1
|
Ding M, Lin J, Qin C, Fu Y, Du Y, Qiu X, Wei P, Xu T. Novel CAR-T Cells Specifically Targeting SIA-CIgG Demonstrate Effective Antitumor Efficacy in Bladder Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400156. [PMID: 39178136 PMCID: PMC11516049 DOI: 10.1002/advs.202400156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/15/2024] [Indexed: 08/25/2024]
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy is a promising cancer treatment method. However, its application in bladder cancer (BC) remains limited, partially because of the absence of appropriate target molecules. Sialylated cancer-derived IgG (SIA-CIgG) is highly expressed in BC and is closely associated with malignant biological behavior. However, its potential as a target for CAR-T cell therapy to treat BC is yet to be established. Here, it is found that SIA-CIgG is highly expressed in most BC samples but displayed limited expression in normal tissues. CAR-T cells specifically targeting SIA-CIgG can effectively lyse BC cells and the cytotoxicity depends on SIA-CIgG expression. Furthermore, SIA-CIgG CAR-T cells demonstrate milder tumor cell lysis and enhanced persistence compared with human epidermal growth factor receptor 2 (HER2) CAR-T cells, which have undergone extensive clinical trials. After repeated tumor antigen challenges, SIA-CIgG CAR-T cells display substantial alterations in both the transcriptome and chromatin accessibility. When combining SIA-CIgG CAR-T cell therapy with FDA-approved drugs to treat BC, the histone deacetylase inhibitor (HDACi), vorinostat, is found to enhance the ablility of CAR-T cells for tumor cell lysis. Therefore, the combination of SIA-CIgG CAR-T cells and vorinostat is promising for BC treatment.
Collapse
Affiliation(s)
- Mengting Ding
- Department of UrologyPeking University People's HospitalBeijing100044China
| | - Jiaxing Lin
- Department of UrologyPeking University People's HospitalBeijing100044China
| | - Caipeng Qin
- Department of UrologyPeking University People's HospitalBeijing100044China
| | - Yuhao Fu
- Center for Cell and Gene Circuit DesignCAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdong518055China
| | - Yiqing Du
- Department of UrologyPeking University People's HospitalBeijing100044China
| | - Xiaoyan Qiu
- Department of ImmunologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Ping Wei
- Center for Cell and Gene Circuit DesignCAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdong518055China
| | - Tao Xu
- Department of UrologyPeking University People's HospitalBeijing100044China
| |
Collapse
|
2
|
Signaling Pathways in Inflammation and Cardiovascular Diseases: An Update of Therapeutic Strategies. IMMUNO 2022. [DOI: 10.3390/immuno2040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Inflammatory processes represent a pivotal element in the development and complications of cardiovascular diseases (CVDs). Targeting these processes can lead to the alleviation of cardiomyocyte (CM) injury and the increase of reparative mechanisms. Loss of CMs from inflammation-associated cardiac diseases often results in heart failure (HF). Evidence of the crosstalk between nuclear factor-kappa B (NF-κB), Hippo, and mechanistic/mammalian target of rapamycin (mTOR) has been reported in manifold immune responses and cardiac pathologies. Since these signaling cascades regulate a broad array of biological tasks in diverse cell types, their misregulation is responsible for the pathogenesis of many cardiac and vascular disorders, including cardiomyopathies and atherosclerosis. In response to a myriad of proinflammatory cytokines, which induce reactive oxygen species (ROS) production, several molecular mechanisms are activated within the heart to inaugurate the structural remodeling of the organ. This review provides a global landscape of intricate protein–protein interaction (PPI) networks between key constituents of NF-κB, Hippo, and mTOR signaling pathways as quintessential targetable candidates for the therapy of cardiovascular and inflammation-related diseases.
Collapse
|
3
|
Hou T, Dan W, Liu T, Liu B, Wei Y, Yue C, Que T, Ma B, Lei Y, Wang Z, Zeng J, Fan Y, Li L. Deubiquitinase OTUD5 modulates mTORC1 signaling to promote bladder cancer progression. Cell Death Dis 2022; 13:778. [PMID: 36085200 PMCID: PMC9463452 DOI: 10.1038/s41419-022-05128-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 01/21/2023]
Abstract
The mechanistic (formally "mammalian") target of rapamycin (mTOR) pathway serves as a crucial regulator of various biological processes such as cell growth and cancer progression. In bladder cancer, recent discoveries showing the cancer-promoting role of mTOR complex 1 have attracted wide attention. However, the regulation of mTOR signaling in bladder cancer is complicated and the underlying mechanism remains elusive. Here, we report that the deubiquitinating enzyme, ovarian tumor domain-containing protein 5 (OTUD5), can activate the mTOR signaling pathway, promote cancer progression, and show its oncogenic potential in bladder cancer. In our study, we found that OTUD5 deubiquitinated a RING-type E3 ligase, RNF186, and stabilized its function. In addition, the stabilization of RNF186 further led to the degradation of sestrin2, which is an inhibitor of the mTOR signaling pathway. Together, we provide novel insights into the pathogenesis of bladder cancer and first prove that OTUD5 can promote bladder cancer progression through the OTUD5-RNF186-sestrin2-mTOR axis, which may be exploited in the future for the diagnosis and treatment of this malignancy.
Collapse
Affiliation(s)
- Tao Hou
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Weichao Dan
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Tianjie Liu
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Bo Liu
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Yi Wei
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Chenyang Yue
- grid.21100.320000 0004 1936 9430Department of Biology, York University, Toronto, ON M3J1P3 Canada
| | - Taotao Que
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Bohan Ma
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Yuzeshi Lei
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Zixi Wang
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Jin Zeng
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Yizeng Fan
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| | - Lei Li
- grid.452438.c0000 0004 1760 8119Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 P.R. China ,grid.43169.390000 0001 0599 1243Xi’an Jiaotong university, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China ,grid.452438.c0000 0004 1760 8119The First Affiliated Hospital of Xi’an Jiaotong University, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi China
| |
Collapse
|
4
|
Selvarani R, Mohammed S, Richardson A. Effect of rapamycin on aging and age-related diseases-past and future. GeroScience 2021; 43:1135-1158. [PMID: 33037985 PMCID: PMC8190242 DOI: 10.1007/s11357-020-00274-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
In 2009, rapamycin was reported to increase the lifespan of mice when implemented later in life. This observation resulted in a sea-change in how researchers viewed aging. This was the first evidence that a pharmacological agent could have an impact on aging when administered later in life, i.e., an intervention that did not have to be implemented early in life before the negative impact of aging. Over the past decade, there has been an explosion in the number of reports studying the effect of rapamycin on various diseases, physiological functions, and biochemical processes in mice. In this review, we focus on those areas in which there is strong evidence for rapamycin's effect on aging and age-related diseases in mice, e.g., lifespan, cardiac disease/function, central nervous system, immune system, and cell senescence. We conclude that it is time that pre-clinical studies be focused on taking rapamycin to the clinic, e.g., as a potential treatment for Alzheimer's disease.
Collapse
Affiliation(s)
- Ramasamy Selvarani
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sabira Mohammed
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Arlan Richardson
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
5
|
Wu CF, Wu CY, Chiou RYY, Yang WC, Lin CF, Wang CM, Hou PH, Lin TC, Kuo CY, Chang GR. The Anti-Cancer Effects of a Zotarolimus and 5-Fluorouracil Combination Treatment on A549 Cell-Derived Tumors in BALB/c Nude Mice. Int J Mol Sci 2021; 22:4562. [PMID: 33925400 PMCID: PMC8123799 DOI: 10.3390/ijms22094562] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/29/2022] Open
Abstract
Zotarolimus is a semi-synthetic derivative of rapamycin and a novel immunosuppressive agent used to prevent graft rejection. The pharmacological pathway of zotarolimus restricts the kinase activity of the mammalian target of rapamycin (mTOR), which potentially leads to reductions in cell division, cell growth, cell proliferation, and inflammation. These pathways have a critical influence on tumorigenesis. This study aims to examine the anti-tumor effect of zotarolimus or zotarolimus combined with 5-fluorouracil (5-FU) on A549 human lung adenocarcinoma cell line implanted in BALB/c nude mice by estimating tumor growth, apoptosis expression, inflammation, and metastasis. We established A549 xenografts in nude mice, following which we randomly divided the mice into four groups: control, 5-FU (100 mg/kg/week), zotarolimus (2 mg/kg/day), and zotarolimus combined with 5-FU. Compared the results with those for control mice, we found that mice treated with zotarolimus or zotarolimus combined with 5-FU retarded tumor growth; increased tumor apoptosis through the enhanced expression of cleaved caspase 3 and extracellular signal-regulated kinase (ERK) phosphorylation; decreased inflammation cytokines levels (e.g., IL-1β, TNF-α, and IL-6); reduced inflammation-related factors such as cyclooxygenase-2 (COX-2) protein and nuclear factor-κB (NF-κB) mRNA; enhanced anti-inflammation-related factors including IL-10 and inhibitor of NF-κB kinase α (IκBα) mRNA; and inhibited metastasis-related factors such as transforming growth factor β (TGF-β), CD44, epidermal growth factor receptor (EGFR), and vascular endothelial growth factor (VEGF). Notably, mice treated with zotarolimus combined with 5-FU had significantly retarded tumor growth, reduced tumor size, and increased tumor inhibition compared with the groups of mice treated with 5-FU or zotarolimus alone. The in vivo study confirmed that zotarolimus or zotarolimus combined with 5-FU could retard lung adenocarcinoma growth and inhibit tumorigenesis. Zotarolimus and 5-FU were found to have an obvious synergistic tumor-inhibiting effect on lung adenocarcinoma. Therefore, both zotarolimus alone and zotarolimus combined with 5-FU may be potential anti-tumor agents for treatment of human lung adenocarcinoma.
