1
|
Huang Y, Zhan Y, Zhan Y. Psychological stress on cancer progression and immunosenescence. Semin Cancer Biol 2025; 113:85-99. [PMID: 40348001 DOI: 10.1016/j.semcancer.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/31/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Diagnosis and treatment of cancer constitute a deeply stressful experience that involves unique and common problems and generates uncertainty, fear and emotional distress. Furthermore, there are reciprocal interactions between psychological stress and cancer in the clinical settings. Therefore, it is crucial to understand the links of stress with cancer. A growing body of epidemiological and preclinical studies have suggested that stress affects cancer progression, and metastasis and treatment outcomes. Furthermore, stress elicits premature aging and deterioration of the immune system (known as immunosenescence), causing vulnerability to infections, autoimmune diseases, and cancers. In this review, we describe recent advances in how stress affects cancer progression through specific stress hormones and receptor systems as well as intracellular molecular processes, and discuss how stress-evoked neuroendocrine molecules regulate local and systemic immune responses in the tumor microenvironment. Furthermore, we review the molecular mechanisms of immunosenescence and evidence of psychological stress-evoked immunosenescence, highlighting the clinical value for available psychological and/or pharmacological interventions for psychological stress in patients with cancer. Based on existing evidence and emerging mechanistic insights, factors linked with psychological stress, immunosenescence and complications in cancer survivors need to be determined in future studies, and screening programs should be added to follow-up.
Collapse
Affiliation(s)
- Yinglin Huang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Yuhong Zhan
- Pharmacy Department, The 966th Hospital of PLA Joint Logistics Support Force, Dandong, Liaoning 118000, China
| | - Yuhua Zhan
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
2
|
Abrignani MG, Lucà F, Abrignani V, Nucara M, Grosseto D, Lestuzzi C, Mistrangelo M, Passaretti B, Rao CM, Parrini I. Risk Factors and Prevention of Cancer and CVDs: A Chicken and Egg Situation. J Clin Med 2025; 14:3083. [PMID: 40364115 PMCID: PMC12072322 DOI: 10.3390/jcm14093083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/30/2025] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular diseases and cancer are the two primary causes of mortality worldwide. Although traditionally regarded as distinct pathologies, they share numerous pathophysiological mechanisms and risk factors, including chronic inflammation, insulin resistance, obesity, and metabolic dysregulation. Notably, several cancers have been identified as closely linked to cardiovascular diseases, including lung, breast, prostate, and colorectal cancers, as well as hematological malignancies, such as leukemia and lymphoma. Additionally, renal and pancreatic cancers exhibit a significant association with cardiovascular complications, partly due to shared risk factors and the cardiotoxic effects of cancer therapies. Addressing the overlapping risk factors through lifestyle modifications-such as regular physical activity, a balanced diet, and cessation of smoking and alcohol-has proven effective in reducing both CV and oncological morbidity and mortality. Furthermore, even in patients with established cancer, structured interventions targeting physical activity, nutritional optimization, and smoking cessation have been associated with improved outcomes. Beyond lifestyle modifications, pharmacological strategies play a crucial role in the prevention of both diseases. Several cardiovascular medications, including statins, aspirin, beta-blockers, and metformin, exhibit pleiotropic effects that extend beyond their primary indications, demonstrating potential anti-neoplastic properties in preclinical and observational studies. Recently, novel therapeutic agents have garnered attention for their possible cardioprotective and metabolic benefits. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and sodium-glucose cotransporter-2 inhibitors (SGLT2is), initially developed for managing type 2 diabetes, have shown CV and renal protective effects, alongside emerging evidence of their role in modulating cancer-related metabolic pathways. Inclisiran, a small interfering RNA targeting PCSK9, effectively lowers LDL cholesterol and may contribute to reducing CV risk, with potential implications for tumor biology. Additionally, sacubitril/valsartan, an angiotensin receptor-neprilysin inhibitor, has revolutionized heart failure management by improving hemodynamic parameters and exerting anti-inflammatory effects that may have broader implications for chronic disease prevention. Given the intricate interplay between CVD and cancer, further research is essential to clarify the exact mechanisms linking these conditions and assessing the potential of CV therapies in cancer prevention. This review aims to examine shared risk factors, consider the role of pharmacological and lifestyle interventions, and emphasize crucial epidemiological and mechanistic insights into the intersection of CV and oncological health.
Collapse
Affiliation(s)
| | - Fabiana Lucà
- O.U. Interventional Cardiology-ICCU, A.O. Bianchi Melacrino Morelli, 89128 Reggio Calabria, Italy;
| | - Vincenzo Abrignani
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90133 Palermo, Italy;
| | - Mariacarmela Nucara
- O.U. Interventional Cardiology-ICCU, A.O. Bianchi Melacrino Morelli, 89128 Reggio Calabria, Italy;
| | | | | | - Marinella Mistrangelo
- Department Rete Oncologica Piemonte e Valle d’Aosta, Città della Salute e della Scienza, 10126 Turin, Italy;
| | - Bruno Passaretti
- Cardiology Unit, Homanitas, Gavazzeni-Castelli, 24125 Bergamo, Italy;
| | | | - Iris Parrini
- Cardiology Department, Ospedale Mauriziano Umberto I, 10128 Turin, Italy;
| |
Collapse
|
3
|
Qi J, Cheng H, Su L, Li J, Cheng F. A novel exosome-related prognostic risk model for thyroid cancer. Asia Pac J Clin Oncol 2025; 21:190-198. [PMID: 38577908 DOI: 10.1111/ajco.14063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/13/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024]
Abstract
AIM The aim was to build an exosome-related gene (ERG) risk model for thyroid cancer (TC) patients. METHODS Note that, 510 TC samples from The Cancer Genome Atlas database and 121 ERGs from the ExoBCD database were obtained. Differential gene expression analysis was performed to get ERGs in TC (TERGs). Functional enrichment analyses including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were conducted on the TERGs. Then we constructed a model based on LASSO Cox regression analysis. Kaplan-Meier survival analysis was applied and a Nomogram model was also built. The immune landscape was evaluated by CIBERSORT. RESULTS Thirty-eight TERGs were identified and their functions were enriched on 591 GO terms and 30 KEGG pathways. We built a Risk Score model based on FGFR3, ADRA1B, and POSTN. Risk Scores were significantly higher in T4 than in other stages, meanwhile, it didn't significantly differ in genders and TNM N or M classifications. The nomogram model could reliably predict the overall survival of TC patients. The mutation rate of BRAF and expression of cytotoxic T-lymphocyte-associated protein 4 were significantly higher in the high-risk group than in the low-risk group. The risk score was significantly correlated to the immune landscape. CONCLUSION We built a Risk Score model using FGFR3, ADRA1B, and POSTN which could reliably predict the prognosis of TC patients.
Collapse
Affiliation(s)
- Junfeng Qi
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Hanshan Cheng
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Long Su
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Jun Li
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Fei Cheng
- Department of Surgical Oncology, Wuwei People's Hospital, Wuwei, China
| |
Collapse
|
4
|
Chen XY, Lai JY, Shen WJ, Wang D, Wei ZX. Investigation of risk signatures associated with anoikis in thyroid cancer through integrated transcriptome and Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1458956. [PMID: 39568815 PMCID: PMC11576184 DOI: 10.3389/fendo.2024.1458956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Background Anoikis is intricately associated with the malignant progression of cancer. Thyroid cancer (THCA) is the most common endocrine tumor, metastasis is closely related to treatment response and prognosis of THCA. Hence, it is imperative to comprehensively identify predictive prognostic genes and novel molecular targets for effective THCA therapy. Methods Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were utilized to mine differentially expressed anoikis-related (DE-ARGs). Then, the prognostic genes were identified and a risk signature was constructed for THCA using univariate Cox analysis and least absolute shrinkage and selection operator (LASSO) method. Furthermore, the associations between risk signature and immune infiltration, immunotherapy, as well as potential mechanisms of action were determined using multiple R packages and Wilcoxon test. Finally, Mendelian randomized (MR) analysis was conducted to investigate the causal relationship between the prognostic genes and THCA. Results In total, six prognostic genes (LRRC75A, METTL7B, ADRA1B, TPD52L1, TNFRSF10C, and CXCL8) related to anoikis were identified, and the corresponding risk signature were constructed to assess the survival time of THCA patients. Immunocorrelation analysis demonstrated the anoikis-relevant risk signature could be used to evaluate immunotherapy effects in THCA patients, and the infiltration of immune cells was correlated with the degree of risk in THCA patients. According to two-sample MR analysis, there was the significant causal relationship between CXCL8 and THCA (odds ratio [OR] > 1 & p< 0.05), and the increase of its gene expression would lead to an increased risk of THCA. Furthermore, real-time quantitative polymerase chain reaction (RT-qPCR) confirmed the upregulated expression patterns of these prognostic genes in THCA tissues. Conclusion In conclusion, we constructed the risk signature related to anoikis for THCA, which might have important clinical significance for improving the quality of life and treatment effect of THCA patients.
Collapse
Affiliation(s)
- Xiang-Yi Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jia-Ying Lai
- Department of Nuclear Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wen-Jun Shen
- Department of Nuclear Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dawei Wang
- Department of Medical Engineering, Medical Supplies Center of PLA General Hospital, Beijing, China
- Department of Nuclear Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zhi-Xiao Wei
- Department of Nuclear Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
5
|
Björnebo L, Razdan S, Discacciati A, Palsdottir T, Aly M, Nordström T, Eklund M, Lundon D, Grönberg H, Tewari A, Wiklund P, Kyprianou N, Lantz A. Prostate cancer incidence and mortality in men exposed to α1-adrenergic receptor antagonists. J Natl Cancer Inst 2024; 116:1459-1465. [PMID: 38718219 PMCID: PMC11378311 DOI: 10.1093/jnci/djae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/26/2024] [Accepted: 05/06/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND α1-Adrenergic receptor antagonists are commonly used to treat benign prostatic hyperplasia. Preclinical studies suggest that they induce cell death and inhibit tumor growth. This study evaluated the risk of prostate cancer death in men using α1-adrenergic receptor antagonists. METHODS A population-based cohort study in Stockholm, Sweden (January 1, 2007, to December 31, 2019) included 451 779 men with a prostate-specific antigen test result. Study entry was 1 year after the first prostate-specific antigen test. Men were considered exposed at their second filled prescription. The primary outcome was prostate cancer mortality. Secondary outcomes were all-cause mortality and prostate cancer incidence. Cox proportional hazards regression models were used to calculate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for all outcomes. Inverse-probability weighting with marginal structural models accounted for time-dependent confounders. RESULTS Of 351 297 men in the final cohort, 39 856 (11.3%) were exposed to α1-adrenergic receptor antagonists. Median (interquartile range) follow-up for prostate cancer mortality was 8.9 (5.1-10.9) years; median (interquartile range) exposure time to α1-adrenergic receptor antagonists was 4.4 (2.0-7.6) years. There was no evidence of an association between α1-adrenergic receptor antagonist use and prostate cancer mortality, all-cause mortality, or high-grade prostate cancer. α1-Adrenergic receptor antagonist use was associated with an increased risk of prostate cancer (HR = 1.11, 95% CI = 1.06 to 1.17) and low-grade prostate cancer (HR = 1.22, 95% CI = 1.11 to 1.33). Men whose prostate cancer was treated with α1-adrenergic receptor antagonists underwent more frequent prostate-specific antigen testing. CONCLUSIONS Our findings show no significant association between α1-adrenergic receptor adrenoceptor antagonist exposure and prostate cancer mortality or high-grade prostate cancer. Although the preclinical evidence indicates a potential chemopreventive effect, this study's findings do not support it.
