Electronic Engineering, Escuela Superior de Ingenieros, University of Seville, 41004 Seville, Spain.
| | - Haobo Ge
- Department of Chemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - Carmen Aracil
- Department of Electronic Engineering, Escuela Superior de Ingenieros, University of Seville, 41004 Seville, Spain.
| | - Despina Moschou
- Centre for Biosensors, Bioelectronics and Biodevices (C3Bio), Department of Electronic and Electrical Engineering, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - Pedro Estrela
- Centre for Biosensors, Bioelectronics and Biodevices (C3Bio), Department of Electronic and Electrical Engineering, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - Jose Manuel Quero
- Department of Electronic Engineering, Escuela Superior de Ingenieros, University of Seville, 41004 Seville, Spain.
| | - Sofia I Pascu
- Department of Chemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| | - Paulo R F Rocha
- Centre for Biosensors, Bioelectronics and Biodevices (C3Bio), Department of Electronic and Electrical Engineering, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
30
|
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
|
31
|
Kermanshah L, Poudineh M, Ahmed S, Nguyen LNM, Srikant S, Makonnen R, Pena Cantu F, Corrigan M, Kelley SO. Dynamic CTC phenotypes in metastatic prostate cancer models visualized using magnetic ranking cytometry. LAB ON A CHIP 2018; 18:2055-2064. [PMID: 29923581 PMCID: PMC6368266 DOI: 10.1039/c8lc00310f] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Tumors can shed thousands of cells into the circulation daily. These circulating tumor cells (CTCs) are heterogeneous, and their phenotypes change dynamically. Real-time monitoring of CTC phenotypes is crucial to elucidate the role of CTCs in the metastatic cascade. Here, we monitor phenotypic changes in CTCs in mice xenografted with tumors with varying aggressiveness during cancer progression and a course of chemotherapy to study the metastatic potential of CTCs and changes in the properties of these cells in response to treatment. A new device that enables magnetic ranking cytometry (MagRC) is employed to profile the phenotypic properties of CTCs. Overall, CTCs from metastatic xenografts in mice display dynamic and heterogeneous profiles while non-metastatic models had static profiles. Decreased heterogeneity followed by a reduction in metastasis incidence was observed after a course of chemotherapy administered to highly metastatic xenografts. Phenotypic profiling of CTCs could be employed to monitor disease progression and predict therapeutic responses.
Collapse
Affiliation(s)
- Leyla Kermanshah
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Vanacore D, Boccellino M, Rossetti S, Cavaliere C, D'Aniello C, Di Franco R, Romano FJ, Montanari M, La Mantia E, Piscitelli R, Nocerino F, Cappuccio F, Grimaldi G, Izzo A, Castaldo L, Pepe MF, Malzone MG, Iovane G, Ametrano G, Stiuso P, Quagliuolo L, Barberio D, Perdonà S, Muto P, Montella M, Maiolino P, Veneziani BM, Botti G, Caraglia M, Facchini G. Micrornas in prostate cancer: an overview. Oncotarget 2018; 8:50240-50251. [PMID: 28445135 PMCID: PMC5564846 DOI: 10.18632/oncotarget.16933] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/25/2017] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer is the second highest cause of cancer mortality after lung tumours. In USA it affects about 2.8 million men and the incidence increases with age in many countries. Therefore, early diagnosis is a very important step for patient clinical evaluation and for a selective and efficient therapy. The study of miRNAs' functions and molecular mechanisms has brought new knowledge in biological processes of cancer. In prostate cancer there is a deregulation of several miRNAs that may function as tumour suppressors or oncogenes. The aim of this review is to analyze the progress made to our understanding of the role of miRNA dysregulation in prostate cancer tumourigenesis.