Collapse
Affiliation(s)
- Ching-Feng Wu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Linkou, 5 Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; (C.-F.W.); (C.-Y.W.)
| | - Ching-Yang Wu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Linkou, 5 Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; (C.-F.W.); (C.-Y.W.)
| | - Robin Y.-Y. Chiou
- Department of Food Science, National Chiayi University, 300 University Road, Chiayi 60004, Taiwan;
| | - Wei-Cheng Yang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 4 Section, 1 Roosevelt Road, Taipei 10617, Taiwan;
| | - Chuen-Fu Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 912301, Taiwan;
| | - Chao-Min Wang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.)
| | - Po-Hsun Hou
- Department of Psychiatry, Taichung Veterans General Hospital, 4 Section, 1650 Taiwan Boulevard, Taichung 40705, Taiwan;
- Faculty of Medicine, National Yang-Ming University, 2 Section, 155 Linong Street, Beitou District, Taipei 11221, Taiwan
| | - Tzu-Chun Lin
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.)
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 289 Jianguo Road, Xindian District, New Taipei City 231405, Taiwan
| | - Geng-Ruei Chang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan; (C.-M.W.); (T.-C.L.)
| |
Collapse
|
6
|
Almeida TC, da Silva GN. Resveratrol effects in bladder cancer: A mini review. Genet Mol Biol 2021; 44:e20200371. [PMID: 33749701 PMCID: PMC7983189 DOI: 10.1590/1678-4685-gmb-2020-0371] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer has a high incidence worldwide and is the most common genitourinary cancer. The treatment of bladder cancer involves surgery and chemotherapy; however high failure rates and toxicity are observed. In this context, the search of new drugs aiming a more effective treatment is extremely necessary. Natural products are an important source of compounds with antiproliferative effects. Resveratrol is a naturally occurring plant polyphenol whose anticancer activity has been demonstrated in different types of cancer. This review summarizes the in vitro and in vivo studies using models of bladder cancer treated with resveratrol and discusses its different mechanisms of action.
Collapse
Affiliation(s)
- Tamires Cunha Almeida
- Universidade Federal de Ouro Preto, Laboratório de Pesquisas
Clínicas, Ouro Preto, MG, Brazil
| | - Glenda Nicioli da Silva
- Universidade Federal de Ouro Preto, Laboratório de Pesquisas
Clínicas, Ouro Preto, MG, Brazil
| |
Collapse
|
7
|
Significant benefit of everolimus in a patient with urothelial bladder cancer harboring a rare M1043I mutation of PIK3CA. Invest New Drugs 2021; 39:1197-1199. [PMID: 33745098 DOI: 10.1007/s10637-021-01103-8] [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: 02/09/2021] [Accepted: 03/12/2021] [Indexed: 10/21/2022]
Abstract
Urothelial bladder cancer (UBC) is a common malignancy with considerable mortality worldwide. However, the treatment options of UBC are mainly chemotherapy and immunotherapy, as few targeted agents have demonstrated efficacy against UBC. In recent studies, everolimus has exhibited antitumor activity in patients harboring aberrations in the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/ mammalian target of rapamycin (mTOR) pathway in multiple tumor types. Herein, we report the case of a patient with metastatic UBC harboring a rare M1043I mutation of PIK3CA which was detected using DNA-based next-generation sequencing. The patient received everolimus as first-line therapy after palliative transurethral resection. The treatment resulted in complete response within 1 month, and the patient achieved a progression-free survival (PFS) of >6 months according to reports from the last follow-up visit. To our knowledge, this is the first reported case of PIK3CA-mutant UBC for which everolimus therapy demonstrated a significant benefit suggesting that the rare M1043I mutation variant may be a potential biomarker of sensitivity to everolimus. Further insights into its mechanism and clinical studies are needed to clarify the effectiveness of everolimus therapy in patients with PIK3CA M1043I mutation.
Collapse
|
8
|
TOR Signaling Pathway in Cardiac Aging and Heart Failure. Biomolecules 2021; 11:biom11020168. [PMID: 33513917 PMCID: PMC7911348 DOI: 10.3390/biom11020168] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
Abstract
Mechanistic Target of Rapamycin (mTOR) signaling is a key regulator of cellular metabolism, integrating nutrient sensing with cell growth. Over the past two decades, studies on the mTOR pathway have revealed that mTOR complex 1 controls life span, health span, and aging by modulating key cellular processes such as protein synthesis, autophagy, and mitochondrial function, mainly through its downstream substrates. Thus, the mTOR pathway regulates both physiological and pathological processes in the heart from embryonic cardiovascular development to maintenance of cardiac homeostasis in postnatal life. In this regard, the dysregulation of mTOR signaling has been linked to many age-related pathologies, including heart failure and age-related cardiac dysfunction. In this review, we highlight recent advances of the impact of mTOR complex 1 pathway and its regulators on aging and, more specifically, cardiac aging and heart failure.
Collapse
|
9
|
Wang Z, Ong WYF, Tong S, Sng JH, Lata RM, Mahendran R, Kesavan E, Chiong E. Beyond diabetes mellitus: role of metformin in non-muscle invasive bladder cancer. Singapore Med J 2020; 63:209-213. [PMID: 32798360 DOI: 10.11622/smedj.2020121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Usage of metformin is associated with improved survival in lung, breast and prostate cancer, and metformin has been shown to inhibit cancer cell growth and proliferation in in vitro studies. Given the lack of clinical data on metformin use in patients with bladder cancer, we aimed to evaluate the role of metformin in their oncological outcomes. METHODS Medication use from a prospectively maintained database of 122 patients with non-muscle invasive bladder cancer treated with intravesical Bacille Calmette-Guerin (BCG), who were recruited under a randomised, double-blinded, controlled clinical trial, was collected and analysed. Kaplan-Meier curves were used to assess overall survival (OS) and disease-specific survival (DSS). RESULTS At a median follow-up duration of 102 (range 3-357) months, 53 (43.4%) patients experienced disease recurrence and 21 (17.2%) experienced disease progression. There was no significant difference in mortality between patients with diabetes mellitus and those without. There was significant difference in OS among patients without diabetes mellitus, patients with diabetes mellitus on metformin and patients with diabetes mellitus not on metformin (p = 0.033); patients with diabetes mellitus on metformin had the best prognosis. Metformin use was associated with significantly lower DSS (p = 0.042). Other oral hypoglycaemic agents, insulin or statins were not associated with disease recurrences or progression. CONCLUSION Metformin use was associated with improved oncological outcomes in patients with non-muscle invasive bladder cancer treated with intravesical BCG. Prospective studies with larger patient populations are needed to validate the role of metformin as potential therapy for bladder cancer.
