1
|
Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010319. [PMID: 36615513 PMCID: PMC9822219 DOI: 10.3390/molecules28010319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is one of the major causes of death worldwide, ranking as the fifth most incident cancer in 2020 and the fourth leading cause of cancer mortality. The majority of GC patients are in an advanced stage at the time of diagnosis, presenting a poor prognosis and outcome. Current GC treatment approaches involve endoscopic detection, gastrectomy and chemotherapy or chemoradiotherapy in an adjuvant or neoadjuvant setting. Drug development approaches demand extreme effort to identify molecular mechanisms of action of new drug candidates. Drug repurposing is based on the research of new therapeutic indications of drugs approved for other pathologies. In this review, we explore GC and the different drugs repurposed for this disease.
Collapse
Affiliation(s)
- Diana Araújo
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
2
|
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: 3.7] [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
|
3
|
Zhong W, Zhang F, Huang C, Lin Y, Huang J. Classification of Clear Cell Renal Cell Carcinoma based on Tumor Suppressor Genomic Profiling. J Cancer 2021; 12:2359-2370. [PMID: 33758612 PMCID: PMC7974878 DOI: 10.7150/jca.50462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/24/2020] [Indexed: 12/30/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent type of malignancy in adults. However, the clinical significance of tumor suppressor genes (TSG) is largely elusive. Herein, the expression profile TSGs and its clinical response in ccRCC were investigated. A total of 603 ccRCC samples from two cohorts (TCGA and ICGC) were retrieved in this study. Three molecular subtypes (C1, C2, and C3) were identified based on the TSGs expression profile in the TCGA dataset. Through Weighted Gene Correlation Network Analysis (WGCNA), six modules associated with three subtypes were identified. Pathway enrichment for the modules revealed that crucial pathways including p53 signaling and immune-related pathways were significantly enriched. We further focused on the relationship between immune infiltration level and subtypes, and found that subtype C1 was associated with higher immune infiltration level, subtype C2 was corresponding with medium immune infiltration level, whereas subtype C3 was correlated with lower immune infiltration level. Interestingly, C2 have a better survival outcome, while C1 and C3 showed a poor prognosis. Considering their survival difference, we then performed a differentially expression analysis between C2 and C1&3, and a total of 99 differentially expressed tumor suppressor genes (DETSGs) were identified. According to these DETSGs, 59 potential compounds with 28 mechanisms of action (MOA) were predicted using the Connectivity Map (CMap) database. Among these compounds, leflunomide, naftopidil, and ribavirin were the most prospective compounds for the treatment of ccRCC. In addition, we found that subtype C2 is more sensitive to sorafenib and sunitinib drugs, and C2 have more likelihood to be responded to immunotherapy. In summary, the three subtypes hinged on the tumor suppressor gene expression for ccRCC might contribute to understanding the underlying molecular mechanisms of ccRCC. Also, its potential compounds might offer guidelines for developing a novel treatment strategy of ccRCC.
Collapse
Affiliation(s)
- Weimin Zhong
- The Fifth Hospital of Xiamen, Xiamen 361101, Fujian Province, China
| | - Fengling Zhang
- The Fifth Hospital of Xiamen, Xiamen 361101, Fujian Province, China
| | - Chaoqun Huang
- The Fifth Hospital of Xiamen, Xiamen 361101, Fujian Province, China
| | - Yao Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, Fujian Province, China
| | - Jiyi Huang
- The Fifth Hospital of Xiamen, Xiamen 361101, Fujian Province, China
| |
Collapse
|
4
|
King L, Christie D, Dare W, Bernaitis N, Chess-Williams R, McDermott C, Forbes A, Anoopkumar-Dukie S. Quinazoline alpha-adrenoreceptor blockers as an adjunct cancer treatment: From bench to bedside. Eur J Pharmacol 2020; 893:173831. [PMID: 33359146 DOI: 10.1016/j.ejphar.2020.173831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 10/22/2022]
Abstract
Drug repurposing has been increasingly used by both researchers and clinicians to identify new cancer treatments. The alpha-1 adrenoreceptor blockers are a class of drugs that have been used for many years in the treatment of hypertension and benign prostatic hyperplasia. Some of the drugs in this class, notably the quinazoline derivatives, have been found to display cytotoxic properties, identifying them as potential options in the treatment of cancer. This review will examine the currently available evidence that investigates the cytotoxic and anti-cancer properties of these agents, the mechanisms behind these properties and how the alpha-1 blockers fit within current cancer therapies. It aims to answer the question of whether these agents can go from the laboratory bench top into cancer clinics.