Collapse
Affiliation(s)
- Daniela Vanacore
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli" Naples, Naples, Italy
| | - Mariarosaria Boccellino
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli" Naples, Naples, Italy
| | - Sabrina Rossetti
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Carla Cavaliere
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Onco-Ematology Medical Oncology, S.G. Moscati Hospital of Taranto, Taranto, Italy
| | - Carmine D'Aniello
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Medical Oncology, A.O.R.N. dei COLLI "Ospedali Monaldi-Cotugno-CTO", Napoli, Italy
| | - Rossella Di Franco
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Francesco Jacopo Romano
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy
| | - Micaela Montanari
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Elvira La Mantia
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Raffaele Piscitelli
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pharmacy Unit, Istituto Nazionale Tumori, Istituto Nazionale Tumori-Fondazione G. Pascale, Naples, Italy
| | - Flavia Nocerino
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Epidemiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Francesca Cappuccio
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Psicology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Giovanni Grimaldi
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Alessandro Izzo
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Luigi Castaldo
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Maria Filomena Pepe
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Maria Gabriella Malzone
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Gelsomina Iovane
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Gianluca Ametrano
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Paola Stiuso
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli" Naples, Naples, Italy
| | - Lucio Quagliuolo
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli" Naples, Naples, Italy
| | - Daniela Barberio
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Psicology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Sisto Perdonà
- Division of Urology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Paolo Muto
- Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Maurizio Montella
- Epidemiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale Tumori, Istituto Nazionale Tumori-Fondazione G. Pascale, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, Naples, Italy.,Scientific Directorate, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli" Naples, Naples, Italy
| | - Gaetano Facchini
- Progetto ONCONET2.0, Linea progettuale 14 per l'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| |
Collapse
|
33
|
Epithelial-mesenchymal transition in prostate cancer: an overview. Oncotarget 2018; 8:35376-35389. [PMID: 28430640 PMCID: PMC5471062 DOI: 10.18632/oncotarget.15686] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/15/2017] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer is a main urological disease associated with significant morbidity and mortality. Radical prostatectomy and radiotherapy are potentially curative for localized prostate cancer, while androgen deprivation therapy is the initial systemic therapy for metastatic prostate disease. However, despite temporary response, most patients relapse and evolve into castration resistant cancer. Epithelial-mesenchymal transition (EMT) is a complex gradual process that occurs during embryonic development and/or tumor progression. During this process, cells lose their epithelial characteristics and acquire mesenchymal features. Increasing evidences indicate that EMT promotes prostate cancer metastatic progression and it is closely correlated with increased stemness and drug resistance. In this review, we discuss the main molecular events that directly or indirectly govern the EMT program in prostate cancer, in order to better define the role and the mechanisms underlying this process in prostate cancer progression and therapeutic resistance.
Collapse
|
34
|
Wang L, Xu M, Li Z, Shi M, Zhou X, Jiang X, Bryant J, Balk S, Ma J, Isaacs W, Xu X. Calcium and CaSR/IP3R in prostate cancer development. Cell Biosci 2018. [DOI: 10.1186/s13578-018-0217-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
35
|
Yin ZY, Qi SY, Cheng X, Guo L, Chen HY, Shi M. [Establishment of mouse model of bone metastasis of prostate cancer and breast cancer via femoral artery injection of tumor cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:914-918. [PMID: 28736367 PMCID: PMC6765506 DOI: 10.3969/j.issn.1673-4254.2017.07.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To explore an efficient and reliable method for establishing an animal model of bone metastasis of tumors. METHODS Male C57BL/6J mice were randomized into 4 equal groups to receive injections of normal saline or prostate cancer RM-1 cell suspension via the femoral artery or the external iliac artery, and breast cancer 4T1-luc cells were injected in 15 female BALB/c mice via the femoral artery. The operation time, postoperative survival rate, and the tumor formation rates in the mice with two different injection methods were compared. The tumor metastasis in the mice was evaluated with in vivo imaging. RESULTS The mean time for hemostasis time was 2.53∓1.75 min in mice receiving tumor cell injection via the femoral artery, significantly shorter than that in mice with injections via the external iliac artery (4.70∓1.63 min; P<0.05); the mean operation time was 14.67∓2.16 min and 22.47∓3.50 min in the two groups, respectively (P<0.05). At 21 days after the operation, the survival rate was 93.3% in femoral artery injection group, significantly higher than that in external iliac artery injection group (66.7%;P<0.05). The tumor metastasis rate was 100% in both groups. CONCLUSION Injection of the tumor cells via the femoral artery is more suitable for establishing mouse models of bone metastasis of cancers.
Collapse
Affiliation(s)
- Zhao-Yang Yin
- The Health Team, Shandong Armed Police Corps Detachment of Laiwu City, Laiwu 271199, China.E-mail:
| | | | | | | | | | | |
Collapse
|
36
|
Yin ZY, Qi SY, Cheng X, Guo L, Chen HY, Shi M. [Establishment of mouse model of bone metastasis of prostate cancer and breast cancer via femoral artery injection of tumor cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:914-918. [PMID: 28736367 PMCID: PMC6765506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To explore an efficient and reliable method for establishing an animal model of bone metastasis of tumors. METHODS Male C57BL/6J mice were randomized into 4 equal groups to receive injections of normal saline or prostate cancer RM-1 cell suspension via the femoral artery or the external iliac artery, and breast cancer 4T1-luc cells were injected in 15 female BALB/c mice via the femoral artery. The operation time, postoperative survival rate, and the tumor formation rates in the mice with two different injection methods were compared. The tumor metastasis in the mice was evaluated with in vivo imaging. RESULTS The mean time for hemostasis time was 2.53∓1.75 min in mice receiving tumor cell injection via the femoral artery, significantly shorter than that in mice with injections via the external iliac artery (4.70∓1.63 min; P<0.05); the mean operation time was 14.67∓2.16 min and 22.47∓3.50 min in the two groups, respectively (P<0.05). At 21 days after the operation, the survival rate was 93.3% in femoral artery injection group, significantly higher than that in external iliac artery injection group (66.7%;P<0.05). The tumor metastasis rate was 100% in both groups. CONCLUSION Injection of the tumor cells via the femoral artery is more suitable for establishing mouse models of bone metastasis of cancers.