Collapse
Affiliation(s)
- Ziting Wang
- Department of Urology, National University Hospital, Singapore
| | | | - Shen Tong
- Department of Urology, National University Hospital, Singapore
| | - Jen-Hwei Sng
- Department of Urology, National University of Singapore, Singapore
| | - Raman Mani Lata
- Department of Urology, National University Hospital, Singapore
| | - Ratha Mahendran
- Department of Urology, National University of Singapore, Singapore
| | | | - Edmund Chiong
- Department of Urology, National University Hospital, Singapore
| |
Collapse
|
10
|
Immunotherapy in Bladder Cancer: Current Methods and Future Perspectives. Cancers (Basel) 2020; 12:cancers12051181. [PMID: 32392774 PMCID: PMC7281703 DOI: 10.3390/cancers12051181] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/02/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer is one of the most significant genitourinary cancer, causing high morbidity and mortality in a great number of patients. Over the years, various treatment methods for this type of cancer have been developed. The most common is the highly efficient method using Bacillus Calmette-Guerin, giving a successful effect in a high percentage of patients. However, due to the genetic instability of bladder cancer, together with individual needs of patients, the search for different therapy methods is ongoing. Immune checkpoints are cell surface molecules influencing the immune response and decreasing the strength of the immune response. Among those checkpoints, the PD-1 (programmed cell death protein-1)/PD-L1 (programmed cell death protein ligand 1) inhibitors aim at blocking those molecules, which results in T cell activation, and in bladder cancer the use of Atezolizumab, Avelumab, Durvalumab, Nivolumab, and Pembrolizumab has been described. The inhibition of another pivotal immune checkpoint, CTLA-4 (cytotoxic T cell antigen), may result in the mobilization of the immune system against bladder cancer and, among anti-CTLA-4 antibodies, the use of Ipilimumab and Tremelimumab has been discussed. Moreover, several different approaches to successful bladder cancer treatment exists, such as the use of ganciclovir and mTOR (mammalian target of rapamycin) kinase inhibitors, IL-12 (interleukin-12) and COX-2 (cyclooxygenase-2). The use of gene therapies and the disruption of different signaling pathways are currently being investigated. Research suggests that the combination of several methods increases treatment efficiency and the positive outcome in individual.
Collapse
|
11
|
Iida K, Naiki T, Naiki-Ito A, Suzuki S, Kato H, Nozaki S, Nagai T, Etani T, Nagayasu Y, Ando R, Kawai N, Yasui T, Takahashi S. Luteolin suppresses bladder cancer growth via regulation of mechanistic target of rapamycin pathway. Cancer Sci 2020; 111:1165-1179. [PMID: 31994822 PMCID: PMC7156788 DOI: 10.1111/cas.14334] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/24/2022] Open
Abstract
Luteolin is a natural flavonoid with strong anti–oxidative properties that is reported to have an anti–cancer effect in several malignancies other than bladder cancer. In this study, we describe the effect of luteolin on a human bladder cancer cell line, T24, in the context of the regulation of p21, thioredoxin‐1 (TRX1) and the mechanistic target of rapamycin (mTOR) pathway. Luteolin inhibited cell survival and induced G2/M cell‐cycle arrest, p21 upregulation and downregulation of phospho(p)‐S6, which is downstream of mTOR signaling. Luteolin also upregulated TRX1 and reduced intracellular reactive oxygen species production. In a subcutaneous xenograft mouse model using the rat bladder cancer cell line, BC31, tumor volumes were significantly decreased in mice orally administered luteolin compared to control. Immunohistochemical analysis revealed that increased p21 and decreased p‐S6 expression were induced in the luteolin treatment group. Moreover, in another in vivo N‐butyl‐N‐(4‐hydroxybutyl) nitrosamine (BBN)‐induced rat bladder cancer model, the oral administration of luteolin led to a trend of decreased bladder tumor dimension and significantly decreased the Ki67‐labeling index and p‐S6 expression. Furthermore, the major findings on the metabolism of luteolin suggest that both plasma and urine luteolin‐3ʹ‐O‐glucuronide concentrations are strongly associated with the inhibition of cell proliferation and mTOR signaling. Moreover, a significant decrease in the squamous differentiation of bladder cancer is attributed to plasma luteolin‐3ʹ‐glucuronide concentration. In conclusion, luteolin, and in particular its metabolized product, may represent another natural product‐derived therapeutic agent that acts against bladder cancer by upregulating p21 and inhibiting mTOR signaling.
Collapse
Affiliation(s)
- Keitaro Iida
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shugo Suzuki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Nozaki
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Nagai
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshiki Etani
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuko Nagayasu
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryosuke Ando
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Noriyasu Kawai
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
12
|
Liu Y, Lin F, Chen Y, Wang R, Liu J, Jin Y, An R. Cryptotanshinone Inhibites Bladder Cancer Cell Proliferation and Promotes Apoptosis via the PTEN/PI3K/AKT Pathway. J Cancer 2020; 11:488-499. [PMID: 31897244 PMCID: PMC6930428 DOI: 10.7150/jca.31422] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 09/22/2019] [Indexed: 12/14/2022] Open
Abstract
Cryptotanshinone (CTT), extracted from the root of Salvia miltiorrhiza Bunge (Danshen), exhibits activities against a variety of human cancers in vitro and in vivo. The purpose of this study was to investigate the potential inhibitory effect of CTT on bladder cancer. In this study, we found that CTT inhibited bladder cancer cell proliferation, migration, and invasion and promoted apoptosis. In addition, CTT modulated the expression of proteins via the PI3K/AKT pathway, and the inhibition of PI3K/AKT signalling was due to induction of PTEN expression. Taken together, the results of the present study demonstrated the anticancer effect of CTT on bladder cancer cells, which might be associated with the downregulation of PI3K/AKT/mTOR and NF-κB signalling pathway proteins, and this inhibition was mediated by the induction of PTEN.
Collapse
Affiliation(s)
- Yadong Liu
- Department of Urology. The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Fanlu Lin
- Department of Urology. The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China.,Department of Urology. Linyi Central Hospital, Linyi, Shandong, 276400, People's Republic of China
| | - Yaodong Chen
- Department of ultrasonic imaging, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Rui Wang
- Department of Urology. The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Jiannan Liu
- Department of Urology. The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Yinshan Jin
- Department of Urology. The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Ruihua An
- Department of Urology. The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| |
Collapse
|
13
|
Hernández-Prat A, Rodriguez-Vida A, Juanpere-Rodero N, Arpi O, Menéndez S, Soria-Jiménez L, Martínez A, Iarchouk N, Rojo F, Albanell J, Brake R, Rovira A, Bellmunt J. Novel Oral mTORC1/2 Inhibitor TAK-228 Has Synergistic Antitumor Effects When Combined with Paclitaxel or PI3Kα Inhibitor TAK-117 in Preclinical Bladder Cancer Models. Mol Cancer Res 2019; 17:1931-1944. [DOI: 10.1158/1541-7786.mcr-18-0923] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 03/14/2019] [Accepted: 05/28/2019] [Indexed: 11/16/2022]
|
14
|
Okubo K, Isono M, Asano T, Sato A. Metformin Augments Panobinostat's Anti-Bladder Cancer Activity by Activating AMP-Activated Protein Kinase. Transl Oncol 2019; 12:669-682. [PMID: 30849634 PMCID: PMC6402380 DOI: 10.1016/j.tranon.2019.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 02/09/2023] Open
Abstract
Panobinostat, a histone deacetylase inhibitor, induces histone acetylation and acts against cancer but attenuates its anticancer activity by activating the mammalian target of rapamycin (mTOR) pathway. AMP-activated protein kinase (AMPK) is a cellular energy sensor that reportedly inhibits the mTOR pathway. The antidiabetic drug metformin is also a potent AMPK activator and we investigated whether it augmented panobinostat's antineoplastic activity in bladder cancer cells (UMUC3, J82, T24 and MBT-2). Metformin enhanced panobinostat-induced apoptosis and the combination inhibited the growth of bladder cancer cells cooperatively in vitro and in vivo. As expected, metformin increased the phosphorylation of AMPK and decreased the panobinostat-caused phosphorylation of S6 ribosomal protein, thus inhibiting the panobinostat-activated mTOR pathway. The AMPK activation was shown to play a pivotal role in the combination's action because the AMPK inhibitor compound C attenuated the combination's anticancer activity. Furthermore, the AMPK activation by metformin enhanced panobinostat-induced histone and non-histone acetylation. This acetylation was especially remarkable in the proteins in the detergent-insoluble fraction, which would be expected if the combination also induced endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Kazuki Okubo
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Makoto Isono
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Takako Asano
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Akinori Sato
- Department of Urology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| |
Collapse
|
15
|
Svatek RS, Ji N, de Leon E, Mukherjee NZ, Kabra A, Hurez V, Nicolas M, Michalek JE, Javors M, Wheeler K, Sharp ZD, Livi CB, Shu ZJ, Henkes D, Curiel TJ. Rapamycin Prevents Surgery-Induced Immune Dysfunction in Patients with Bladder Cancer. Cancer Immunol Res 2019; 7:466-475. [PMID: 30563829 PMCID: PMC6926429 DOI: 10.1158/2326-6066.cir-18-0336] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/18/2018] [Accepted: 12/10/2018] [Indexed: 11/16/2022]
Abstract
The mechanistic target of rapamycin (mTOR) integrates environmental inputs to regulate cellular growth and metabolism in tumors. However, mTOR also regulates T-cell differentiation and activation, rendering applications of mTOR inhibitors toward treating cancer complex. Preclinical data support distinct biphasic effects of rapamycin, with higher doses directly suppressing tumor cell growth and lower doses enhancing T-cell immunity. To address the translational relevance of these findings, the effects of the mTOR complex 1 (mTORC1) inhibitor, rapamycin, on tumor and T cells were monitored in patients undergoing cystectomy for bladder cancer. MB49 syngeneic murine bladder cancer models were tested to gain mechanistic insights. Surgery-induced T-cell exhaustion in humans and mice and was associated with increased pulmonary metastasis and decreased PD-L1 antibody efficacy in mouse bladder cancer. At 3 mg orally daily, rapamycin concentrations were 2-fold higher in bladder tissues than in blood. Rapamycin significantly inhibited tumor mTORC1, shown by decreased rpS6 phosphorylation in treated versus control patients (P = 0.008). Rapamycin reduced surgery-induced T-cell exhaustion in patients, evidenced by a significant decrease in the prevalence of dysfunctional programmed death-1 (PD-1)-expressing T cells. Grade 3 to 4 adverse event rates were similar between groups, but rapamycin-treated patients had a higher rate of wound complications versus controls. In conclusion, surgery promoted bladder cancer metastasis and decreased the efficacy of postoperative bladder cancer immunotherapy. Low-dose (3 mg daily) oral rapamycin has favorable pharmacodynamic and immune modulating activity in surgical patients and has the potential to decrease surgery-induced immune dysfunction.