Collapse
Affiliation(s)
- Lars Björnebo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Shirin Razdan
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Discacciati
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Thorgerdur Palsdottir
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Markus Aly
- Department of Urology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tobias Nordström
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Dara Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ash Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Wiklund
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Lantz
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Urology, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
6
|
Zhang Z, Lv ZG, Lu M, Li H, Zhou J. Nerve-tumor crosstalk in tumor microenvironment: From tumor initiation and progression to clinical implications. Biochim Biophys Acta Rev Cancer 2024; 1879:189121. [PMID: 38796026 DOI: 10.1016/j.bbcan.2024.189121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/25/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024]
Abstract
The autonomic nerve system (ANS) innervates organs and tissues throughout the body and maintains functional balance among various systems. Further investigations have shown that excessive activation of ANS not only causes disruption of homeostasis, but also may promote tumor formation. In addition, the dynamic interaction between nerve and tumor cells in the tumor microenvironment also regulate tumor progression. On the one hand, nerves are passively invaded by tumor cells, that is, perineural invasion (PNI). On the other hand, compared with normal tissues, tumor tissues are subject to more abundant innervation, and nerves can influence tumor progression through regulating tumor proliferation, metastasis and drug resistance. A large number of studies have shown that nerve-tumor crosstalk, including PNI and innervation, is closely related to the prognosis of patients, and contributes to the formation of cancer pain, which significantly deteriorates the quality of life for patients. These findings suggest that nerve-tumor crosstalk represents a potential target for anti-tumor therapies and the management of cancer pain in the future. In this review, we systematically describe the mechanism by which nerve-tumor crosstalk regulates tumorigenesis and progression.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Surgery, School of Medicine, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Zhen Gang Lv
- Department of Surgery, School of Medicine, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Miao Lu
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Haifeng Li
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Jiahua Zhou
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu Province, China.
| |
Collapse
|
7
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
8
|
Abstract
Nerves not only regulate the homeostasis and energetic metabolism of normal epithelial cells but also are critical for cancer, as cancer recapitulates the biology of neural regulation of epithelial tissues. Cancer cells rarely develop in denervated organs, and denervation affects tumorigenesis, in vivo and in humans. Axonogenesis occurs to supply the new malignant epithelial growth with nerves. Neurogenesis happens later, first in ganglia around organs or the spinal column and subsequently through recruitment of neuroblasts from the central nervous system. The hallmark of this stage is regulation of homeostasis and energetic metabolism. Perineural invasion is the most efficient interaction between cancer cells and nerves. The hallmark of this stage is increased proliferation and decreased apoptosis. Finally, carcinoma cells transdifferentiate into a neuronal profile in search of neural independence. The latter is the last stage in neuroepithelial interactions. Treatments for cancer must address the biology of neural regulation of cancer.
Collapse
Affiliation(s)
- Gustavo Ayala
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston, McGovern School of Medicine, Houston, Texas, USA;
| |
Collapse
|
9
|
Leitão C, Matos B, Roque F, Herdeiro MT, Fardilha M. The Impact of Lifestyle on Prostate Cancer: A Road to the Discovery of New Biomarkers. J Clin Med 2022; 11:2925. [PMID: 35629050 PMCID: PMC9148038 DOI: 10.3390/jcm11102925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers among men, and its incidence has been rising through the years. Several risk factors have been associated with this disease and unhealthy lifestyles and inflammation were appointed as major contributors for PCa development, progression, and severity. Despite the advantages associated with the currently used diagnostic tools [prostate-specific antigen(PSA) serum levels and digital rectal examination (DRE)], the development of effective approaches for PCa diagnosis is still necessary. Finding lifestyle-associated proteins that may predict the development of PCa seems to be a promising strategy to improve PCa diagnosis. In this context, several biomarkers have been identified, including circulating biomarkers (CRP, insulin, C-peptide, TNFα-R2, adiponectin, IL-6, total PSA, free PSA, and p2PSA), urine biomarkers (PCA3, guanidine, phenylacetylglycine, and glycine), proteins expressed in exosomes (afamin, vitamin D-binding protein, and filamin A), and miRNAs expressed in prostate tissue (miRNA-21, miRNA-101, and miRNA-182). In conclusion, exploring the impact of lifestyle and inflammation on PCa development and progression may open doors to the identification of new biomarkers. The discovery of new PCa diagnostic biomarkers should contribute to reduce overdiagnosis and overtreatment.
Collapse
Affiliation(s)
- Catarina Leitão
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (C.L.); (M.T.H.)
| | - Bárbara Matos
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal;
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine—iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Fátima Roque
- Research Unit for Inland Development, Polytechnic of Guarda (UDI-IPG), Avenida Doutor Francisco Sá Carneiro, 6300-559 Guarda, Portugal;
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Maria Teresa Herdeiro
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (C.L.); (M.T.H.)
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine—iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
10
|
Zhong LK, Deng XY, Shen F, Cai WS, Feng JH, Gan XX, Jiang S, Liu CZ, Zhang MG, Deng JW, Zheng BX, Xie XZ, Ning LQ, Huang H, Chen SS, Miao JH, Xu B. Identification of a 3-Gene Prognostic Index for Papillary Thyroid Carcinoma. Front Mol Biosci 2022; 9:807931. [PMID: 35372518 PMCID: PMC8966665 DOI: 10.3389/fmolb.2022.807931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
The accurate determination of the risk of cancer recurrence is a critical unmet need in managing thyroid cancer (TC). Although numerous studies have successfully demonstrated the use of high throughput molecular diagnostics in TC prediction, it has not been successfully applied in routine clinical use, particularly in Chinese patients. In our study, we objective to screen for characteristic genes specific to PTC and establish an accurate model for diagnosis and prognostic evaluation of PTC. We screen the differentially expressed genes by Python 3.6 in The Cancer Genome Atlas (TCGA) database. We discovered a three-gene signature Gap junction protein beta 4 (GJB4), Ripply transcriptional repressor 3 (RIPPLY3), and Adrenoceptor alpha 1B (ADRA1B) that had a statistically significant difference. Then we used Gene Expression Omnibus (GEO) database to establish a diagnostic and prognostic model to verify the three-gene signature. For experimental validation, immunohistochemistry in tissue microarrays showed that thyroid samples’ proteins expressed by this three-gene are differentially expressed. Our protocol discovered a robust three-gene signature that can distinguish prognosis, which will have daily clinical application.
Collapse
Affiliation(s)
- Lin-Kun Zhong
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Xing-Yan Deng
- Thyroid, Vascular Surgery Department, Maoming People’s Hospital, Maoming, China
| | - Fei Shen
- Department of Thyroid Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wen-Song Cai
- Department of Thyroid Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jian-Hua Feng
- Department of Thyroid Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiao-Xiong Gan
- Department of Thyroid Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shan Jiang
- Reproductive Medicine Center, Boai Hsopital of Zhongshan, Zhongshan, China
| | - Chi-Zhuai Liu
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Ming-Guang Zhang
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Jiang-Wei Deng
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Bing-Xing Zheng
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Xiao-Zhang Xie
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Li-Qing Ning
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Hui Huang
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
| | - Shan-Shan Chen
- Department of Intensive Care Medicine, Zhongshan City People’s Hospital, Zhongshan, China
| | - Jian-Hang Miao
- Department of General Surgery, Zhongshan City People’s Hospital, Zhongshan, China
- *Correspondence: Jian-Hang Miao, ; Bo Xu, , https://orcid.org/0000-0001-6384-6685
| | - Bo Xu
- Thyroid, Vascular Surgery Department, Maoming People’s Hospital, Maoming, China
- *Correspondence: Jian-Hang Miao, ; Bo Xu, , https://orcid.org/0000-0001-6384-6685
| |
Collapse
|
11
|
Archer M, Dogra N, Dovey Z, Ganta T, Jang HS, Khusid JA, Lantz A, Mihalopoulos M, Stockert JA, Zahalka A, Björnebo L, Gaglani S, Noh MR, Kaplan SA, Mehrazin R, Badani KK, Wiklund P, Tsao K, Lundon DJ, Mohamed N, Lucien F, Padanilam B, Gupta M, Tewari AK, Kyprianou N. Role of α- and β-adrenergic signaling in phenotypic targeting: significance in benign and malignant urologic disease. Cell Commun Signal 2021; 19:78. [PMID: 34284799 PMCID: PMC8290582 DOI: 10.1186/s12964-021-00755-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 01/17/2023] Open
Abstract
The urinary tract is highly innervated by autonomic nerves which are essential in urinary tract development, the production of growth factors, and the control of homeostasis. These neural signals may become dysregulated in several genitourinary (GU) disease states, both benign and malignant. Accordingly, the autonomic nervous system is a therapeutic target for several genitourinary pathologies including cancer, voiding dysfunction, and obstructing nephrolithiasis. Adrenergic receptors (adrenoceptors) are G-Protein coupled-receptors that are distributed throughout the body. The major function of α1-adrenoceptors is signaling smooth muscle contractions through GPCR and intracellular calcium influx. Pharmacologic intervention of α-and β-adrenoceptors is routinely and successfully implemented in the treatment of benign urologic illnesses, through the use of α-adrenoceptor antagonists. Furthermore, cell-based evidence recently established the antitumor effect of α1-adrenoceptor antagonists in prostate, bladder and renal tumors by reducing neovascularity and impairing growth within the tumor microenvironment via regulation of the phenotypic epithelial-mesenchymal transition (EMT). There has been a significant focus on repurposing the routinely used, Food and Drug Administration-approved α1-adrenoceptor antagonists to inhibit GU tumor growth and angiogenesis in patients with advanced prostate, bladder, and renal cancer. In this review we discuss the current evidence on (a) the signaling events of the autonomic nervous system mediated by its cognate α- and β-adrenoceptors in regulating the phenotypic landscape (EMT) of genitourinary organs; and (b) the therapeutic significance of targeting this signaling pathway in benign and malignant urologic disease. Video abstract.
Collapse
Affiliation(s)
- M. Archer
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - N. Dogra
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Z. Dovey
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - T. Ganta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - H.-S. Jang
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - J. A. Khusid
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Lantz
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institute, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - M. Mihalopoulos
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - J. A. Stockert
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Zahalka
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - L. Björnebo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - S. Gaglani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. R. Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - S. A. Kaplan
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - R. Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. K. Badani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - P. Wiklund
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. Tsao
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - D. J. Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Mohamed
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - F. Lucien
- Department of Urology, Mayo Clinic, Rochester, MN USA
| | - B. Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. Gupta
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. K. Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
12
|
Enwald M, Lehtimäki T, Mishra PP, Mononen N, Murtola TJ, Raitoharju E. Human Prostate Tissue MicroRNAs and Their Predicted Target Pathways Linked to Prostate Cancer Risk Factors. Cancers (Basel) 2021; 13:cancers13143537. [PMID: 34298752 PMCID: PMC8307951 DOI: 10.3390/cancers13143537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 01/15/2023] Open
Abstract
MicroRNAs are important in prostate cancer development, progression and metastasis. The aim of this study was to test microRNA expression profile in prostate tissue obtained from prostate cancer patients for associations with various prostate cancer related factors and to pinpoint the predicted target pathways for these microRNAs. Prostate tissue samples were obtained at prostatectomy from patients participating in a trial evaluating impact of pre-operative atorvastatin on serum prostate specific antigen (PSA) and Ki-67 expression in prostate tissue. Prostate tissue microRNA expression profiles were analyzed using OpenArray® MicroRNA Panel. Pathway enrichment analyses were conducted for predicted target genes of microRNAs that correlated significantly with studied factors. Eight microRNAs correlated significantly with studied factors of patients after Bonferroni multiple testing correction. MiR-485-3p correlated with serum HDL-cholesterol levels. In atorvastatin-treated subjects, miR-34c-5p correlated with a change in serum PSA and miR-138-3p with a change in total cholesterol. In the placebo arm, both miR-576-3p and miR-550-3p correlated with HDL-cholesterol and miR-627 with PSA. In pathway analysis, these eight microRNAs related significantly to several pathways relevant to prostate cancer. This study brings new evidence from the expression of prostate tissue microRNAs and related pathways that may link risk factors to prostate cancer and pinpoint new therapeutic possibilities.