Collapse
Affiliation(s)
- Zhao-Yang Yin
- The Health Team, Shandong Armed Police Corps Detachment of Laiwu City, Laiwu 271199, China.E-mail:
| | | | | | | | | | | |
Collapse
|
37
|
Nadar RA, Margiotta N, Iafisco M, van den Beucken JJJP, Boerman OC, Leeuwenburgh SCG. Bisphosphonate-Functionalized Imaging Agents, Anti-Tumor Agents and Nanocarriers for Treatment of Bone Cancer. Adv Healthc Mater 2017; 6. [PMID: 28207199 DOI: 10.1002/adhm.201601119] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Indexed: 12/14/2022]
Abstract
Bone metastases result from the invasion of primary tumors to bone. Current treatment modalities include local treatments such as surgery and radiotherapy, while systemic treatments include chemotherapy and (palliative) treatment of skeletal metastases. Nevertheless, once bone metastases have been established they remain incurable leading to morbidity and mortality. Bisphosphonates are a well-established class of drugs, which are increasingly applied in the treatment of bone cancers owing to their effective inhibition of tumor cells and suppression of bone metastases. The increased understanding of the mechanism of action of bisphosphonates on bone and tumor cells has prompted the development of novel bisphosphonate-functionalized imaging and therapeutic agents. This review provides an update on the preclinical efficacy of bisphosphonate-functionalized fluorophore, anti-tumor agents and nanocarriers for the treatment of bone metastases. After an overview of the general characteristics of bisphosphonates and their mechanisms of action, an outline is provided on the various conjugation strategies that have become available to functionalize imaging agents, anti-tumor agents and nanocarriers with bisphosphonates. Finally, the efficacy of these bisphosphonate-modified agents and carriers in preclinical studies is evaluated by reviewing their potential to target tumors and inhibit tumor growth in clinically relevant animal models for the treatment of bone cancer.
Collapse
Affiliation(s)
- Robin A. Nadar
- Department of Biomaterials; Radboud University Medical Center; Philips van Leydenlaan 25 6525 EX Nijmegen The Netherlands
| | - Nicola Margiotta
- Dipartimento di Chimica; Università degli Studi di Bari Aldo Moro; Via E. Orabona 4 70125 Bari Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics (ISTEC); National Research Council (CNR); Via Granarolo 64 48018 Faenza Italy
| | | | - Otto C. Boerman
- Department of Nuclear Medicine; Radboud University Medical Center; Geert Grooteplein Zuid 10 6525 AG Nijmegen The Netherlands
| | - Sander C. G. Leeuwenburgh
- Department of Biomaterials; Radboud University Medical Center; Philips van Leydenlaan 25 6525 EX Nijmegen The Netherlands
| |
Collapse
|
38
|
Xu C, Gu L. The diagnostic effect of serum miR-196b as biomarker in colorectal cancer. Biomed Rep 2016; 6:39-45. [PMID: 28123705 PMCID: PMC5244757 DOI: 10.3892/br.2016.815] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 11/11/2016] [Indexed: 12/11/2022] Open
Abstract
The microRNA, miR-196b, serves a role in normal cell differentiation, proliferation and tumorigenesis of different types of cancer. The aim of the present study was to explore the serum expression of miR-196b in colorectal cancer (CRC) and its correlation with clinicopathological features. Sera samples were obtained from 103 patients with CRC, 51 patients with colorectal adenoma (Ad) and 100 healthy individuals for the present study. The serum expression of miR-196b in sera samples of the three cohorts was detected using reverse transcription-quantitative polymerase chain reaction. The diagnostic value of miR-196b in the serum of the patients with CRC was evaluated by receiver operating characteristic (ROC) curve and survival analysis, using the Kaplan-Meier method, which was performed with the data from a 5-year follow-up. The expression of miR-196b in the serum of patients with CRC was significantly higher compared with that in Ad patients or healthy individuals (all P<0.001), and the overexpression of serum miR-196b was clearly associated with lymph node invasion, differentiation, and the tumor-lymph nodes-metastasis stage (all P<0.05). ROC curve analysis demonstrated that, comparing patients with CRC with healthy individuals, the area under the curve of serum miR-196b was 0.8135, and its specificity and sensitivity were 63 and 87.38%, respectively, at a diagnostic threshold of −4.785. Patients with CRC of miR-196b-high status had shorter overall survival and disease-free survival rates compared with those of miR-196b-low status. In conclusion, the results of the present study demonstrated that serum miR-196b is upregulated in CRC, and may have an application as a diagnostic and prognostic biomarker for patients with CRC.
Collapse
Affiliation(s)
- Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|