Collapse
Affiliation(s)
- Robert S Svatek
- Experimental Developmental Therapeutics (EDT) Program, UT Health MD Anderson, San Antonio, Texas.
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | - Niannian Ji
- Experimental Developmental Therapeutics (EDT) Program, UT Health MD Anderson, San Antonio, Texas
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | - Essel de Leon
- Department of Pathology, UT Health San Antonio, San Antonio, Texas
| | - Neelam Z Mukherjee
- Experimental Developmental Therapeutics (EDT) Program, UT Health MD Anderson, San Antonio, Texas
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | - Aashish Kabra
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | - Vincent Hurez
- Experimental Developmental Therapeutics (EDT) Program, UT Health MD Anderson, San Antonio, Texas
| | - Marlo Nicolas
- Department of Pathology, UT Health San Antonio, San Antonio, Texas
| | - Joel E Michalek
- Department of Epidemiology and Biostatistics, UT Health San Antonio, San Antonio, Texas
| | - Martin Javors
- Department of Psychiatry, UT Health San Antonio, San Antonio, Texas
| | - Karen Wheeler
- Experimental Developmental Therapeutics (EDT) Program, UT Health MD Anderson, San Antonio, Texas
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | - Z Dave Sharp
- The Population Science and Prevention (PSP) Program, Mays Cancer Center at UT Health MD Anderson, San Antonio, Texas
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio
| | - Carolina B Livi
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas
- Agilent Technologies, Santa Clara, California
| | - Zhen-Ju Shu
- Experimental Developmental Therapeutics (EDT) Program, UT Health MD Anderson, San Antonio, Texas
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | - David Henkes
- Department of Pathology, CHRISTUS Santa Rosa Medical Center, San Antonio, Texas
| | - Tyler J Curiel
- Experimental Developmental Therapeutics (EDT) Program, UT Health MD Anderson, San Antonio, Texas.
- Division of Hematology/Medical Oncology at the UT Health San Antonio, San Antonio, Texas
| |
Collapse
|
16
|
Tian T, Li X, Zhang J. mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy. Int J Mol Sci 2019; 20:ijms20030755. [PMID: 30754640 PMCID: PMC6387042 DOI: 10.3390/ijms20030755] [Citation(s) in RCA: 383] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) pathway plays a crucial role in regulation of cell survival, metabolism, growth and protein synthesis in response to upstream signals in both normal physiological and pathological conditions, especially in cancer. Aberrant mTOR signaling resulting from genetic alterations from different levels of the signal cascade is commonly observed in various types of cancers. Upon hyperactivation, mTOR signaling promotes cell proliferation and metabolism that contribute to tumor initiation and progression. In addition, mTOR also negatively regulates autophagy via different ways. We discuss mTOR signaling and its key upstream and downstream factors, the specific genetic changes in the mTOR pathway and the inhibitors of mTOR applied as therapeutic strategies in eight solid tumors. Although monotherapy and combination therapy with mTOR inhibitors have been extensively applied in preclinical and clinical trials in various cancer types, innovative therapies with better efficacy and less drug resistance are still in great need, and new biomarkers and deep sequencing technologies will facilitate these mTOR targeting drugs benefit the cancer patients in personalized therapy.
Collapse
Affiliation(s)
- Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Xiaoyi Li
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| |
Collapse
|
17
|
Liu Y, Li B, Yang X, Zhang C. MiR-99a-5p inhibits bladder cancer cell proliferation by directly targeting mammalian target of rapamycin and predicts patient survival. J Cell Biochem 2018; 120:19330-19337. [PMID: 30560585 DOI: 10.1002/jcb.27318] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/27/2018] [Indexed: 12/14/2022]
Abstract
Bladder cancer is a common malignancy and miR-99a-5p has been reported to be downregulated in bladder cancer, but its function and the underlying mechanism in bladder cancer development remains largely unclear. Here, we report that miR-99a-5p expression was decreased in bladder cancer compared with the adjacent normal tissues. Receiver operating characteristic curve revealed that miR-99a-5p expression signature had area under curve value of 0.7989 in differing bladder cancer from the adjacent normal tissues. Bladder cancer patients with low expression of miR-99a-5p had a poor survival rate. Gain-of-function and loss-of-function approaches demonstrated that miR-99a-5p inhibited bladder cell proliferation and cell cycle. Furthermore, we identified that mammalian target of rapamycin (mTOR) was a direct target of miR-99a-5p and mTOR restore could rescue the proliferative ability of bladder cancer cells. Moreover, miR-99a-5p/mTOR axis regulated S6K1 phosphorylation. These suggested that miR-99a-5p/mTOR axis might be a therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Yan Liu
- Department of Urinary Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Bingxun Li
- Department of Urinary Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xianxu Yang
- Department of Urinary Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chenglong Zhang
- Department of Urinary Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
18
|
Shetty AK, Kodali M, Upadhya R, Madhu LN. Emerging Anti-Aging Strategies - Scientific Basis and Efficacy. Aging Dis 2018; 9:1165-1184. [PMID: 30574426 PMCID: PMC6284760 DOI: 10.14336/ad.2018.1026] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
The prevalence of age-related diseases is in an upward trend due to increased life expectancy in humans. Age-related conditions are among the leading causes of morbidity and death worldwide currently. Therefore, there is an urgent need to find apt interventions that slow down aging and reduce or postpone the incidence of debilitating age-related diseases. This review discusses the efficacy of emerging anti-aging approaches for maintaining better health in old age. There are many anti-aging strategies in development, which include procedures such as augmentation of autophagy, elimination of senescent cells, transfusion of plasma from young blood, intermittent fasting, enhancement of adult neurogenesis, physical exercise, antioxidant intake, and stem cell therapy. Multiple pre-clinical studies suggest that administration of autophagy enhancers, senolytic drugs, plasma from young blood, drugs that enhance neurogenesis and BDNF are promising approaches to sustain normal health during aging and also to postpone age-related neurodegenerative diseases such as Alzheimer's disease. Stem cell therapy has also shown promise for improving regeneration and function of the aged or Alzheimer's disease brain. Several of these approaches are awaiting critical appraisal in clinical trials to determine their long-term efficacy and possible adverse effects. On the other hand, procedures such as intermittent fasting, physical exercise, intake of antioxidants such as resveratrol and curcumin have shown considerable promise for improving function in aging, some of which are ready for large-scale clinical trials, as they are non-invasive, and seem to have minimal side effects. In summary, several approaches are at the forefront of becoming mainstream therapies for combating aging and postponing age-related diseases in the coming years.