Collapse
Affiliation(s)
- Max Enwald
- Pirkanmaa Hospital District, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Terho Lehtimäki
- Pirkanmaa Hospital District, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Pashupati P Mishra
- Pirkanmaa Hospital District, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Nina Mononen
- Pirkanmaa Hospital District, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Teemu J Murtola
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- TAYS Cancer Center, Department of Urology, 33520 Tampere, Finland
| | - Emma Raitoharju
- Pirkanmaa Hospital District, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| |
Collapse
|
13
|
Sung SY, Le TTH, Chen JH, Hsieh TF, Hsieh CL. Alpha-1 blocker use increased risk of subsequent renal cell carcinoma: A nationwide population-based study in Taiwan. PLoS One 2020; 15:e0242429. [PMID: 33211759 PMCID: PMC7676733 DOI: 10.1371/journal.pone.0242429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/03/2020] [Indexed: 11/18/2022] Open
Abstract
Elevated Renal cell carcinoma (RCC) risk has been associated with the use of several antihypertensive medications but has not yet been elucidated in the populations prescribed alpha-1 blockers that are commonly used in the treatment of hypertension and lower urinary tract symptoms associated with benign prostatic hyperplasia (LUTS-BPH). The aim of the present study was to investigate the association between alpha-1 blocker use and the risk of developing RCC using a nationwide population-based database in Taiwan. Patients who were treated with alpha-1 blockers for at least 28 days were identified through the Taiwan National Health Insurance Research Database from 2000 to 2010. The unexposed participants were matched with the exposed cases according to age, sex, and index year at a ratio of 3:1. Cox proportional hazards regression, stratified by sex and comorbidities and adjusted for age, was performed to estimate hazard ratios (HRs) for the risk of subsequent RCC. Among 2,232,092 subjects, patients who received alpha-1 blocker treatment had a higher risk of RCC than the unexposed group. Taking into account hypertension and BPH, the adjusted HR was significantly higher in male alpha-1 blocker users who had no BPH and either the presence (HR: 1.63, 95% confidence interval [CI] = 1.22–2.18) or absence (HR: 2.31, 95% CI = 1.40–3.81) of hypertension than in men not receiving these drugs. Taken together, male alpha-1 blocker users who had no comorbidity of BPH exhibited an increased risk for developing RCC independent of hypertension. Further study is warranted to elucidate the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Shian-Ying Sung
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Trang Thi Huynh Le
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jin- Hua Chen
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
- Research Center of Biostatistics, College of Management, Taipei Medical University, Taipei, Taiwan
- Biostatistics Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Teng-Fu Hsieh
- Department of Urology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
14
|
Repurposing of α1-Adrenoceptor Antagonists: Impact in Renal Cancer. Cancers (Basel) 2020; 12:cancers12092442. [PMID: 32872127 PMCID: PMC7564811 DOI: 10.3390/cancers12092442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 11/17/2022] Open
Abstract
Renal cancer ranks twelfth in incidence among cancers worldwide. Despite improving outcomes due to better therapeutic options and strategies, prognosis for those with metastatic disease remains poor. Current systemic therapeutic approaches include inhibiting pathways of angiogenesis, immune checkpoint blockade, and mTOR inhibition, but inevitably resistance develops for those with metastatic disease, and novel treatment strategies are urgently needed. Emerging molecular and epidemiological evidence suggests that quinazoline-based α1-adrenoceptor-antagonists may have both chemopreventive and direct therapeutic actions in the treatment of urological cancers, including renal cancer. In human renal cancer cell models, quinazoline-based α1-adrenoceptor antagonists were shown to significantly reduce the invasion and metastatic potential of renal tumors by targeting focal adhesion survival signaling to induce anoikis. Mechanistically these drugs overcome anoikis resistance in tumor cells by targeting cell survival regulators AKT and FAK, disrupting integrin adhesion (α5β1 and α2β1) and engaging extracellular matrix (ECM)-associated tumor suppressors. In this review, we discuss the current evidence for the use of quinazoline-based α1-adrenoceptor antagonists as novel therapies for renal cell carcinoma (RCC) and highlight their potential therapeutic action through overcoming anoikis resistance of tumor epithelial and endothelial cells in metastatic RCC. These findings provide a platform for future studies that will retrospectively and prospectively test repurposing of quinazoline-based α1-adrenoceptor-antagonists for the treatment of advanced RCC and the prevention of metastasis in neoadjuvant, adjuvant, salvage and metastatic settings.
Collapse
|
15
|
Suzuki S, Yamamoto M, Sanomachi T, Togashi K, Sugai A, Seino S, Okada M, Yoshioka T, Kitanaka C. Doxazosin, a Classic Alpha 1-Adrenoceptor Antagonist, Overcomes Osimertinib Resistance in Cancer Cells via the Upregulation of Autophagy as Drug Repurposing. Biomedicines 2020; 8:biomedicines8080273. [PMID: 32764319 PMCID: PMC7460424 DOI: 10.3390/biomedicines8080273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 12/19/2022] Open
Abstract
Osimertinib, which is a third-generation epidermal growth factor receptor tyrosine kinase inhibitor, is an important anticancer drug because of its high efficacy and excellent safety profile. However, resistance against osimertinib is inevitable; therefore, therapeutic strategies to overcome the resistance are needed. Doxazosin, a classic quinazoline-based alpha 1-adrenoceptor antagonist is used to treat hypertension and benign prostatic hyperplasia with a known safety profile. The anticancer effects of doxazosin have been examined in various types of malignancies from the viewpoint of drug repositioning or repurposing. However, it currently remains unclear whether doxazosin sensitizes cancer cells to osimertinib. Herein, we demonstrated that doxazosin induced autophagy and enhanced the anticancer effects of osimertinib on the cancer cells and cancer stem cells of non-small cell lung cancer, pancreatic cancer, and glioblastoma at a concentration at which the growth of non-tumor cells was not affected. The osimertinib-sensitizing effects of doxazosin were suppressed by 3-methyladenine, an inhibitor of autophagy, which suggested that the effects of doxazosin were mediated by autophagy. The present study provides evidence for the efficacy of doxazosin as a combination therapy with osimertinib to overcome resistance against osimertinib.
Collapse
Affiliation(s)
- Shuhei Suzuki
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
- Department of Clinical Oncology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan;
- Correspondence: (S.S.); (M.Y.); Tel.: +81-23-628-5224 (S.S.); +81-23-628-5214 (M.Y.)
| | - Masahiro Yamamoto
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
- Correspondence: (S.S.); (M.Y.); Tel.: +81-23-628-5224 (S.S.); +81-23-628-5214 (M.Y.)
| | - Tomomi Sanomachi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
- Department of Clinical Oncology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan;
| | - Keita Togashi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
- Department of Ophthalmology and Visual Sciences, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | - Asuka Sugai
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
| | - Shizuka Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
| | - Masashi Okada
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
| | - Takashi Yoshioka
- Department of Clinical Oncology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan;
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (T.S.); (K.T.); (A.S.); (S.S); (M.O.); (C.K.)
- Research Institute for Promotion of Medical Sciences, Yamagata University Faculty of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| |
Collapse
|
16
|
Drug Repositioning of the α 1-Adrenergic Receptor Antagonist Naftopidil: A Potential New Anti-Cancer Drug? Int J Mol Sci 2020; 21:ijms21155339. [PMID: 32727149 PMCID: PMC7432507 DOI: 10.3390/ijms21155339] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/16/2022] Open
Abstract
Failure of conventional treatments is often observed in cancer management and this requires the development of alternative therapeutic strategies. However, new drug development is known to be a high-failure process because of the possibility of a lower efficacy than expected for the drug or appearance of non-manageable side effects. Another way to find alternative therapeutic drugs consists in identifying new applications for drugs already approved for a particular disease: a concept named "drug repurposing". In this context, several studies demonstrated the potential anti-tumour activity exerted by α1-adrenergic receptor antagonists and notably renewed interest for naftopidil as an anti-cancer drug. Naftopidil is used for benign prostatic hyperplasia management in Japan and a retrospective study brought out a reduced incidence of prostate cancer in patients that had been prescribed this drug. Further studies showed that naftopidil exerted anti-proliferative and cytotoxic effects on prostate cancer as well as several other cancer types in vitro, as well as ex vivo and in vivo. Moreover, naftopidil was demonstrated to modulate the expression of Bcl-2 family pro-apoptotic members which could be used to sensitise cancer cells to targeting therapies and to overcome resistance of cancer cells to apoptosis. For most of these anti-cancer effects, the molecular pathway is either not fully deciphered or shown to involve α1-adrenergic receptor-independent pathway, suggesting off target transduction signals. In order to improve its efficacy, naftopidil analogues were designed and shown to be effective in several studies. Thereby, naftopidil appears to display anti-cancer properties on different cancer types and could be considered as a candidate for drug repurposing although its anti-cancerous activities need to be studied more deeply in prospective randomized clinical trials.
Collapse
|
17
|
Goldberg H, Mohsin FK, Saskin R, Kulkarni GS, Berlin A, Kenk M, Wallis CJD, Klaassen Z, Chandrasekar T, Ahmad AE, Sayyid RK, Saarela O, Penn L, Alibhai SMH, Fleshner N. The Suggested Unique Association Between the Various Statin Subgroups and Prostate Cancer. Eur Urol Focus 2020; 7:537-545. [PMID: 32620539 DOI: 10.1016/j.euf.2020.06.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/25/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The chemopreventive effect of various medications in prostate cancer (PCa) has gained interest. Specifically, the potential impact of statins on PCa incidence has been studied, but solely as a "drug family" overlooking the distinctive pharmacological properties of its two main subgroups: hydrophilic and hydrophobic statins. OBJECTIVE To assess the impact of statin subgroups on PCa-specific mortality (PCSM), PCa diagnosis, and undergoing another prostate biopsy. DESIGN, SETTING, AND PARTICIPANTS This is a population-based cohort study in Ontario identifying all men aged ≥66 yr with a history of a single negative prostate biopsy (representing healthy men at risk for PCa) between 1994 and 2016, who were not on any of the analyzed medications prior to the study, with a median follow-up of 9.42 yr (interquartile range 8.03 yr). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Using multivariable cause-specific hazard models with time-dependent covariates, the association of hydrophobic and hydrophilic statins with all study outcomes was analyzed. Other putative chemopreventive medications (including alpha-blockers, 5-alpha-reductase inhibitors, and proton-pump inhibitors), age, rurality, comorbidities, and study inclusion year were included in the models. RESULTS AND LIMITATIONS Overall, 21 512 men were identified. Statins were taken by 11 401 patients (50.3%), 5184 men (24.1%) were diagnosed with PCa, and 805 (3.7%) died from it. Overall, 7556 patients (35.1%) underwent another biopsy. Any use of hydrophilic statins was associated with a 32.4% (95% confidence interval [CI] 12.9-47.5%), a 20% (95% CI 10-28%), and an 18% (95% CI 6.1-27.3%) decreased risk of PCSM, undergoing another prostate biopsy, and being diagnosed with PCa, respectively. Hydrophobic statins were associated with 17% (95% CI 2-31%) decreased PCSM. The study is limited by its retrospective nature, selection bias, and accompanying health-administrative database inaccuracies. CONCLUSIONS Use of any statin may be associated with a lower hazard of PCSM, with hydrophilic statins showing a greater association with decreased PCa diagnosis rates. Preferentially prescribing one statin subgroup over another in men needs further exploration. PATIENT SUMMARY Use of any statin may be associated with a lower probability of dying from prostate cancer. Hydrophilic statins (rosuvastatin and pravastatin) may also be more positively associated with a lower risk of undergoing an additional prostate biopsy and being diagnosed with prostate cancer in men aged ≥66 yr.