Collapse
Affiliation(s)
- Liam King
- School of Pharmacy and Pharmacology, Griffith University, Queensland, Australia; Ramsay Pharmacy, John Flynn Private Hospital, Queensland, Australia
| | | | - Wendy Dare
- Ramsay Pharmacy, John Flynn Private Hospital, Queensland, Australia
| | - Nijole Bernaitis
- Ramsay Pharmacy, John Flynn Private Hospital, Queensland, Australia
| | | | | | - Amanda Forbes
- Faculty of Health Sciences, Bond University, Queensland, Australia
| | | |
Collapse
|
5
|
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: 0.8] [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
|
6
|
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: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
7
|
Florent R, Weiswald LB, Lambert B, Brotin E, Abeilard E, Louis MH, Babin G, Poulain L, N'Diaye M. Bim, Puma and Noxa upregulation by Naftopidil sensitizes ovarian cancer to the BH3-mimetic ABT-737 and the MEK inhibitor Trametinib. Cell Death Dis 2020; 11:380. [PMID: 32424251 PMCID: PMC7235085 DOI: 10.1038/s41419-020-2588-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022]
Abstract
Ovarian cancer represents the first cause of mortality from gynecologic malignancies due to frequent chemoresistance occurrence. Increasing the [BH3-only Bim, Puma, Noxa proapoptotic]/[Bcl-xL, Mcl-1 antiapoptotic] proteins ratio was proven to efficiently kill ovarian carcinoma cells and development of new molecules to imbalance Bcl-2 member equilibrium are strongly required. Drug repurposing constitutes an innovative approach to rapidly develop therapeutic strategies through exploitation of established drugs already approved for the treatment of noncancerous diseases. This strategy allowed a renewed interest for Naftopidil, an α1-adrenergic receptor antagonist commercialized in Japan for benign prostatic hyperplasia. Naftopidil was reported to decrease the incidence of prostate cancer and its derivative was described to increase BH3-only protein expression in some cancer models. Based on these arguments, we evaluated the effects of Naftopidil on ovarian carcinoma and showed that Naftopidil reduced cell growth and increased the expression of the BH3-only proteins Bim, Puma and Noxa. This effect was independent of α1-adrenergic receptors blocking and involved ATF4 or JNK pathway depending on cellular context. Finally, Naftopidil-induced BH3-only members sensitized our models to ABT-737 and Trametinib treatments, in vitro as well as ex vivo, in patient-derived organoid models.
Collapse
Affiliation(s)
- Romane Florent
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Louis-Bastien Weiswald
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Bernard Lambert
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
- CNRS-Regional Delegation of Normandy, Caen, France
| | - Emilie Brotin
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Edwige Abeilard
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Marie-Hélène Louis
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Guillaume Babin
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Laurent Poulain
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
- Biological Ressources Center «OvaRessources», Cancer Center François Baclesse, Caen, France
| | - Monique N'Diaye
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France.
- UNICANCER, Cancer Center François Baclesse, Caen, France.
| |
Collapse
|
8
|
Urushiyama H, Terasaki Y, Nagasaka S, Kokuho N, Endo Y, Terasaki M, Kunugi S, Makita K, Isago H, Hosoki K, Souma K, Ishii T, Matsuzaki H, Hiraishi Y, Mikami Y, Noguchi S, Tamiya H, Mitani A, Yamauchi Y, Shimizu A, Nagase T. Naftopidil reduced the proliferation of lung fibroblasts and bleomycin-induced lung fibrosis in mice. J Cell Mol Med 2019; 23:3563-3571. [PMID: 30873733 PMCID: PMC6484423 DOI: 10.1111/jcmm.14255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/05/2019] [Accepted: 02/09/2019] [Indexed: 01/16/2023] Open
Abstract
Naftopidil, an α‐1 adrenoceptor antagonist with few adverse effects, is prescribed for prostate hyperplasia. Naftopidil inhibits prostate fibroblast proliferation; however, its effects on lung fibroblasts and fibrosis remain largely unknown. Two normal and one idiopathic pulmonary fibrosis human lung fibroblast lines were cultured with various naftopidil concentrations with or without phenoxybenzamine, an irreversible α‐1 adrenoceptor inhibitor. We examined the incorporation of 5‐bromo‐2ʹ‐deoxyuridine into DNA and lactic acid dehydrogenase release by enzyme‐linked immunosorbent assay, cell cycle analysis by flow cytometry, scratch wound‐healing assay, and mRNA expressions of type IV collagen and α‐smooth muscle actin by polymerase chain reaction. Effects of naftopidil on bleomycin‐induced lung fibrosis in mice were evaluated using histology, micro‐computed tomography, and surfactant protein‐D levels in serum. Naftopidil, dose‐dependently but independently of phenoxybenzamine, inhibited 5‐bromo‐2ʹ‐deoxyuridine incorporation in lung fibroblasts. Naftopidil induced G1 cell cycle arrest, but lactic acid dehydrogenase release and migration ability of lung fibroblasts were unaffected. Naftopidil decreased mRNA expressions of type IV collagen and α‐smooth muscle actin in one normal lung fibroblast line. Histological and micro‐computed tomography examination revealed that naftopidil attenuated lung fibrosis and decreased serum surfactant protein‐D levels in bleomycin‐induced lung fibrosis in mice. In conclusion, naftopidil may have therapeutic effects on lung fibrosis.