Collapse
Affiliation(s)
- Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
| |
Collapse
|
19
|
Zhao YY, Tian Y, Liu L, Zhan JH, Hou X, Chen X, Zhou T, Huang Y, Zhang L. Inhibiting eEF-2 kinase-mediated autophagy enhanced the cytocidal effect of AKT inhibitor on human nasopharyngeal carcinoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2655-2663. [PMID: 30214154 PMCID: PMC6120571 DOI: 10.2147/dddt.s169952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aim Our previous research showed that AKT inactivation via small molecule inhibitors did not induce significant apoptosis, but rather markedly increased autophagy in nasopharyngeal carcinoma (NPC). The purpose of the current study was to determine whether autophagy inhibition can enhance the anticancer efficacy of an AKT inhibitor (MK-2206). Materials and methods NPC cell lines CNE-2 (Epstein–Barr virus negative) and C666-1 (Epstein–Barr virus positive) were used to conduct the research. Autophagy induction effects were evaluated via Western blotting. Eukaryotic elongation factor-2 (eEF-2) kinase was specifically and stably knocked down using shRNA. The growth and proliferation of the cells were assessed by Cell Counting Kit-8. In CNE-2 xenograft tumors, the antitumor effects of an AKT inhibitor (MK-2206) combined with an eEF-2 kinase inhibitor (NH125) were tested. Results MK-2206 induced eEF-2 kinase-dependent autophagy in NPC cell lines. Knockdown of eEF-2 kinase using shRNA blunted the autophagy activated by MK-2206. Compared with treatment with MK-2206 alone, shRNA or NH125 suppressed eEF-2 kinase and increased the growth-inhibitory effect of MK-2206 on the human NPC cell lines. The synergistic effects of eEF-2 kinase inhibition and MK-2206 were similar to those of the combination of hydroxychloroquine and MK-2206. Moreover, NH125 showed good synergistic effects with MK-2206 in vivo. Conclusion eEF-2 kinase-mediated autophagy induced by AKT inhibition played a protective role in NPC cells. Inhibition of eEF-2 kinase may be an effective method for increasing the efficacy of an AKT inhibitor such as MK-2206 in NPC.
Collapse
Affiliation(s)
- Yuan-Yuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| | - Ying Tian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| | - Lin Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China, .,Zhaoqing Medical College, Zhaoqing, Guangdong Province, People's Republic of China
| | - Jian-Hua Zhan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| | - Xue Hou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| | - Xi Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| | - Ting Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China,
| |
Collapse
|
20
|
Richard PO, Ahmad AE, Bashir S, Zlotta A, Bhindi B, Leao R, Nayan M, Mohammed A, Fleshner NE, Kulkarni GS. Impact of oral hypoglycemic agents on mortality among diabetic patients with non-muscle-invasive bladder cancer: A populationbased analysis. Can Urol Assoc J 2018; 12:203-210. [PMID: 29485035 DOI: 10.5489/cuaj.4870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Non-muscle-invasive bladder cancer (NMIBC) accounts for 75-85% of all urothelial bladder cancers (UBC). Many UBC patients are also afflicted by diabetes mellitus (DM). It has been postulated that several oral hypoglycemic agents could impact disease-specific survival (DSS), but the data are sparse among NMIBC patients. Our primary objective was to evaluate the impact of metformin on DSS and overall survival (OS) in NMIBC patients. METHODS This is a retrospective, population-based study that used linked administrative databases to identify diabetic patients ≥66 years who were subsequently diagnosed with NMIBC in Ontario between 1992 and 2012. Cumulative use of metformin and other hypoglycemic agent were calculated before and after NMIBC diagnosis. DSS and OS were estimated using multivariable competing risk and Cox proportional hazards models, respectively. RESULTS A total of 1742 subjects were included in the study. After a median followup of 5.2 years, 1122 (64%) had died, including 247 (15%) deaths as a result of UBC. On multivariable analysis, cumulative duration of metformin use after NMIBC diagnosis did not appear to impact DSS (hazard ratio [HR] 1.1; 95% confidence interval [CI] 0.92-1.2), whereas glyburide use appeared to have a detrimental effect (HR 1.17; 95% CI 1.02-1.3). None of the other hypoglycemic agents had an impact on OS. CONCLUSIONS In this large, population-based study, we have provided further evidence that metformin use does not significantly impact DSS among diabetic patients diagnosed with NMIBC. However, our findings demonstrate that glyburide use inversely affects DSS. The detrimental effect of glyburide on DSS will require further validation.
Collapse
Affiliation(s)
- Patrick O Richard
- Division of Urology, Departments of Surgery, Centre Hospitalier Universitaire de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, QC; Canada
| | - Ardalan E Ahmad
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada
| | - Shaheena Bashir
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada
| | - Alexandre Zlotta
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada.,Division of Urology, Departments of Surgery, Mount Sinai Hospital and the University of Toronto, Toronto, ON; Canada
| | - Bimal Bhindi
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada
| | - Ricardo Leao
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada
| | - Madhur Nayan
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada
| | - Aza Mohammed
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada
| | - Neil E Fleshner
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and the University of Toronto, Toronto, ON; Canada
| | - Girish S Kulkarni
- Division of Urology, Departments of Surgery and 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
| |
Collapse
|
21
|
Abstract
The PI3K/AKT/mTOR signaling pathway shows frequent molecular alterations and increased activity in cancer. Given its role in the regulation of cell growth, survival and metastasis, molecules within this pathway are promising targets for pharmacologic intervention. Metastatic bladder cancer (BLCA) continues to have few treatment options. Although various molecular alterations in PI3K/AKT/mTOR signaling have been described in BLCA, clinical trials with small molecule inhibitors have not met their endpoints. In this article, we summarize results from preclinical studies and clinical trials that examined PI3K pathway inhibitors in BLCA focusing on technical challenges that might result in contradictory findings in preclinical studies. Based on published data from our group, we also address challenges that need to be overcome to optimize PI3K inhibition in BLCA and enable its successful translation into the clinic.
Collapse
Affiliation(s)
- Anuja Sathe
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
22
|
RAPTOR gene polymorphism is independently correlated with urothelial cancer susceptibility compared with environmental toxin exposure. UROLOGICAL SCIENCE 2017. [DOI: 10.1016/j.urols.2016.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
23
|
Dalbagni G, Benfante N, Sjoberg DD, Bochner BH, Machele Donat S, Herr HW, Mc Coy AS, Fahrner AJ, Retinger C, Rosenberg JE, Bajorin DF. Single Arm Phase I/II Study of Everolimus and Intravesical Gemcitabine in Patients with Primary or Secondary Carcinoma In Situ of the Bladder who failed Bacillus Calmette Guerin (NCT01259063). Bladder Cancer 2017; 3:113-119. [PMID: 28516156 PMCID: PMC5409047 DOI: 10.3233/blc-170095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Standard treatment for BCG-refractory urothelial cancer is radical cystectomy. Identification of active agents is clearly warranted. Objective: To determine a safe dose of oral everolimus in combination with standard intravesical gemcitabine and to evaluate the efficacy of this combination. Methods: Patients with carcinoma in situ refractory to intravesical bacillus Calmette-Guérin and refusing cystectomy were eligible. Patients in the phase I part of the trial received one of three dose levels of oral everolimus. Patients also received a fixed dose of intravesical gemcitabine. Maintenance everolimus was given for 12 months in patients achieving a complete response confirmed by cystoscopy and cytology. Patients in phase II received continuous everolimus administered at 10 mg daily with intravesical gemcitabine followed by everolimus maintenance for 12 months of total therapy. The enrollment goal for the phase II was 33 patients. Results: 14 patients were enrolled in phase I of the trial. 23 patients were enrolled in phase II of the trial and 19 were evaluable for primary and secondary endpoints. Four patients withdrew consent prior to treatment initiation. Of the 19 patients evaluable for response, 3 (16%, 95% confidence interval [CI] 3% – 40%) were disease free at 1 yr. The probability of RFS was 20% (95% CI 5% – 42%) at 12 months. Ten patients out of 19 had grade 3 or greater toxicity events. Seven withdrew consent or were taken off study. Conclusions: Many patients withdrew, and enrollment was halted. Continuous oral everolimus plus intravesical gemcitabine was not well tolerated in this patient population where the threshold for tolerability is low.