Collapse
Affiliation(s)
- Hanan Goldberg
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA; Institute for Clinical Evaluative Sciences, Toronto, ON, Canada.
| | - Faizan K Mohsin
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Refik Saskin
- Institute for Clinical Evaluative Sciences, Toronto, ON, Canada
| | - Girish S Kulkarni
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada; Institute for Clinical Evaluative Sciences, Toronto, ON, Canada
| | - Alejandro Berlin
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Techna Institute, University Health Network, Toronto, ON, Canada
| | - Miran Kenk
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada
| | - Christopher J D Wallis
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada; Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zachary Klaassen
- Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Augusta, GA, USA
| | - Thenappan Chandrasekar
- Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia PA, USA
| | - Ardalan E Ahmad
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Rashid K Sayyid
- Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Olli Saarela
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Linda Penn
- Department of Medical Biophysics, University of Toronto, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Shabbir M H Alibhai
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Medicine, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Neil Fleshner
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Hart J, Spencer B, McDermott CM, Chess-Williams R, Sellers D, Christie D, Anoopkumar-Dukie S. A Pilot retrospective analysis of alpha-blockers on recurrence in men with localised prostate cancer treated with radiotherapy. Sci Rep 2020; 10:8191. [PMID: 32424131 PMCID: PMC7235269 DOI: 10.1038/s41598-020-65238-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 04/29/2020] [Indexed: 12/17/2022] Open
Abstract
While alpha-blockers are commonly used to reduce lower urinary tract symptoms in prostate cancer patients receiving radiotherapy, their impact on response to radiotherapy remains unknown. Therefore, this pilot study aimed to retrospectively determine if alpha-blockers use, influenced response to radiotherapy for localised prostate cancer. In total, 303 prostate cancer patients were included, consisting of 84 control (alpha-blocker naïve), 72 tamsulosin and 147 prazosin patients. The main outcomes measured were relapse rates (%), time to biochemical relapse (months) and PSA velocity (ng/mL/year). Recurrence free survival was calculated using Kaplan-Meier analysis. Prazosin significantly reduced biochemical relapse at both two and five-years (2.72%, 8.84%) compared to control (22.61%, 34.52%). Recurrence free survival was also significantly higher in the prazosin group. This remained after multivariable analysis (HR: 0.09, 95% CI: 0.04-0.26, p < 0.001). Patients receiving prazosin had a 3.9 times lower relative risk of biochemical relapse compared to control. Although not statistically significant, tamsulosin and prazosin extended recurrence free survival by 13.15 and 9.21 months respectively. We show for the first time that prazosin may reduce risk of prostate cancer recurrence and delay time to biochemical relapse and provides justification for prospective studies to examine its potential as an adjunct treatment option for localised prostate cancer.
Collapse
Affiliation(s)
- Jordan Hart
- Menzies Health Institute, Griffith University, Queensland, Australia
- School of Pharmacy and Pharmacology, Griffith University, Queensland, Australia
- Quality Use of Medicines Network, Griffith University, Queensland, Australia
| | - Briohny Spencer
- Menzies Health Institute, Griffith University, Queensland, Australia
- School of Pharmacy and Pharmacology, Griffith University, Queensland, Australia
- Quality Use of Medicines Network, Griffith University, Queensland, Australia
| | - Catherine M McDermott
- Centre for Urology Research, Bond University, Gold Coast, Queensland, Australia
- Quality Use of Medicines Network, Griffith University, Queensland, Australia
| | - Russ Chess-Williams
- Centre for Urology Research, Bond University, Gold Coast, Queensland, Australia
- Quality Use of Medicines Network, Griffith University, Queensland, Australia
| | - Donna Sellers
- Centre for Urology Research, Bond University, Gold Coast, Queensland, Australia
- Quality Use of Medicines Network, Griffith University, Queensland, Australia
| | - David Christie
- School of Pharmacy and Pharmacology, Griffith University, Queensland, Australia
- Genesis Cancer Care, Gold Coast, Queensland, Australia
- Quality Use of Medicines Network, Griffith University, Queensland, Australia
| | - Shailendra Anoopkumar-Dukie
- Menzies Health Institute, Griffith University, Queensland, Australia.
- School of Pharmacy and Pharmacology, Griffith University, Queensland, Australia.
- Quality Use of Medicines Network, Griffith University, Queensland, Australia.
| |
Collapse
|
19
|
Wang T, Qin Y, Lai H, Wei W, Li Z, Yang Y, Huang M, Chen J. The prognostic value of ADRA1 subfamily genes in gastric carcinoma. Oncol Lett 2019; 18:3150-3158. [PMID: 31452791 PMCID: PMC6704286 DOI: 10.3892/ol.2019.10660] [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: 09/19/2018] [Accepted: 06/04/2019] [Indexed: 01/05/2023] Open
Abstract
Adrenergic receptor α1 (ADRA1) subfamily members, including ADRA1A, ADRA1B and ADRA1D, are understood to participate in cardiac disease and benign prostatic hyperplasia. In addition, adrenergic signals in cell pathways can promote the development of cancer. However, little is understood regarding the associations between ADRA1 subfamily members and gastric carcinoma (GC). The present study investigated the prognostic value of the ADRA1 subfamily genes in GC. Data from a total of 379 patients with GC were obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases. Kaplan-Meier analysis and Cox regression analysis were used to determine associations with overall survival (OS) and to evaluate the median survival time using hazard ratios (HRs) and 95% confidence intervals (CIs). Multivariate survival analysis revealed that low expression levels of ADRA1A (HR, 0.595; 95% CI, 0.426–0.831; adjusted P=0.002) ADRA1B (HR, 0.576; 95% CI, 0.412–0.805; adjusted P=0.001) and ADRA1D (HR, 0.559; 95% CI, 0.398–0.787; adjusted P=0.001) were associated with a favourable OS. Joint-effects analysis demonstrated that combinations of low expression levels of ARDA1A, ARDA1B and ARDA1D were significantly associated with a favourable OS. Overall, the current results suggested that the mRNA expression levels of ARDA1 subfamily members may serve as potential prognostic markers for GC.
Collapse
Affiliation(s)
- Tingan Wang
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yuzhou Qin
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hao Lai
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Weiyuan Wei
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhao Li
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yang Yang
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Mingwei Huang
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jiansi Chen
- Department of Gastrointestinal Surgery, Affiliated Tumour Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
20
|
Wade CA, Goodwin J, Preston D, Kyprianou N. Impact of α-adrenoceptor antagonists on prostate cancer development, progression and prevention. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2019; 7:46-60. [PMID: 30906804 PMCID: PMC6420703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 06/09/2023]
Abstract
Two decades following the discovery that α1-adrenoceptor antagonists suppress prostate tumor growth at the molecular and cellular level, the impact of α-blockade as re-purposed treatment strategy in the medical management of prostate cancer is gradually being recognized. Prostate cancer is the second most common cause of cancer deaths among males in the United States, yet the disease maintains inconsistent recommendations for prevention and screening. The functional relationship between α-adrenergic signaling and smooth muscle cells in the stroma of the prostate gland and the bladder neck empowered the use of α-adrenoceptor antagonists for the relief of urethral obstruction and clinical symptoms associated with benign prostatic hyperplasia (BPH). Adrenoceptors are G-protein-coupled receptors (GCPRs) that are functionally bound by catecholamines: epinephrine (ER) and norepinephrine (NE). The α1A adrenoceptor subtype is primarily responsible for smooth muscle contraction in the bladder neck and prostate gland. α1-adrenoceptor antagonists are clinically indicated as first-line therapies for the relief of BPH, hypertension, and post-traumatic stress disorder (PTSD). Compelling evidence from cellular and pre-clinical models have identified additional effects of α1-adrenoceptor antagonists regarding their ability to induce apoptosis-mediated suppression of prostate tumor growth and metastasis. Additionally, early epidemiologic data suggest that they may serve as a safe treatment to reduce the risk of prostate cancer. Optimization of quinazoline based compounds (doxazosin) to exploit pharmacologic targeting of tumor growth and vascularization revealed high efficacy of the lead novel compound DZ-50 against prostate tumors. This review discusses the experimental and pre-clinical evidence on the impact of α-blockade on prostate cancer.
Collapse
Affiliation(s)
- Cameron A Wade
- Department of Urology, University of Kentucky College of MedicineLexington, Kentucky 40536, USA
| | - Jeffrey Goodwin
- Department of Urology, University of Kentucky College of MedicineLexington, Kentucky 40536, USA
| | - David Preston
- Department of Urology, University of Kentucky College of MedicineLexington, Kentucky 40536, USA
| | - Natasha Kyprianou
- Department of Urology, University of Kentucky College of MedicineLexington, Kentucky 40536, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of MedicineLexington, Kentucky 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky College of MedicineLexington, Kentucky 40536, USA
| |
Collapse
|
21
|
Suzuki N, Niikura R, Ihara S, Hikiba Y, Kinoshita H, Higashishima N, Hayakawa Y, Yamada A, Hirata Y, Nakata R, Okamoto M, Sano M, Kushiyama A, Ichinose M, Woods SL, Worthley D, Iwamoto Y, Koike K. Alpha-Blockers As Colorectal Cancer Chemopreventive: Findings from a Case-Control Study, Human Cell Cultures, and In Vivo Preclinical Testing. Cancer Prev Res (Phila) 2019; 12:185-194. [PMID: 30700439 DOI: 10.1158/1940-6207.capr-18-0288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/17/2018] [Accepted: 01/18/2019] [Indexed: 11/16/2022]
Abstract
A retrospective case-controlled analysis was performed to identify drug candidates in the current use that may prevent colorectal cancer, outside of aspirin. A total of 37,510 patients aged ≥20 years were assessed to identify subjects who had been diagnosed with colorectal cancer by colonoscopy without a previous diagnosis of colorectal cancer, inflammatory bowel disease (IBD), or gastrointestinal symptoms; 1,560 patients were identified who were diagnosed with colorectal cancer by colonoscopy. The patients with colorectal cancer were matched with 1,560 age, gender, family history of colorectal cancer and comorbidity-matched control patients who were not diagnosed with colorectal cancer at colonoscopy. The medication histories were compared between the two groups. Next, candidate drugs that were more frequently used by the control patients were selected and their effects on human colorectal cancer cell lines in vitro and an inflammation-induced mouse model of colorectal cancer were tested. Putative colorectal cancer preventative agents were identified, including aspirin, vitamin D, vitamin B, vitamin C, vitamin E, xanthine oxidase inhibitor, alpha-blockers, angiotensin receptor blocker, nateglinide, probiotics, thienopyridine, folic acid, nitrovasodilators, bisphosphonates, calcium channel blockers, steroids, and statins (P < 0.05). Alpha-blockers and xanthine oxidase inhibitors were selected for further study because these agents have not been analyzed previously as factors that may affect colorectal cancer outcomes. In vitro doxazosin (alpha-blocker), but not febuxostat (xanthine oxidase inhibitor), suppressed the proliferation of human colorectal cancer cells. Doxazosin also decreased tumorigenesis in an AOM/DSS mouse colorectal cancer model. Alpha-blockers may prevent colorectal cancer.