Collapse
Affiliation(s)
- Hirokazu Urushiyama
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Yasuhiro Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Shinya Nagasaka
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Nariaki Kokuho
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Youko Endo
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Shinobu Kunugi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Kosuke Makita
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hideaki Isago
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Keisuke Hosoki
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kunihiko Souma
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Takashi Ishii
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hirotaka Matsuzaki
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yoshihisa Hiraishi
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yu Mikami
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Satoshi Noguchi
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hiroyuki Tamiya
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Akihisa Mitani
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yasuhiro Yamauchi
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
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 2017; 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] [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
|
10
|
Iwamoto Y, Ishii K, Kanda H, Kato M, Miki M, Kajiwara S, Arima K, Shiraishi T, Sugimura Y. Combination treatment with naftopidil increases the efficacy of radiotherapy in PC-3 human prostate cancer cells. J Cancer Res Clin Oncol 2017; 143:933-939. [PMID: 28243746 DOI: 10.1007/s00432-017-2367-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/06/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE Clinically, radiotherapy (RT) often leads to the development of prostate cancer (PCa) resistance because of protective responses in cancer cells. One of the mechanisms includes the upregulation of RT-induced antioxidant enzymes. Thus, combination therapy with RT and certain pharmaceutical drugs targeting antioxidant enzymes may be ideal for increasing the efficacy of RT with minimum side effects. Naftopidil is a subtype-selective α1D-adrenoceptor antagonist used for the treatment of benign prostatic hyperplasia (BPH). In our drug repositioning study, naftopidil showed not only unique growth-inhibitory effects but also AKT phosphorylation-inhibitory effects in PC-3 human PCa cells. Here, we examined the efficacy of additive naftopidil treatment in combination with RT in PC-3 cells. METHODS The effects of combination therapy with RT plus naftopidil were analyzed using an animal model of PC-3 xenografts in BALB/c nude mice. The expression of the antioxidant enzyme manganese superoxide dismutase (MnSOD) was evaluated by western blotting. RESULTS Combination therapy with RT plus naftopidil induced a more efficacious delay in PC-3 xenograft tumor growth as compared with monotherapy with naftopidil or RT. In PC-3 tumors, combination therapy with RT plus naftopidil suppressed the upregulation of RT-induced MnSOD expression. In vitro, neither AKT inhibitor IV nor naftopidil directly altered MnSOD expression. Upregulation of RT-induced MnSOD expression was markedly suppressed by combination treatment with RT plus AKT inhibitor IV or naftopidil. CONCLUSIONS These results suggest that additive naftopidil treatment in combination with RT may increase the efficacy of RT for the treatment of PCa.