Collapse
Affiliation(s)
- Guido Dalbagni
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicole Benfante
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel D Sjoberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bernard H Bochner
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Machele Donat
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harry W Herr
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Asia S Mc Coy
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alicia J Fahrner
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caitlyn Retinger
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan E Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dean F Bajorin
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
24
|
Hau AM, Nakasaki M, Nakashima K, Krish G, Hansel DE. Differential mTOR pathway profiles in bladder cancer cell line subtypes to predict sensitivity to mTOR inhibition. Urol Oncol 2017; 35:593-599. [PMID: 28427860 DOI: 10.1016/j.urolonc.2017.03.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Molecular classification of bladder cancer has been increasingly proposed as a potential tool to predict clinical outcomes and responses to chemotherapy. Here we focused on mechanistic target of rapamycin (mTOR) inhibition as a chemotherapeutic strategy and characterized the expression profile of mTOR signaling targets in representative bladder cancer cell lines from basal, luminal, and either basal/luminal ("non-type") molecular subtypes. MATERIALS AND METHODS Protein and mRNA expression of mTOR signaling components from representative luminal (RT4 and RT112), basal (SCaBER and 5637), and nontype (T24 and J82) bladder cancer cell line subtypes were determined by Western blot and database mining analysis of the Cancer Cell Line Encyclopedia. Cell viability following treatment with either, Torin-2 or KU-0063794, 2 dual mTOR complex 1/2 inhibitors, was determined by MTT assay. Immunoblot analysis of cells treated with Torin-2 or KU-0063794 was performed to determine the effects of mTOR inhibition on expression and phosphorylation status of mTOR signaling components, Akt, 4E-BP1, and ribosomal protein S6. RESULTS Molecular subtypes of bladder cancer cell lines each exhibited a distinct pattern of expression of mTOR-associated genes and baseline phosphorylation level of Akt and 4E-BP1. Cells with low levels of Akt Ser-473 phosphorylation were more resistant to the cytotoxic effects of mTOR inhibition with Torin-2, but not KU-0063794. Exposure to Torin-2 and KU-0063794 both potently and rapidly inhibited phosphorylation of Akt Ser-473 and Thr-308, and 4E-BP1 T37/46 in cell lines that included basal and nontype subtypes. CONCLUSIONS Differential gene expression and protein activity associated with mTOR signaling is observed among bladder cancer cell lines stratified into basal, luminal, and nontype subtypes. Urothelial carcinomas characterized by high baseline Akt Ser-473 phosphorylation may be best suited for targeted mTOR therapies.
Collapse
Affiliation(s)
- Andrew M Hau
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - Manando Nakasaki
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - Kazufumi Nakashima
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - Goutam Krish
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - Donna E Hansel
- Department of Pathology, University of California, San Diego, La Jolla, CA; Department of Urology, University of California, San Diego, La Jolla, CA.
| |
Collapse
|
25
|
Simultaneous Targeting of Bladder Tumor Growth, Survival, and Epithelial-to-Mesenchymal Transition with a Novel Therapeutic Combination of Acetazolamide (AZ) and Sulforaphane (SFN). Target Oncol 2017; 11:209-27. [PMID: 26453055 DOI: 10.1007/s11523-015-0386-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Current chemotherapies for advanced stage metastatic bladder cancer often result in severe side effects, and most patients become drug resistant over time. Thus, there is a need for more effective therapies with minimal side effects. OBJECTIVE The acid/base balance in tumor cells is essential for tumor cell functioning. We reasoned that simultaneous targeting of pH homeostasis and survival pathways would improve therapeutic efficacy. We evaluated the effectiveness of targeting pH homeostasis with the carbonic anhydrase inhibitor acetazolamide (AZ) in combination with the survival pathway targeting isothiocyanate sulforaphane (SFN) on the HTB-9 and RT112(H) human bladder tumor cell lines. MATERIALS AND METHODS We assessed viability, proliferation, and survival in vitro and effect on xenografts in vivo. RESULTS Combination AZ + SFN treatment induced dose-dependent suppression of growth, produced a potent anti-proliferative and anti-clonogenic effect, and induced apoptosis through caspase-3 and PARP activation. The anti-proliferative effect was corroborated by significant reductions in Ki-67, pHH3, cyclin D1, and sustained induction of the cell cycle inhibitors, p21 and p27. Both active p-Akt (Ser473) and p-S6 were significantly downregulated in the AZ + SFN combination treated cells with a concomitant inhibition of Akt kinase activity. The inhibitory effects of the AZ + SFN combination treatment showed similar efficacy as the dual PI3K/mTOR pathway inhibitor NVP-BEZ235, albeit at an expected higher dose. In terms of the effect on the metastatic potential of these bladder cancers, we found downregulated expression of carbonic anhydrase 9 (CA9) concomitant with reductions in both E-cadherin, N-cadherin, and vimentin proteins mitigating the epithelial-to-mesenchymal transition (EMT), suggesting negation of this program. CONCLUSION We suggest that reductions in these components could be linked with downregulation of the survival mediated Akt pathway and suggested an active role of the Akt pathway in bladder cancer. Altogether, our in vitro and pre-clinical model data support the potential use of an AZ + SFN combination for the treatment of bladder cancer.
Collapse
|
26
|
Alayev A, Salamon RS, Schwartz NS, Berman AY, Wiener SL, Holz MK. Combination of Rapamycin and Resveratrol for Treatment of Bladder Cancer. J Cell Physiol 2016; 232:436-446. [DOI: 10.1002/jcp.25443] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/24/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Anya Alayev
- Department of Biology; Yeshiva University; New York New York
| | | | | | - Adi Y. Berman
- Department of Biology; Yeshiva University; New York New York
| | - Sara L. Wiener
- Department of Biology; Yeshiva University; New York New York
| | - Marina K. Holz
- Department of Biology; Yeshiva University; New York New York
- Department of Molecular Pharmacology, Albert Einstein Cancer Center; Albert Einstein College of Medicine; Bronx New York
| |
Collapse
|
27
|
Pinto-Leite R, Arantes-Rodrigues R, Sousa N, Oliveira PA, Santos L. mTOR inhibitors in urinary bladder cancer. Tumour Biol 2016; 37:11541-11551. [PMID: 27235118 DOI: 10.1007/s13277-016-5083-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/15/2016] [Indexed: 02/07/2023] Open
Abstract
Despite the great scientific advances that have been made in cancer treatment, there is still much to do, particularly with regard to urinary bladder cancer. Some of the drugs used in urinary bladder cancer treatment have been in use for more than 30 years and show reduced effectiveness and high recurrence rates. There have been several attempts to find new and more effective drugs, to be used alone or in combination with the drugs already in use, in order to overcome this situation.The biologically important mammalian target of rapamycin (mTOR) pathway is altered in cancer and mTOR inhibitors have raised many expectations as potentially important anticancer drugs. In this article, the authors will review the mTOR pathway and present their experiences of the use of some mTOR inhibitors, sirolimus, everolimus and temsirolimus, in isolation and in conjunction with non-mTOR inhibitors cisplatin and gemcitabine, on urinary bladder tumour cell lines. The non-muscle-invasive cell line, 5637, is the only one that exhibits a small alteration in the mTOR and AKT phosphorylation after rapalogs exposure. Also, there was a small inhibition of cell proliferation. With gemcitabine plus everolimus or temsirolimus, the results were encouraging as a more effective response was noticed with both combinations, especially in the 5637 and T24 cell lines. Cisplatin associated with everolimus or temsirolimus also gave promising results, as an antiproliferative effect was observed when the drugs were associated, in particular on the 5637 and HT1376 cell lines. Everolimus or temsirolimus in conjunction with gemcitabine or cisplatin could have an important role to play in urinary bladder cancer treatment, depending on the tumour grading.
Collapse
Affiliation(s)
- R Pinto-Leite
- Genetic Service, Cytogenetic Laboratory, Hospital Center of Trás-os-Montes and Alto Douro, Vila Real, Portugal. .,Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.
| | - R Arantes-Rodrigues
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,QOPNA, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, Aveiro, Portugal.,Institute for Research and Innovation in Health (I3S), Porto, Portugal
| | - Nuno Sousa
- Health School, University Fernando Pessoa, Porto, Portugal
| | - P A Oliveira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal
| | - L Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal.,Health School, University Fernando Pessoa, Porto, Portugal.,Medical Oncology Department, Portuguese Institute of Oncology, Porto, Portugal
| |
Collapse
|
28
|
Lin YC, Lin JF, Wen SI, Yang SC, Tsai TF, Chen HE, Chou KY, Hwang TIS. Inhibition of High Basal Level of Autophagy Induces Apoptosis in Human Bladder Cancer Cells. J Urol 2015; 195:1126-35. [PMID: 26519656 DOI: 10.1016/j.juro.2015.10.128] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2015] [Indexed: 02/04/2023]
Abstract
PURPOSE Cancer cells adapt to stress by activation of the autophagy pathway primed for survival. A high basal level of autophagic activity was found in human bladder cancer cell lines. We studied the significance of the phenomenon on cancer cell survival. MATERIALS AND METHODS The immortalized human bladder epithelial cell line SV-HUC-1 and the human bladder cancer cell lines RT-4 and 5637 together with human bladder cancer specimens collected from patients were used. A commercially available bladder cancer microarray was applied to confirm the findings. LC3 (light chain-3) II protein detection was done to determine the presence of autophagy. Caspase 3 and DNA fragmentation was performed to detect apoptosis. RESULTS Bladder cancer cell lines showed activated autophagic flux compared to SV-HUC-1 cells, prostate cancer cells and breast cancer cells. Results were confirmed in human bladder cancer specimens. Autophagy inhibition by Baf (bafilomycin) A1, or by knockdown of ATG (autophagy related protein) 7 or 12 induced cytotoxicity in multiple human bladder cell lines. Induction of apoptosis was found in cells with autophagy inhibition. Although the disruption of mitochondria membrane potential or the generation of reactive oxygen species was detected in Baf A1 treated cells, intensity was mild and not thought to be related to apoptosis of bladder cancer cells. CONCLUSIONS Our results indicate that autophagy is required for the growth and survival of human bladder cancer cells.