Collapse
Affiliation(s)
- Nobumi Suzuki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. .,Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan.,School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Ryota Niikura
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sozaburo Ihara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Yohko Hikiba
- Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Hiroto Kinoshita
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Naoko Higashishima
- Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsuo Yamada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Hirata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryo Nakata
- Department of Gastroenterology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Makoto Okamoto
- Department of Gastroenterology, JR Tokyo General Hospital, Tokyo, Japan
| | - Munetaka Sano
- Department of Gastroenterology, Yaizu City Hospital, Shizuoka, Japan
| | - Akifumi Kushiyama
- Division of Diabetes and Metabolism, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Mari Ichinose
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Susan L Woods
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Daniel Worthley
- School of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Yasuhiko Iwamoto
- Division of Diabetes and Metabolism, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Gao X, Abdelkarim H, Albee LJ, Volkman BF, Gaponenko V, Majetschak M. Partial agonist activity of α1-adrenergic receptor antagonists for chemokine (C-X-C motif) receptor 4 and atypical chemokine receptor 3. PLoS One 2018; 13:e0204041. [PMID: 30248140 PMCID: PMC6152952 DOI: 10.1371/journal.pone.0204041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/01/2018] [Indexed: 12/15/2022] Open
Abstract
We observed in PRESTO-Tango β-arrestin recruitment assays that the α1-adrenergic receptor (AR) antagonist prazosin activates chemokine (C-X-C motif) receptor (CXCR)4. This prompted us to further examine this unexpected pharmacological behavior. We screened a panel of 14 α1/2- and β1/2/3-AR antagonists for CXCR4 and atypical chemokine receptor (ACKR)3 agonist activity in PRESTO-Tango assays against the cognate agonist CXCL12. We observed that multiple α1-AR antagonists activate CXCR4 (CXCL12 = prazosin = cyclazosin > doxazosin) and ACKR3 (CXCL12 = prazosin = cyclazosin > alfuzosin = doxazosin = phentolamine > terazosin = silodosin = tamsulosin). The two strongest CXCR4/ACKR3 activators, prazosin and cyclazosin, were selected for a more detailed evaluation. We found that the drugs dose-dependently activate both receptors in β-arrestin recruitment assays, stimulate ERK1/2 phosphorylation in HEK293 cells overexpressing each receptor, and that their effects on CXCR4 could be inhibited with AMD3100. Both α1-AR antagonists induced significant chemical shift changes in the 1H-13C-heteronuclear single quantum correlation spectrum of CXCR4 and ACKR3 in membranes, suggesting receptor binding. Furthermore, prazosin and cyclazosin induced internalization of endogenous CXCR4/ACKR3 in human vascular smooth muscle cells (hVSMC). While these drugs did not in induce chemotaxis in hVSMC, they inhibited CXCL12-induced chemotaxis with high efficacy and potency (IC50: prazosin—4.5 nM, cyclazosin 11.6 pM). Our findings reveal unexpected pharmacological properties of prazosin, cyclazosin, and likely other α1-AR antagonists. The results of the present study imply that prazosin and cyclazosin are biased or partial CXCR4/ACKR3 agonists, which function as potent CXCL12 antagonists. Our findings could provide a mechanistic basis for previously observed anti-cancer properties of α1-AR antagonists and support the concept that prazosin could be re-purposed for the treatment of disease processes in which CXCR4 and ACKR3 are thought to play significant pathophysiological roles, such as cancer metastases or various autoimmune pathologies.
Collapse
Affiliation(s)
- Xianlong Gao
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Hazem Abdelkarim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Lauren J. Albee
- Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States of America
| | - Brian F. Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Matthias Majetschak
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
23
|
Cao L, Zhang S, Jia CM, He W, Wu LT, Li YQ, Wang W, Li Z, Ma J. Antihypertensive drugs use and the risk of prostate cancer: a meta-analysis of 21 observational studies. BMC Urol 2018. [PMID: 29514670 PMCID: PMC5842557 DOI: 10.1186/s12894-018-0318-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Due to the lack of strong evidence to identify the relationship between antihypertensive drugs use and the risk of prostate cancer, it was needed to do a systematic review to go into the subject. Methods We systematically searched PubMed, Web of Science and Embase to identify studies published, through May 2015. Two evaluators independently reviewed and selected articles involving the subject. We used the Newcastle-Ottawa Scale (NOS) to assess the quality of the studies. All extracted results to evaluate the relationship between antihypertensive drugs usage and prostate cancer risk were pool-analysed using Stata 12.0 software. Results A total of 12 cohort and 9 case-control studies were ultimately included in our review. Most of the studies were evaluated to be of high quality. There was no significant relationship between angiotensin converting enzyme inhibitors (ACEI) usage and the risk of prostate cancer (RR 1.07, 95% CI 0.96–1.20), according to the total pool-analysed. Use of angiotensin receptor blocker (ARB) was not associated with the risk of prostate cancer (RR 1.09, 95% CI 0.97–1.21), while use of CCB may well increase prostate cancer risk based on the total pool-analysed (RR 1.08, 95% CI 1–1.16). Moreover, subgroup analysis suggested that use of CCB clearly increased prostate cancer risk (RR 1.10, 95% CI 1.04–1.16) in terms of case-control studies. There was also no significant relationship between use of diuretic (RR 1.09, 95% CI 0.95–1.25) or antiadrenergic agents (RR 1.22, 95% CI 0.76–1.96) and prostate cancer risk. Conclusions There is no significant relationship between the use of antihypertensive drugs (ACEI, ARB, beta-blockers and diuretics) and prostate cancer risk, but CCB may well increase prostate cancer risk, according to existing observational studies. Electronic supplementary material The online version of this article (10.1186/s12894-018-0318-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liang Cao
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Sha Zhang
- Shanxi University of Chinese Medicine, Xian yang, 712046, People's Republic of China
| | - Cheng-Ming Jia
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Wei He
- Shanxi University of Chinese Medicine, Xian yang, 712046, People's Republic of China
| | - Lei-Tao Wu
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Ying-Qi Li
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Wen Wang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Zhe Li
- Shanxi University of Chinese Medicine, Xian yang, 712046, People's Republic of China.
| | - Jing Ma
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| |
Collapse
|
24
|
Ishii K, Matsuoka I, Kajiwara S, Sasaki T, Miki M, Kato M, Kanda H, Arima K, Shiraishi T, Sugimura Y. Additive naftopidil treatment synergizes docetaxel-induced apoptosis in human prostate cancer cells. J Cancer Res Clin Oncol 2018; 144:89-98. [PMID: 29098395 DOI: 10.1007/s00432-017-2536-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 10/24/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE Docetaxel (DTX) is a standard chemotherapeutic drug for castration-resistant prostate cancer (CRPC), although adverse events are common. To overcome this problem, researchers have evaluated the efficacy of DTX treatment in combination with other drugs. Naftopidil is a tubulin-binding drug with fewer adverse events, implying the usefulness of this drug in clinical applications when combined with DTX. Here, we investigated the efficacy of additive naftopidil treatment in combination with DTX on prostate cancer (PCa) cells. METHODS The effects of combination treatment with DTX plus naftopidil were analyzed using two animal models of LNCaP cells plus PrSC xenografts (sub-renal capsule grafting) and PC-3 xenografts (intratibial injection). RESULTS Combination treatment with DTX plus naftopidil significantly inhibited cell growth in LNCaP cells compared with DTX alone. Analysis of the cooperativity index (CI) showed that combination treatment exhibited additive effects on DTX-induced growth inhibition in LNCaP cells. In contrast, combination treatment showed more than an additive (synergistic) effect on DTX-induced apoptosis in LNCaP and PC-3 cells. In LNCaP cells plus PrSC xenografts, combination treatment showed synergistic effects on DTX-induced apoptosis. The synergistic effects of naftopidil on DTX-induced apoptosis were also observed in PC-3 xenografts. CONCLUSIONS Our results demonstrated that additive naftopidil treatment in combination with DTX increased the efficacy of DTX for the treatment of LNCaP and PC-3 tumors in vivo. Thus, additive naftopidil treatment showed a synergistic effect on DTX-induced apoptosis in PCa cells in vitro and in vivo, suggesting that this treatment approach may yield improved clinical benefits compared with DTX alone.
Collapse
Affiliation(s)
- Kenichiro Ishii
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Izumi Matsuoka
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Shinya Kajiwara
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takeshi Sasaki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Manabu Miki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Manabu Kato
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hideki Kanda
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Kiminobu Arima
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Taizo Shiraishi
- Department of Diagnostic Pathology, Kuwana City Medical Center, Kuwana, Mie, Japan
| | - Yoshiki Sugimura
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
25
|
Huan HB, Wen XD, Chen XJ, Wu L, Wu LL, Zhang L, Yang DP, Zhang X, Bie P, Qian C, Xia F. Sympathetic nervous system promotes hepatocarcinogenesis by modulating inflammation through activation of alpha1-adrenergic receptors of Kupffer cells. Brain Behav Immun 2017; 59:118-134. [PMID: 27585737 DOI: 10.1016/j.bbi.2016.08.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/23/2016] [Accepted: 08/27/2016] [Indexed: 02/07/2023] Open
Abstract
The sympathetic nervous system (SNS) is known to play a significant role in tumor initiation and metastasis. Hepatocellular carcinoma (HCC) frequently occurs in cirrhotic livers after chronic inflammation, and the SNS is hyperactive in advanced liver cirrhosis. However, it remains unclear whether the SNS promotes hepatocarcinogenesis by modulating chronic liver inflammation. In this study, a retrospective pathological analysis and quantification of sympathetic nerve fiber densities (tyrosine hydroxylase, TH+) in HCC patients, and diethylnitrosamine (DEN)-induced hepatocarcinogenesis in rats were performed. Our data showed that high density of sympathetic nerve fibers and α1-adrenergic receptors (ARs) of Kupffer cells (KCs) were associated with a poor prognosis of HCC. Sympathetic denervation or blocking of α1-ARs decreased DEN-induced HCC incidence and tumor development. In addition, synergistic effects of interleukin-6 (IL-6) and transforming growth factor-beta (TGF-β) in hepatocarcinogenesis were observed. The suppression of the SNS reduced IL-6 and TGF-β expression, which suppressed hepatocarcinogenesis, and KCs play a key role in this process. After the ablation of KCs, IL-6 and TGF-β expression and the development of HCC were inhibited. This study demonstrates that sympathetic innervation is crucial for hepatocarcinogenesis and that the SNS promotes hepatocarcinogenesis by activating α1-ARs of KCs to boost the activation of KCs and to maintain the inflammatory microenvironment. These results indicate that sympathetic denervation or α1-ARs blockage may represent novel treatment approaches for HCC.
Collapse
Affiliation(s)
- Hong-Bo Huan
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xu-Dong Wen
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xue-Jiao Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunology and Pathology of Ministry of Education China, Chongqing, China
| | - Lin Wu
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Li-Li Wu
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Liang Zhang
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Da-Peng Yang
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunology and Pathology of Ministry of Education China, Chongqing, China
| | - Ping Bie
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunology and Pathology of Ministry of Education China, Chongqing, China
| | - Feng Xia
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
26
|
The Role of α1-Adrenoceptor Antagonists in the Treatment of Prostate and Other Cancers. Int J Mol Sci 2016; 17:ijms17081339. [PMID: 27537875 PMCID: PMC5000736 DOI: 10.3390/ijms17081339] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022] Open
Abstract
This review evaluates the role of α-adrenoceptor antagonists as a potential treatment of prostate cancer (PCa). Cochrane, Google Scholar and Pubmed were accessed to retrieve sixty-two articles for analysis. In vitro studies demonstrate that doxazosin, prazosin and terazosin (quinazoline α-antagonists) induce apoptosis, decrease cell growth, and proliferation in PC-3, LNCaP and DU-145 cell lines. Similarly, the piperazine based naftopidil induced cell cycle arrest and death in LNCaP-E9 cell lines. In contrast, sulphonamide based tamsulosin did not exhibit these effects. In vivo data was consistent with in vitro findings as the quinazoline based α-antagonists prevented angiogenesis and decreased tumour mass in mice models of PCa. Mechanistically the cytotoxic and antitumor effects of the α-antagonists appear largely independent of α 1-blockade. The proposed targets include: VEGF, EGFR, HER2/Neu, caspase 8/3, topoisomerase 1 and other mitochondrial apoptotic inducing factors. These cytotoxic effects could not be evaluated in human studies as prospective trial data is lacking. However, retrospective studies show a decreased incidence of PCa in males exposed to α-antagonists. As human data evaluating the use of α-antagonists as treatments are lacking; well designed, prospective clinical trials are needed to conclusively demonstrate the anticancer properties of quinazoline based α-antagonists in PCa and other cancers.