Collapse
Affiliation(s)
- Yoichi Iwamoto
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Kenichiro Ishii
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Hideki Kanda
- 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
| | - Manabu Miki
- 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
| | - 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
|
11
|
Nakano T, Kuribayashi K, Mikami K. Possible new therapeutic agents for malignant pleural mesothelioma: anti-CD26 monoclonal antibody and naftopidil. Expert Rev Anticancer Ther 2016; 16:1097-1099. [PMID: 27718761 DOI: 10.1080/14737140.2016.1241150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Takashi Nakano
- a Division of Respiratory Diseases, Department of Internal Medicine , Hyogo College of Medicine , Hyogo , Japan
| | - Kozo Kuribayashi
- a Division of Respiratory Diseases, Department of Internal Medicine , Hyogo College of Medicine , Hyogo , Japan
| | - Koji Mikami
- a Division of Respiratory Diseases, Department of Internal Medicine , Hyogo College of Medicine , Hyogo , Japan
| |
Collapse
|
12
|
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: 3.1] [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
|
13
|
Fibroblasts prolong serum prostate-specific antigen decline after androgen deprivation therapy in prostate cancer. J Transl Med 2016; 96:338-49. [PMID: 26641067 DOI: 10.1038/labinvest.2015.136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 09/29/2015] [Accepted: 10/08/2015] [Indexed: 11/09/2022] Open
Abstract
In patients with prostate cancer (PCa), serum prostate-specific antigen (PSA) is a useful marker for evaluating the effects of androgen deprivation therapy (ADT). Intuitively, most urologists expect that a more rapid PSA decline in response to ADT would be positively associated with extended survival. Recently, we have reported that prolonged gradual serum PSA decline after ADT is strongly associated with favorable prognosis in PCa patients, however, the mechanism remains unknown. We investigated the role of fibroblasts in serum PSA decline after ADT. We performed in vitro experiments using androgen-sensitive, androgen receptor (AR)-positive prostate epithelial cell lines (LNCaP, 22Rv1, and RWPE-1 cells), commercially available prostate stromal cells (PrSC), and primary cultures of prostate fibroblasts (pcPrFs). In LNCaP and 22Rv1 cells, PSA production was increased by co-culture with fibroblasts under androgen-deprived conditions. In an in vivo model using LNCaP cells, serum PSA declined rapidly after ADT becoming undetectable within 14 days in mice inoculated with LNCaP cells alone. In contrast, when LNCaP cells were co-inoculated with fibroblasts, serum PSA levels were still high on 14 days post ADT and did not drop to undetectable levels until 21 days post ADT. Tumor volumes and Ki67 labeling indices were not altered between days 14 and 21 post ADT in mice inoculated with LNCaP cells; however, those in mice inoculated with LNCaP cells plus fibroblasts decreased gradually. PSA protein was detected in all tumors on 21 days post ADT by immunohistochemical staining. Microvessel densities were higher on 14 days post ADT for tumors from mice inoculated with LNCaP cells plus fibroblasts as compared with LNCaP cells alone. In summary, co-inoculation of fibroblasts with LNCaP cells prolonged serum PSA decline after ADT and enhanced the efficacy of ADT. Prolonged serum PSA decline may indicate the presence of protective fibroblasts that preserve the AR dependence of PCa cells, improving treatment efficacy.
Collapse
|
14
|
Ishii K, Sugimura Y. Identification of a new pharmacological activity of the phenylpiperazine derivative naftopidil: tubulin-binding drug. J Chem Biol 2014; 8:5-9. [PMID: 25584077 DOI: 10.1007/s12154-014-0122-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 08/25/2014] [Indexed: 11/26/2022] Open
Abstract
The phenylpiperazine derivative naftopidil is an α1-adrenoceptor (AR) antagonist that has been used clinically to treat benign prostatic hyperplasia. In our drug repositioning research, naftopidil shows the unique growth-inhibitory effects. Naftopidil inhibits cell cycle progression not only in cancer cells, but also in fibroblasts and vascular endothelial cells. Naftopidil-inhibited cell cycle progression is independent of α1-AR expression in cells. Therefore, the antiproliferative effects of naftopidil may be due to the off-target effects of the drug. In this study, we attempted to identify the off-target molecules of naftopidil using the magnetic nanobeads, ferrite glycidyl metharcrylate (FG) beads. Similar to naftopidil, its derivatives TG09-01 and TG09-02, which were introduced with amino groups for immobilizing to FG beads, inhibited cell growth in human HT29 colon adenocarcinoma cells. Both derivatives were associated with inhibition of cell cycle progression in HT29 cells. This observation is consistent with that seen with naftopidil. Using TG09-02-immobilized FG beads, α- and β-tubulins were identified as the specific binding proteins of naftopidil. The tubulin polymerization assay clearly indicated that naftopidil bound directly to tubulin and inhibited the polymerization of tubulin. Other phenylpiperazine derivatives, such as RS100329, BMY7378, and KN-62, also inhibited the polymerization of tubulin. These results suggest that phenylpiperazine derivatives including naftopidil may have broad spectrum of cellular cytotoxicity in various types of cells. In addition, the tubulin polymerization-inhibiting activity of phenylpiperazine derivatives may be a specific feature of the phenylpiperazine-based structure. These findings can allow us to design and synthesize new tubulin-binding drugs derived from naftopidil as a lead compound.
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
| | - Yoshiki Sugimura
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507 Japan
| |
Collapse
|