Collapse
Affiliation(s)
- Yi-Chia Lin
- Division of Urology, Department of Surgery, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China; School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Ji-Fan Lin
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China
| | - Sheng-I Wen
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China
| | - Shan-Che Yang
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China
| | - Te-Fu Tsai
- Division of Urology, Department of Surgery, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China; School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Hung-En Chen
- Division of Urology, Department of Surgery, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China
| | - Kuang-Yu Chou
- Division of Urology, Department of Surgery, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China; School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Thomas I-Sheng Hwang
- Division of Urology, Department of Surgery, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China; Department of Urology, Taipei Medical University, Taipei, Taiwan, Republic of China; School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, Republic of China.
| |
Collapse
|
29
|
Earl J, Rico D, Carrillo-de-Santa-Pau E, Rodríguez-Santiago B, Méndez-Pertuz M, Auer H, Gómez G, Grossman HB, Pisano DG, Schulz WA, Pérez-Jurado LA, Carrato A, Theodorescu D, Chanock S, Valencia A, Real FX. The UBC-40 Urothelial Bladder Cancer cell line index: a genomic resource for functional studies. BMC Genomics 2015; 16:403. [PMID: 25997541 PMCID: PMC4470036 DOI: 10.1186/s12864-015-1450-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 03/09/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Urothelial bladder cancer is a highly heterogeneous disease. Cancer cell lines are useful tools for its study. This is a comprehensive genomic characterization of 40 urothelial bladder carcinoma (UBC) cell lines including information on origin, mutation status of genes implicated in bladder cancer (FGFR3, PIK3CA, TP53, and RAS), copy number alterations assessed using high density SNP arrays, uniparental disomy (UPD) events, and gene expression. RESULTS Based on gene mutation patterns and genomic changes we identify lines representative of the FGFR3-driven tumor pathway and of the TP53/RB tumor suppressor-driven pathway. High-density array copy number analysis identified significant focal gains (1q32, 5p13.1-12, 7q11, and 7q33) and losses (i.e. 6p22.1) in regions altered in tumors but not previously described as affected in bladder cell lines. We also identify new evidence for frequent regions of UPD, often coinciding with regions reported to be lost in tumors. Previously undescribed chromosome X losses found in UBC lines also point to potential tumor suppressor genes. Cell lines representative of the FGFR3-driven pathway showed a lower number of UPD events. CONCLUSIONS Overall, there is a predominance of more aggressive tumor subtypes among the cell lines. We provide a cell line classification that establishes their relatedness to the major molecularly-defined bladder tumor subtypes. The compiled information should serve as a useful reference to the bladder cancer research community and should help to select cell lines appropriate for the functional analysis of bladder cancer genes, for example those being identified through massive parallel sequencing.
Collapse
Affiliation(s)
- Julie Earl
- Epithelial Carcinogenesis Group, F BBVA Cancer Cell Biology Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain. .,Servicio de Oncología Médica, Hospital Ramón y Cajal, Madrid, Spain.
| | - Daniel Rico
- Structural Computational Biology Group, Structural Biology and Biocomputing Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain.
| | - Enrique Carrillo-de-Santa-Pau
- Epithelial Carcinogenesis Group, F BBVA Cancer Cell Biology Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain.
| | - Benjamín Rodríguez-Santiago
- Quantitative Genomic Medicine Laboratory, qGenomics, Barcelona, Spain. .,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.
| | - Marinela Méndez-Pertuz
- Epithelial Carcinogenesis Group, F BBVA Cancer Cell Biology Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain.
| | - Herbert Auer
- Institut de Recerca Biomèdica de Barcelona, Parc Científic de Barcelona, Barcelona, Spain.
| | - Gonzalo Gómez
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain.
| | | | - David G Pisano
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain.
| | - Wolfgang A Schulz
- Department of Urology, Heinrich-Heine-University, Düsseldorf, Germany.
| | - Luis A Pérez-Jurado
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.
| | - Alfredo Carrato
- Servicio de Oncología Médica, Hospital Ramón y Cajal, Madrid, Spain.
| | - Dan Theodorescu
- University of Colorado Comprehensive Cancer Center, 80045, Aurora, CO, USA.
| | - Stephen Chanock
- Translational Genomics Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA.
| | - Alfonso Valencia
- Structural Computational Biology Group, Structural Biology and Biocomputing Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain.
| | - Francisco X Real
- Epithelial Carcinogenesis Group, F BBVA Cancer Cell Biology Programme, CNIO (Spanish National Cancer Research Centre), Madrid, Spain. .,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain. .,Cancer Cell Biology Programme, Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
30
|
Matsushima M, Kikuchi E, Matsumoto K, Hattori S, Takeda T, Kosaka T, Miyajima A, Oya M. Intravesical dual PI3K/mTOR complex 1/2 inhibitor NVP-BEZ235 therapy in an orthotopic bladder cancer model. Int J Oncol 2015; 47:377-83. [PMID: 25963317 DOI: 10.3892/ijo.2015.2995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/14/2015] [Indexed: 01/08/2023] Open
Abstract
NVP-BEZ235 is an inhibitor of both phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin complex 1/2 (mTORC1/2), and its antitumor activity is expected to be higher than that of mTORC1 inhibitors because it inhibits the upregulation of pAkt through mTORC2. We examined the efficacy of intravesical NVP-BEZ235 therapy in the treatment of bladder cancer using an orthotopic bladder cancer model. The cytotoxic effects of various concentrations of NVP-BEZ235 in MBT-2 cells were examined using a WST assay. The expression of pAkt, pS6 and p4EBP1 was evaluated in MBT-2 cells treated with NVP-BEZ235 using western blotting. Orthotopic models were established by implanting MBT-2 cells into the bladders of female C3H/He mice. We assigned C3H/He mice to 2 groups: a control group treated with vehicle control (n=15), and a group intravesically administered 40 µM (18.78 mg/l) of NVP-BEZ235 (n=15). NVP-BEZ235 inhibited the viability of MBT-2 cells in a dose-dependent manner. Furthermore, the expression of pAkt, pS6, and p4EBP1 was inhibited in NVP-BEZ235-treated MBT-2 cells. Bladder weights were significantly lower in the NVP-BEZ235-treated group than in the control group (P<0.05). An analysis of the tumor tissues revealed that the NVP-BEZ235 treatment strongly reduced pAkt, pS6 and p4EBP1 levels. An immunohistochemical analysis showed that NVP-BEZ235 significantly inhibited the expression of pS6. Intravesically administered NVP-BEZ235 exerted significant antitumor effects in the orthotopic bladder cancer model by inhibiting the PI3K/Akt/mTOR pathway. The intravesical instillation of a dual PI3K/mTORC1/2 inhibitor may represent a novel therapy for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Masashi Matsushima
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Eiji Kikuchi
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuhiro Matsumoto
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Seiya Hattori
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshikazu Takeda
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Akira Miyajima
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
31
|
Combined inhibition of AKT/mTOR and MDM2 enhances Glioblastoma Multiforme cell apoptosis and differentiation of cancer stem cells. Sci Rep 2015; 5:9956. [PMID: 25898313 PMCID: PMC4404683 DOI: 10.1038/srep09956] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/24/2015] [Indexed: 02/07/2023] Open
Abstract
The poor prognosis of Glioblastoma Multiforme (GBM) is due to a high resistance to conventional treatments and to the presence of a subpopulation of glioma stem cells (GSCs). Combination therapies targeting survival/self-renewal signals of GBM and GSCs are emerging as useful tools to improve GBM treatment. In this context, the hyperactivated AKT/mammalian target of the rapamycin (AKT/mTOR) and the inhibited wild-type p53 appear to be good candidates. Herein, the interaction between these pathways was investigated, using the novel AKT/mTOR inhibitor FC85 and ISA27, which re-activates p53 functionality by blocking its endogenous inhibitor murine double minute 2 homologue (MDM2). In GBM cells, FC85 efficiently inhibited AKT/mTOR signalling and reactivated p53 functionality, triggering cellular apoptosis. The combined therapy with ISA27 produced a synergic effect on the inhibition of cell viability and on the reactivation of p53 pathway. Most importantly, FC85 and ISA27 blocked proliferation and promoted the differentiation of GSCs. The simultaneous use of these compounds significantly enhanced GSC differentiation/apoptosis. These findings suggest that FC85 actively enhances the downstream p53 signalling and that a combination strategy aimed at inhibiting the AKT/mTOR pathway and re-activating p53 signalling is potentially effective in GBM and in GSCs.