Collapse
|
27
|
Forbes A, Anoopkumar-Dukie S, Chess-Williams R, McDermott C. Relative cytotoxic potencies and cell death mechanisms of α1 -adrenoceptor antagonists in prostate cancer cell lines. Prostate 2016; 76:757-66. [PMID: 26880388 DOI: 10.1002/pros.23167] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 01/27/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Some α1 -adrenoceptor antagonists possess anti-cancer actions that are independent of α1 -adrenoceptors and the aim of these studies was to assess the relative cytotoxic potencies of α1 -adrenoceptor antagonists and the mechanisms involved in these actions. METHODS PC-3 and LNCap human prostate cancer cells were exposed to α1 -adrenoceptor antagonists (0.01-100 μM) and cell survival assessed after 24-72 hr. The levels of apoptosis, autophagy and stress related proteins were also determined. RESULTS The relative cytotoxic potency order was prazosin = doxazosin > terazosin = silodosin = alfuzosin > tamsulosin on both cell types, but LNCaP cells were significantly more sensitive to these effects than PC-3 cells. Prazosin and doxazosin increased levels of apoptotsis and autophagy in both cell lines, and activated EphA2 receptors in PC-3 cells. Autophagy contributed to survival of LNCaP, but promoted cell death in PC-3 cells. Treatment with prazosin (30 μM) altered the expression of several cell stress-related proteins: elevating phospho-p38α and reducing S6 kinase in both cell lines. Surprisingly some proteins were differentially affected in the two prostate cancer cell lines: Akt and p27 increasing and HIF-1α decreasing in LNCap cells but not PC-3, while ADAMTS1 was increased in PC-3 cells only. CONCLUSIONS Prazosin and doxazosin demonstrated cytotoxic actions on both castration-resistant PC-3 and androgen-sensitive LNCap prostate cancer cells. The mechanisms involved included changes in a number of proliferation and apoptosis regulatory proteins. The role of autophagy depended on the cell type, but contributed to cell death in PC3 cells.
Collapse
Affiliation(s)
- Amanda Forbes
- Centre for Urology Research, Faculty of Health Sciences and Medicine, Bond University, Queensland, Australia
| | - Shailendra Anoopkumar-Dukie
- Menzies Health Institute Queensland, Griffith University, Queensland Australia School of Pharmacy, Griffith University, Queensland, Australia
| | - Russ Chess-Williams
- Centre for Urology Research, Faculty of Health Sciences and Medicine, Bond University, Queensland, Australia
| | - Catherine McDermott
- Centre for Urology Research, Faculty of Health Sciences and Medicine, Bond University, Queensland, Australia
| |
Collapse
|
28
|
Hoegestoel EA, Berg-Johnsen J. Regression of Intracranial Meningioma during Treatment with α1-Adrenoceptor Blocker. J Neurol Surg Rep 2016; 77:e62-5. [PMID: 27175325 PMCID: PMC4863873 DOI: 10.1055/s-0035-1571204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Regression of meningioma has been reported after hemorrhage or hormonal withdrawal. Here, we report a case of an incidentally diagnosed meningioma that regressed in association with α1-adrenoceptor antagonist. Case report A 59-year old male patient with an incidentally diagnosed lateral sphenoid wing meningioma was followed with serial magnetic resonance imaging. The tumor with a maximum diameter of 43 mm showed progressive regression, and after 3 years the size was reduced to 22% of the initial volume. During follow-up the patient was treated with an α1-adrenoceptor antagonist (tamsulosin) for benign prostatic hyperplasia. Possible mechanisms are discussed, including our main hypothesis of reduced mitogenic effects through phospholipase C-signal transduction. Conclusion This is the first report of regression of an incidentally diagnosed meningioma associated with α1-adrenoceptor antagonist treatment.
Collapse
Affiliation(s)
| | - Jon Berg-Johnsen
- Department of Neurosurgery, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
29
|
Delgado-González E, Sánchez-Tusie AA, Morales G, Aceves C, Anguiano B. Triiodothyronine Attenuates Prostate Cancer Progression Mediated by β-Adrenergic Stimulation. Mol Med 2016; 22:1-11. [PMID: 26928389 DOI: 10.2119/molmed.2015.00047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 02/19/2016] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer cells are responsive to adrenergic and thyroid stimuli. It is well established that β-adrenergic activation (protein kinase A [PKA]/cAMP response element binding protein [CREB]) promotes cancer progression, but the role of thyroid hormones is poorly understood. We analyzed the effects of β-adrenergic stimulation (isoproterenol [ISO]) and/or thyroid hormone on neuroendocrine (NE) differentiation and cell invasion, using in vivo (LNCaP tumor) and in vitro models (LNCaP and DU145 human cells). Nude mice were inoculated with LNCaP cells and were treated for 6 wks with ISO (200 μg/d), triiodothyronine (T3, 2.5 μg/d) or both. ISO alone reduced tumor growth but increased tumor expression of cAMP response element (CRE)-dependent genes (real-time polymerase chain reaction, chromogranin A, neuron-specific enolase, survivin, vascular endothelial growth factor [VEGF], urokinase plasmin activator [uPA] and metalloproteinase-9 [MMP-9]) and some proteins related to NE differentiation and/or invasiveness (synaptophysin, VEGF, pCREB). T3 reduced tumor growth and prevented the overexpression of ISO-stimulated factors through a pCREB-independent mechanism. In low invasive LNCaP cells, 50 μmol/L ISO or 100 nmol/L thyroxine (T4) induced the acquisition of NE-like morphology (phase-contrast microscopy), increased VEGF secretion (ELISA) and invasive capacity (Transwell assay), but no synergistic effects were observed after the coadministration of ISO + T4. In contrast, 10 nmol/L T3 alone had no effect, but it prevented the NE-like morphology and invasiveness stimulated by ISO. None of these treatments had any effect on highly invasive DU145 cells. In summary, this study showed that ISO and T4 increase cancer progression, and T3 attenuates ISO-stimulated progression. Further studies are required to determine if changes in the ratio of T4/T3 could be relevant for prostate cancer progression.
Collapse
Affiliation(s)
- Evangelina Delgado-González
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Ana Alicia Sánchez-Tusie
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Giapsy Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Carmen Aceves
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - Brenda Anguiano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| |
Collapse
|
30
|
Colciago A, Mornati O, Ferri N, Castelnovo LF, Fumagalli L, Bolchi C, Pallavicini M, Valoti E, Negri-Cesi P. A selective alpha1D-adrenoreceptor antagonist inhibits human prostate cancer cell proliferation and motility “in vitro”. Pharmacol Res 2016; 103:215-26. [DOI: 10.1016/j.phrs.2015.11.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/26/2015] [Accepted: 11/19/2015] [Indexed: 01/09/2023]
|
31
|
Patil KC, McPherson L, Daly CJ. Co-Localization of Alpha1-Adrenoceptors and GPR55: A Novel Prostate Cancer Paradigm? ACTA ACUST UNITED AC 2015. [DOI: 10.4236/pp.2015.64023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
32
|
Zhang MM, Lu L, Wang XS. A Selective Method for the Synthesis ofN,N’-diarylbenzene-1,4-diamine and Dispirocyclic Quinazolinone Derivatives Catalyzed by Iodine. J Heterocycl Chem 2014. [DOI: 10.1002/jhet.1826] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Mei-Mei Zhang
- The Key Laboratory of Biotechnology on Medical Plant of Jiangsu Province; Jiangsu Normal University; Xuzhou Jiangsu 221116 People's Republic of China
| | - Lian Lu
- School of Chemistry and Chemical Engineering, Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials; Jiangsu Normal University; Xuzhou Jiangsu 221116 People's Republic of China
| | - Xiang-Shan Wang
- The Key Laboratory of Biotechnology on Medical Plant of Jiangsu Province; Jiangsu Normal University; Xuzhou Jiangsu 221116 People's Republic of China
- School of Chemistry and Chemical Engineering, Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials; Jiangsu Normal University; Xuzhou Jiangsu 221116 People's Republic of China
| |
Collapse
|
33
|
Bilbro J, Mart M, Kyprianou N. Therapeutic value of quinazoline-based compounds in prostate cancer. Anticancer Res 2013; 33:4695-4700. [PMID: 24222103 PMCID: PMC5451155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Certain α1-adrenoreceptor antagonists induce significant apoptosis and impair tumor vascularity without affecting cellular proliferation, effects specific to the quinazoline structure. These anticancer effects have been attributed to both induction of classical apoptosis and reversal of anoikis resistance via disruption of integrin-mediated cell survival pathways. Recent drug optimization efforts have generated several novel compounds with quinazoline-derived chemical structure that exert potent anti-tumor activity via anoikis. Results from pre-clinical and clinical studies implicate a potential value of quinazoline-based analogues in prostate cancer prevention and therapy. A retrospective study of a large patient cohort at our center, revealed that treatment with α1-andrenoreceptor antagonists significantly reduced the risk of developing prostate cancer, indicating a potential chemopreventative mechanism for these FDA-approved agents. In the present review we discuss the current understanding of the signaling mechanisms reversing anoikis resistance by the quinazoline-based compounds in prostate tumors, towards enabling identification of novel therapeutic targets for the treatment of metastatic castration-resistant prostate cancer (CRPC).
Collapse
Affiliation(s)
- Johnathan Bilbro
- Combs Res. Bldg., Rm. 306, University of Kentucky Medical Center, Lexington, KY 40536, U.S.A.
| | | | | |
Collapse
|
34
|
Ventura S, Evans BA. Does the autonomic nervous system contribute to the initiation and progression of prostate cancer? Asian J Androl 2013; 15:715-6. [PMID: 24141535 PMCID: PMC3854038 DOI: 10.1038/aja.2013.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In the July 12 issue of Science magazine, researchers from the Albert Einstein College of Medicine, the Mount Sinai School of Medicine, the Durham VA Medical Centre and Duke University published an elegant study demonstrating that the sympathetic nervous system, acting through β2 and β3-adrenoceptors in the prostate, plays an important role in the initiation of prostate cancer, while the parasympathetic nervous system plays a role in the dissemination of tumour metastases via M1 muscarinic receptors. These findings are significant because they indicate that receptors associated with the autonomic nervous system may be viable targets for prostate cancer therapy.
Collapse
|
35
|
Iwamoto Y, Ishii K, Sasaki T, Kato M, Kanda H, Yamada Y, Arima K, Shiraishi T, Sugimura Y. Oral naftopidil suppresses human renal-cell carcinoma by inducing G(1) cell-cycle arrest in tumor and vascular endothelial cells. Cancer Prev Res (Phila) 2013; 6:1000-6. [PMID: 23901046 DOI: 10.1158/1940-6207.capr-13-0095] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Renal cell carcinoma (RCC) is an angiogenesis-dependent and hypoxia-driven malignancy. As a result, several targeting agents are being investigated. However, the efficacy of current regimens is generally insufficient for their toxicity and poor overall response rates. We have recently reported that naftopidil exerts growth-inhibitory effects on human prostate cancer cells. In this study, we investigated the biochemical mechanisms by which naftopidil produces growth-inhibitory and antiangiogenic effects on RCC. We first tested the effects of naftopidil on the proliferation of ACHN and Caki-2 RCC cells. Next, we set up a model simulating the tumor microenvironment, in which ACHN cells were grafted onto the renal capsule of mice. We then tested the effects of naftopidil on human umbilical vein endothelial cells' cell proliferation and Matrigel plug vascularization. Finally, to establish the antitumor activity of naftopidil on RCC, we tested the antitumor effects of naftopidil on excised tumor specimens from 20 patients with RCC that were grafted beneath the renal capsule of mice. Naftopidil showed similar in vitro growth-inhibitory effects on all cell lines. Fluorescence-activated cell sorting analysis revealed an increase in G1 cell-cycle arrest in all naftopidil-treated cell lines. In vivo tumorigenic studies showed a significant reduction of ACHN tumor weight, Ki-67 index, and microvessel density (MVD) in naftopidil-treated mice. Naftopidil attenuated neovascularization in an in vivo Matrigel plug assay. Studies in mouse xenograft models also showed a significant MVD reduction in naftopidil-treated excised human RCC. The growth-inhibitory effects of naftopidil suggest it may be a novel anticancer agent and a potential preventive option for RCC.
Collapse
Affiliation(s)
- Yoichi Iwamoto
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Mie, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hendrix LN, Hamilton DA, Kyprianou N. Emerging therapeutics targeting castration-resistant prostate cancer: the AR-mageddon of tumor epithelial-mesenchymal transition. Expert Rev Endocrinol Metab 2013; 8:403-416. [PMID: 30736155 DOI: 10.1586/17446651.2013.811914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Advanced prostate cancer will claim nearly 30,000 lives among men in the USA in the year 2013. Most of these will be castration-resistant prostate cancers that are not responsive to traditional therapeutic modalities, and there is no available regimen that fully eradicates metastatic disease. This poses a significant clinical challenge for practitioners and has stimulated the development of novel agents that target these castration-resistant tumor cells. Development of metastatic prostate cancer is orchestrated by multiple signaling pathways that regulate cell survival, apoptosis, anoikis, epithelial-mesenchymal transition (EMT), invasion, the androgen signaling axis and angiogenesis. Disruption of the mechanisms underlying these processes is critical for development of agents that can target otherwise resistant tumor cells. Insights into the mechanisms by which rounds of EMT/mesenchymal-epithelial transition conversions facilitate the progression of localized prostate carcinomas to advanced metastatic and castration-resistant disease emerge as attractive targets for drug development. In this review, the authors discuss the current understanding of therapeutic resistance in castration-resistant prostate cancer with focus on the androgen receptor signaling axis and EMT. Novel therapeutic approaches targeting critical players of both pathways as well as the results from ongoing clinical trials will be discussed in this review.
Collapse
Affiliation(s)
- Lauren N Hendrix
- a Division of Urology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - David A Hamilton
- a Division of Urology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Natasha Kyprianou
- b Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
- c Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA
- d Division of Urology, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
37
|
Murtola TJ, Kujala PM, Tammela TLJ. High-grade prostate cancer and biochemical recurrence after radical prostatectomy among men using 5α-reductase inhibitors and alpha-blockers. Prostate 2013; 73:923-31. [PMID: 23334943 DOI: 10.1002/pros.22638] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 12/07/2012] [Indexed: 11/09/2022]
Abstract
BACKGROUND Two clinical trials have shown that users of 5α-reductase inhibitors finasteride and dutasteride (5-ARIs) have reduced overall prostate cancer risk, while the proportion of high-grade tumors is increased. We studied tumor characteristics, risk of biochemical recurrence and mortality after radical prostatectomy in 5-ARI and alpha-blocker users. METHODS The study cohort consisted of 1,315 men who underwent radical prostatectomy at the Tampere University Hospital during 1995-2009. Biochemical relapse was defined as serum PSA ≥ 0.2 ng/ml after the operation. Information on mortality and medication purchases was obtained from national registries. Cox proportional regression was used to analyze hazard ratios (HRs) and 95% confidence intervals (95% CI) of biochemical relapse and death. RESULTS The proportion of high-grade (Gleason 7-10) tumors was significantly elevated among men who had used 5-ARIs for 4 years or longer compared to the non-users (83.3% vs. 53.3%, respectively). Survival curves for biochemical relapse-free survival differed between long-term and short-term 5-ARI users, but the hazard ratio remained statistically non-significant. Risk of biochemical recurrence was elevated among alpha-blocker users (HR 1.68, 95% CI 1.37-2.06), but in sensitivity analyses this was evident only in men using alpha-blockers after prostatectomy. Mortality was not associated with medication usage. CONCLUSIONS Long-term users of finasteride or dutasteride had more often high-grade prostate cancer. Our results suggest also worse progression-free survival. The association between risk of biochemical recurrence and post-operative alpha-blocker usage suggests that voiding or storage symptoms after prostatectomy may predict biochemical relapse.
Collapse
Affiliation(s)
- Teemu J Murtola
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
| | | | | |
Collapse
|
38
|
White CW, Xie JH, Ventura S. Age-related changes in the innervation of the prostate gland: implications for prostate cancer initiation and progression. Organogenesis 2013; 9:206-15. [PMID: 23872639 PMCID: PMC3896592 DOI: 10.4161/org.24843] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The adult prostate gland grows and develops under hormonal control while its physiological functions are controlled by the autonomic nervous system. The prostate gland receives sympathetic input via the hypogastric nerve and parasympathetic input via the pelvic nerve. In addition, the hypogastric and pelvic nerves also provide sensory inputs to the gland. This review provides a summary of the innervation of the adult prostate gland and describes the changes which occur with age and disease. Growth and development of the prostate gland is age dependent as is the occurrence of both benign prostate disease and prostate cancer. In parallel, the activity and influence of both the sympathetic and parasympathetic nervous system changes with age. The influence of the sympathetic nervous system on benign prostatic hyperplasia is well documented and this review considers the possibility of a link between changes in autonomic innervation and prostate cancer progression.
Collapse
Affiliation(s)
- Carl W White
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville, VIC Australia
| | | | | |
Collapse
|
39
|
Does benign prostatic hyperplasia treatment with alpha-blockers affect prostate cancer risk? Curr Opin Urol 2013; 23:2-4. [PMID: 23159990 DOI: 10.1097/mou.0b013e32835abcf2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To determine whether alpha-blockers, commonly used for the treatment of benign prostatic hyperplasia, are associated with prostate cancer risk. RECENT FINDINGS Alpha-blockers have been associated with a reduced risk of prostate cancer aggressiveness in some observational studies and an increased risk in other studies. However, this relationship is complex as different alpha-blockers have divergent effects in laboratory studies and there are many confounders in daily practice such as differential screening practices. SUMMARY Both benign prostatic hyperplasia and prostate cancer are common conditions in the aging male population, such that an interaction between alpha-blockers and prostate cancer risk is clinically relevant. Prospective evidence is necessary to establish a definitive link.
Collapse
|
40
|
Yamada D, Nishimatsu H, Kumano S, Hirano Y, Suzuki M, Fujimura T, Fukuhara H, Enomoto Y, Kume H, Homma Y. Reduction of prostate cancer incidence by naftopidil, an α1-adrenoceptor antagonist and transforming growth factor-β signaling inhibitor. Int J Urol 2013; 20:1220-7. [DOI: 10.1111/iju.12156] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 03/06/2013] [Indexed: 11/27/2022]
Affiliation(s)
- Daisuke Yamada
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
- Department of Urology; The Fraternity Memorial Hospital; Tokyo Japan
| | | | - Shintaro Kumano
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
| | - Yoshikazu Hirano
- Department of Urology; The Fraternity Memorial Hospital; Tokyo Japan
| | - Motofumi Suzuki
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
| | - Tetsuya Fujimura
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
| | - Hiroshi Fukuhara
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
| | - Yutaka Enomoto
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
| | - Haruki Kume
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
| | - Yukio Homma
- Department of Urology; The University of Tokyo Hospital; Tokyo Japan
| |
Collapse
|
41
|
Kjellman A, Friis S, Granath F, Gustafsson O, Sørensen HT, Akre O. Treatment with finasteride and prostate cancer survival. Scand J Urol 2012; 47:265-71. [DOI: 10.3109/00365599.2012.737366] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
42
|
Wang L, Liu H, Chen X, Zhang M, Xie K, Ma Q. Immune sculpting of norepinephrine on MHC-I, B7-1, IDO and B7-H1 expression and regulation of proliferation and invasion in pancreatic carcinoma cells. PLoS One 2012; 7:e45491. [PMID: 23029049 PMCID: PMC3446877 DOI: 10.1371/journal.pone.0045491] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 08/23/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The sympathetic neurotransmitter Norepinephrine (NE) contributes to tumorigenesis and cancer progression. This study aims to investigate the role of NE in modulating the immune phenotype and allowing pancreatic carcinoma (PC) cells to escape the immune response. METHODS Varied concentrations of NE and interferon-gamma (IFN-γ) were administrated to MIA PaCa-2 and BxPC-3 cell lines for 48 hours. Proliferation and invasion were then investigated using an MTT assay and a membrane invasion culture system respectively. MHC-I, B7-1, IDO and B7-H1 expression were measured using real-time quantitative RT-PCR, western blotting and immunocytochemistry. The synergistic and time-dependent effects of NE/IFN-γ were also investigated. Adrenergic antagonists were used to identify the relevant target receptor of NE. RESULTS The results showed that NE had dose-dependent and time-dependent effects on cell biological processes as well as on the expression of MHC-I, B7-1, IDO and B7-H1. These effects occurred mainly via the β(2)-adrenergic receptor. Long-term NE treatment was able to antagonize some of the effects of IFN-γ (after 2 weeks of treatment), but NE and IFN-γ had significant synergistic stimulatory effects on IDO and B7-H1 expression. The residual effects on biological activities lasted for 2 weeks, while the immunophenotypic changes decreased at early time points after treatment. CONCLUSIONS NE plays important roles in modulating PC cell biological activities and affecting MHC-I, B7-1, IDO and B7-H1 expression in vitro, mainly via the β2-adrenergic receptor (β2-AR) in a time- and dose-dependent fashion. Only at extended treatment durations could NE affect PC cell progression and immune evasion.
Collapse
Affiliation(s)
- Liancai Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
- Henan Province People’s Hospital, Zhengzhou, Henan Province, China
| | - Han Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Xiangli Chen
- Henan Province People’s Hospital, Zhengzhou, Henan Province, China
| | - Min Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Keping Xie
- Department of Gastrointestinal Medical Oncology, the University of Texas, MD Anderson Cancer Centre, Houston, Texas
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| |
Collapse
|
43
|
Gotoh A, Nagaya H, Kanno T, Nishizaki T. Antitumor action of α(1)-adrenoceptor blockers on human bladder, prostate and renal cancer cells. Pharmacology 2012; 90:242-6. [PMID: 23007551 DOI: 10.1159/000342797] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 08/21/2012] [Indexed: 11/19/2022]
Abstract
The present study investigated the antitumor action of α(1)-adrenoceptor blockers on human bladder, prostate and renal cancer cells. For bladder cancer cell lines used here such as 253J, 5637, KK-47, T24 and UM-UC-3 cells, prazosin, a selective α(1)-adrenoceptor blocker, reduced cell viability at concentrations more than 30 µmol/l. Likewise, naftopidil, a blocker of α(1A)- and α(1D)-adrenoceptors, reduced cell viability for all the bladder cancer cells used here in a concentration (10-100 µmol/l)-dependent manner, with a much greater advantage than prazosin. Naftopidil also reduced cell viability for human prostate cancer cell lines such as DU145, LNCap and PC-3 cells and ACHN human renal cancer cells, with a much higher potential than prazosin. Thus, the results of the present study suggest that naftopidil could be a beneficial antitumor drug for the treatment of urological cancers.
Collapse
Affiliation(s)
- Akinobu Gotoh
- Laboratory of Cell and Gene Therapy, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | | | | | | |
Collapse
|
44
|
Jansen L, Below J, Chang-Claude J, Brenner H, Hoffmeister M. Beta blocker use and colorectal cancer risk. Cancer 2012; 118:3911-9. [DOI: 10.1002/cncr.26727] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 10/11/2011] [Accepted: 11/07/2011] [Indexed: 12/31/2022]
|
45
|
Takara K, Yamamoto K, Matsubara M, Minegaki T, Takahashi M, Yokoyama T, Okumura K. Effects of α-adrenoceptor antagonists on ABCG2/BCRP-mediated resistance and transport. PLoS One 2012; 7:e30697. [PMID: 22355323 PMCID: PMC3280247 DOI: 10.1371/journal.pone.0030697] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 12/23/2011] [Indexed: 01/05/2023] Open
Abstract
Acquired resistance of cancer cells to various chemotherapeutic agents is known as multidrug resistance, and remains a critical factor in the success of cancer treatment. It is necessary to develop the inhibitors for multidrug resistance. The aim of this study was to examine the effects of eight α-adrenoceptor antagonists on ABCG2/BCRP-mediated resistance and transport. Previously established HeLa/SN100 cells, which overexpress ABCG2/BCRP but not ABCB1/MDR1, were used. The effects of the antagonists on sensitivity to mitoxantrone and the transport activity of Hoehst33342, both substrates for ABCG2/BCRP, were evaluated using the WST-1 assay and cellular kinetics, respectively. ABCG2/BCRP mRNA expression and the cell cycle were also examined by real-time RT-PCR and flow cytometry, respectively. Sensitivity to mitoxantrone was reversed by the α-adrenoceptor antagonists in a concentration-dependent manner, although such effects were also found in the parental HeLa cells. Levels of ABCG2/BCRP mRNA expression were not influenced by the antagonists. The transport activity of Hoechst33342 was decreased by doxazosin and prazosin, but unaffected by the other antagonists. In addition, doxazosin and prazosin increased the proportion of S phase cells in the cultures treated with mitoxantrone, whereas the other α-adrenoceptor antagonists increased the percentage of cells in G2/M phase. These findings suggested that doxazosin and prazosin reversed resistance mainly by inhibiting ABCG2/BCRP-mediated transport, but the others affected sensitivity to mitoxantrone via a different mechanism.
Collapse
Affiliation(s)
- Kohji Takara
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, Himeji, Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Liao CH, Guh JH, Chueh SC, Yu HJ. Anti-angiogenic effects and mechanism of prazosin. Prostate 2011; 71:976-84. [PMID: 21541974 DOI: 10.1002/pros.21313] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Accepted: 10/26/2010] [Indexed: 12/20/2022]
Abstract
BACKGROUND Alpha1-adrenoceptors antagonists (doxazosin, terazosin, prazosin) are commonly prescribed for benign prostate hyperplasia and hypertension. Doxazosin and terazosin exhibit anti-angiogenic effects and apoptotic activities against multiple cell types and are potential preventive agents for prostate cancer. Prazosin induces apoptosis in three prostate cancer cell lines. We hypothesized that prazosin, a more potent alpha1-adrenoceptor antagonist with a distinct mechanism, exhibits anti-angiogenic activity. METHODS We examined the effect of prazosin on growth and tube formation of human umbilical vascular endothelial cells (HUVECs). We used flow cytometry to assess the effect of prazosin on cell cycle progression and Western blotting to assess its effect on the expression of various apoptotic proteins. RESULTS Prazosin inhibited the growth of HUVEC with an IC(50) of 6.53 µM and suppressed tube formation in a dose-dependent manner. Unlike prostate cancer cells, prazosin did not arrest cell cycle progression at the G2/M checkpoint. We used rhodamine 123 staining to show that prazosin (20 µM) treatment induced a loss of mitochondrial membrane potential by 12 hr. Prazosin treatment of HUVECs resulted in reduced MCL-1 expression, increased Bad, and Bcl-xL expression, cytochrome c release, and induction of apoptosis via the intrinsic apoptosis pathway. Prazosin induced apoptosis in prostate cancer cells and normal HUVEC cells via different mechanisms. CONCLUSIONS These data suggest that prazosin exhibits anti-angiogenic activity and differentially modulates apoptotic pathways depending on the cell type.
Collapse
Affiliation(s)
- Chun-Hou Liao
- Department of Surgery, Fu Jen Catholic University, Taipei, Taiwan
| | | | | | | |
Collapse
|
47
|
Powe DG, Voss MJ, Zänker KS, Habashy HO, Green AR, Ellis IO, Entschladen F. Beta-blocker drug therapy reduces secondary cancer formation in breast cancer and improves cancer specific survival. Oncotarget 2011; 1:628-38. [PMID: 21317458 DOI: 10.18632/oncotarget.101009] [Citation(s) in RCA: 215] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Laboratory models show that the beta-blocker, propranolol, can inhibit norepinephrine-induced breast cancer cell migration. We hypothesised that breast cancer patients receiving beta-blockers for hypertension would show reduced metastasis and improved clinical outcome. Three patient subgroups were identified from the medical records of 466 consecutive female patients (median age 57, range 28-71) with operable breast cancer and follow-up (>10 years). Two subgroups comprised 43 and 49 hypertensive patients treated with beta-blockers or other antihypertensives respectively, prior to cancer diagnosis. 374 patients formed a non-hypertensive control group. Metastasis development, disease free interval, tumour recurrence and hazards risk were statistically compared between groups. Kaplan-Meier plots were used to model survival and DM. Beta-blocker treated patients showed a significant reduction in metastasis development (p=0.026), tumour recurrence (p=0.001), and longer disease free interval (p=0.01). In addition, there was a 57% reduced risk of metastasis (Hazards ratio=0.430; 95% CI=0.200-0.926, p=0.031), and a 71% reduction in breast cancer mortality after 10 years (Hazards ratio=0.291; 95% CI=0.119-0.715, p=0.007). This proof-of-principle study showed beta-blocker therapy significantly reduces distant metastases, cancer recurrence, and cancer-specific mortality in breast cancer patients suggesting a novel role for beta-blocker therapy. A larger epidemiological study leading to randomised clinical trials is needed for breast and other cancer types including colon, prostate and ovary.
Collapse
Affiliation(s)
- Desmond G Powe
- Department of Cellular Pathology, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, NG7 2UH, UK.
| | | | | | | | | | | | | |
Collapse
|
48
|
Hori Y, Ishii K, Kanda H, Iwamoto Y, Nishikawa K, Soga N, Kise H, Arima K, Sugimura Y. Naftopidil, a selective {alpha}1-adrenoceptor antagonist, suppresses human prostate tumor growth by altering interactions between tumor cells and stroma. Cancer Prev Res (Phila) 2011; 4:87-96. [PMID: 21205739 DOI: 10.1158/1940-6207.capr-10-0189] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In prostate cancer, tumor-stroma interactions play a critical role in the promotion of tumorigenesis, and thus the prevention of those interactions is a promising target to suppress tumor growth. Several studies demonstrated that alpha(1)-adrenoceptor (α(1)-AR) antagonists, therapeutic drugs for benign prostatic hyperplasia, have growth inhibitory effects on human prostate cancer (PCa) cells through induction of apoptosis or G(1) cell-cycle arrest. However, their direct actions on stromal cells surrounding cancer cells have not yet been elucidated. In this study, we investigated the effects of subtype-selective α(1)-AR antagonists (naftopidil, tamsulosin, and silodosin) on prostate tumor growth with a focus on the role of stroma, using commercially available fibroblast cells (PrSC). Tumorigenic studies in vivo showed significant reductions in tumor growth when E9 cells (an androgen low-sensitive LNCaP subline) grafted with PrSC were treated with naftopidil. In in vitro analyses, naftopidil and silodosin showed antiproliferative effects on PCa cells regardless of androgen sensitivity and α(1)-AR subtype expression. In PrSC, a strong growth inhibitory effect was observed with naftopidil but not silodosin. Flow cytometric analysis revealed that naftopidil, but not silodosin, induced G(1) cell-cycle arrest in both PCa cells and PrSC. In naftopidil-treated PrSC, total interleukin-6 protein was significantly reduced with increased suppression of cell proliferation. Silodosin induced weak early apoptosis only in PCa cells. These findings demonstrated that naftopidil strongly suppressed cell proliferation of stromal cells, resulting in decreased tumorigenic soluble factor, suggesting that naftopidil might be effective in preventing stromal support of tumor cells.
Collapse
Affiliation(s)
- Yasuhide Hori
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Mie, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Takeshita K, Takahashi S, Tang M, Seeni A, Asamoto M, Shirai T. Hypertension is positively associated with prostate cancer development in the TRAP transgenic rat model. Pathol Int 2011; 61:202-9. [PMID: 21418392 DOI: 10.1111/j.1440-1827.2011.02645.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Epidemiological data on the relationship between hypertension and prostate cancer development are conflicting. To cast light on this question, we performed animal experiments using hybrid rats generated by crossing the spontaneously hypertensive rat (SHR) or its normotensive control Wistar Kyoto (WKY) rat with a transgenic rat for adenocarcinoma of prostate (TRAP) that features development of adenocarcinoma at high incidence by 15 weeks of age. The number of adenocarcinomatous foci in the lateral prostate of hypertensive (TRAP × SHR)F1 rats was demonstrated to be significantly increased compared with those of normotensive (TRAP × WKY)F1 rats. In the ventral prostate, increase of carcinoma foci was also observed but did not reach significance. The number of cancer foci showing microinvasion in (TRAP × SHR)F1 rats was higher than that of (TRAP × WKY)F1 rats, but again without significance, while treatment with prazosin, an anti-hypertensive agent, tended to decrease microinvasive carcinoma foci in both the ventral and lateral prostate. In conclusion, the present study provided additional evidence that high blood pressure is associated with prostate cancer risk.
Collapse
Affiliation(s)
- Kentaro Takeshita
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Powe DG, Voss MJ, Habashy HO, Zänker KS, Green AR, Ellis IO, Entschladen F. Alpha- and beta-adrenergic receptor (AR) protein expression is associated with poor clinical outcome in breast cancer: an immunohistochemical study. Breast Cancer Res Treat 2011; 130:457-63. [PMID: 21298476 DOI: 10.1007/s10549-011-1371-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 01/23/2011] [Indexed: 12/21/2022]
Abstract
Breast cancer mortality is frequently associated with metastatic disease. Metastasis models have shown adrenoceptor (AR) stimulation induces cell migration which is inhibited by adrenoceptor antagonist drugs. We investigated adrenoceptor protein expression in clinical breast tumours and its association with disease progression and prognosis. Immunohistochemistry on tissue microarrays was used to characterise α1b, α2c and β(2)2 adrenoceptor protein expression in operable breast tumours. Associations with tumour-relevant biological markers and clinical outcome were statistically assessed. Strong α1b expression occurred in large high grade (P < 0.0001), HER2+ (P < 0.0001) or basal-like (CK5/6, P = 0.0005; CK14, P = 0.0001; EGFR, P = 0.003) cancers, showing increased proliferation (Mib1, P = 0.002), decreased apoptosis (Bcl2, P < 0.0001) and poor NPI membership (P = 0.001). α1b expression correlated with poor cancer-specific survival (LR = 7.628, P = 0.022) and tumour recurrence (LR = 6.128, P = 0.047). Strong α2c was over-expressed in high grade (P = 0.007), HER3+ (P = 0.002) and HER4+ (P < 0.0001) cancers with borderline increase in EGFR, p53 and MIB1 proteins, and inverse association with hormonal (PgR, P = 0.002) phenotype. In contrast, strong β(2) expression occurred in small-size, luminal-like (ER+, P < 0.001) tumours of low grade (P < 0.001) and lymph node stage (P = 0.027) that showed poor prognosis when hormonal treatment was withheld. Adrenoceptors were not found to be independent predictors of clinical outcome. Alpha1b and α2c AR is over-expressed in basal-like breast tumours of poor prognosis. Strong β(2) adrenoceptor expression is seen in patients with a luminal (ER+) tumour phenotype and good prognosis, due to benefits derived from hormonal therapy. These findings suggest a possible role for targeted therapy using adrenoceptor antagonists.
Collapse
Affiliation(s)
- D G Powe
- Department of Cellular Pathology, Queen's Medical Centre, Nottingham University Hospitals Trust and School of Molecular Medical Sciences, University of Nottingham, Nottingham NG7 2UH, UK.
| | | | | | | | | | | | | |
Collapse
|