Collapse
|
32
|
Abstract
Metastatic bladder cancer is a lethal disease. Cisplatin-based chemotherapy, including the combination regimens gemcitabine-cisplatin and methotrexate-vinblastine-doxorubicin-cisplatin, are the standard first-line therapies. Second-line therapies have modest activity and no significant improvement in patient outcomes. Agents targeting growth, survival, and proliferation pathways have been added to cytotoxic therapy with limited added benefit to date. Modulating host immune response to cancer-associated antigens appears promising, with multiple new therapeutic approaches being pursued. Next-generation sequencing of invasive urothelial carcinoma has provided insights into the biology of this disease and potential actionable targets. Alterations in the receptor tyrosine kinase/Ras pathway and the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin pathway represent potential therapeutic targets in advanced disease, and novel agents are in development. Recent data from the Cancer Genome Atlas Research Network bladder cancer cohort and other efforts suggest that mutations in chromatin-regulatory genes are very common in invasive bladder tumors, and are more frequent than in other studied tumors. The discovery of new genomic alterations challenges drug development to change the course of this disease.
Collapse
|
33
|
Vashistha V, Quinn DI, Dorff TB, Daneshmand S. Current and recent clinical trials for perioperative systemic therapy for muscle invasive bladder cancer: a systematic review. BMC Cancer 2014; 14:966. [PMID: 25515347 PMCID: PMC4301463 DOI: 10.1186/1471-2407-14-966] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/11/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Although Muscle Invasive Bladder Cancer (MIBC) is increasing in incidence, treatment has largely remained limited to radical cystectomy with or without cisplatin-based neoadjuvant and/or adjuvant chemotherapy. We reviewed the current and recent clinical trials evaluating perioperative chemotherapy, targeted therapy, and novel therapeutic regimens for MIBC patients undergoing radical cystectomy. METHODS An overview of perioperative MIBC management was conducted initially using MEDLINE. The Clinical Trials Registry and MEDLINE were further searched specifically for perioperative MIBC chemotherapy, targeted therapy, and other novel therapeutic approaches. Trials involving non-perioperative management, operative management other than radical cystectomy, multiple tumors, or purely superficial or metastatic disease were excluded from selection. These criteria were not specifically fulfilled for mTOR inhibitor and immune therapy trials. Only phase III chemotherapy and phase II targeted therapy trials found in the Clinical Trials Registry were selected. MEDLINE searches of specific treatments were limited to January 2009 to January 2014 whereas the Clinical Trials Registry search had no timeline. Systematic MEDLINE searches had no phase restrictions. Trials known by the authors to fulfill search criteria but were not found via searches were also selected. RESULTS Twenty-five trials were selected from the Clinical Trials Registry including 7 phase III chemotherapy trials, 11 Phase II targeted therapy trials, 3 immune therapy trials, 1 mammalian target of rapamycin (mTOR) inhibitor trial, and 3 gene and vaccine therapy trials. Nine trials have been completed and 5 have been terminated early or withdrawn. Nine trials have data available when individually searched using MEDLINE and/or Google. Systematic searches of MEDLINE separately found 12 trials in the past 5 years. Two phase III chemotherapy trials were selected based on knowledge by the authors. No phase III trials of targeted therapy have been registered or published. CONCLUSIONS New trials are currently being conducted that may revolutionize MIBC treatment preceding or following cystectomy. Head-to-head phase III trials of perioperative chemotherapy and further phase II and phase III trials of targeted therapy and other therapeutic approaches are necessary before the current cisplatin-based perioperative chemotherapy paradigm is altered.
Collapse
Affiliation(s)
- Vishal Vashistha
- />Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH USA
| | - David I Quinn
- />Division of Oncology, USC/Norris Comprehensive Cancer Center, USC Institute of Urology, Los Angeles, CA USA
| | - Tanya B Dorff
- />Division of Oncology, USC/Norris Comprehensive Cancer Center, USC Institute of Urology, Los Angeles, CA USA
| | - Siamak Daneshmand
- />Department of Urology, USC/Norris Comprehensive Cancer Center, USC Institute of Urology, 1441 Eastlake Abe, Suite 7416, Los Angeles, CA 90089 USA
| |
Collapse
|
34
|
Li SH, Chen CH, Lu HI, Huang WT, Tien WY, Lan YC, Lee CC, Chen YH, Huang HY, Chang AYW, Lin WC. Phosphorylated p70S6K expression is an independent prognosticator for patients with esophageal squamous cell carcinoma. Surgery 2014; 157:570-80. [PMID: 25726316 DOI: 10.1016/j.surg.2014.10.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 09/22/2014] [Accepted: 10/23/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although marked improvements have been made in surgical technique and chemoradiotherapy, the prognosis for patients with esophageal squamous cell carcinoma (ESCC) is still unsatisfactory. The mammalian target of rapamycin (mTOR) and its downstream signaling, p70 ribosomal S6 protein kinase (p70S6K) and eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1), seem to play central roles in the regulation of cancer cell proliferation and survival. The significance of mTOR and its downstream targets, p70S6K and 4E-BP1, on the prognosis of ESCC remains uncertain, but this pathway is of particular concern because effective inhibitors are already available. METHODS Immunohistochemistry performed to evaluate the expression of phosphorylated mTOR (p-mTOR), phosphorylated p70S6K (p-p70S6K), phosphorylated 4E-binding protein 1 (p-4E-BP1), and Ki-67 using 105 surgically resected ESCC correlated with treatment outcome. The effect of the mTOR signaling pathway inhibitor everolimus on ESCC cell lines were investigated in vitro by the 3-(4.5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and in vivo by a nude mouse xenograft model. RESULTS Univariate analysis showed that p-mTOR overexpression (P = .022), p-p70S6K overexpression (P = .002), and Ki-67 labeling index >50% (P = .045) were associated with inferior overall survival (OS). In a multivariate comparison, p-p70S6K overexpression (P = .001; hazard ratio, 2.247) remained independently associated with worse OS. In cell lines and the xenograft model, everolimus significantly inhibited ESCC growth. CONCLUSION Overexpression of p-p70S6K is associated independently with a poor prognosis among patients with ESCC. The mTOR signaling pathway inhibitor everolimus can inhibit ESCC growth in vitro and in vivo. Our findings suggest that inhibition of mTOR signaling pathway may be a promising novel target for ESCC.
Collapse
Affiliation(s)
- Shau-Hsuan Li
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC.
| | - Chang-Han Chen
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Hung-I Lu
- Department of Thoracic & Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Wan-Ting Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Wan-Yu Tien
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Ya-Chun Lan
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Ching-Chang Lee
- Department of Gastroenterology, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan, ROC
| | - Yen-Hao Chen
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Hsuan-Ying Huang
- Department of Thoracic & Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Alice Y W Chang
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Wei-Che Lin
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital and the Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| |
Collapse
|
35
|
Evaluation of the Prognostic Significance of Altered Mammalian Target of Rapamycin Pathway Biomarkers in Upper Tract Urothelial Carcinoma. Urology 2014; 84:1134-40. [DOI: 10.1016/j.urology.2014.07.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 07/09/2014] [Accepted: 07/29/2014] [Indexed: 11/19/2022]
|
36
|
Mamtani R, Pfanzelter N, Haynes K, Finkelman BS, Wang X, Keefe SM, Haas NB, Vaughn DJ, Lewis JD. Incidence of bladder cancer in patients with type 2 diabetes treated with metformin or sulfonylureas. Diabetes Care 2014; 37:1910-7. [PMID: 24496803 PMCID: PMC4067396 DOI: 10.2337/dc13-1489] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Previous studies evaluating the effect of metformin on cancer risk have been impacted by time-related biases. To avoid these biases, we examined the incidence of bladder cancer in new users of metformin and sulfonylureas (SUs). RESEARCH DESIGN AND METHODS This cohort study included 87,600 patients with type 2 diabetes in The Health Improvement Network database. Use of metformin or an SU was treated as a time-dependent variable. Cox regression-generated hazard ratios (HRs) compared metformin use with SU use, adjusted for age, sex, smoking, obesity, and HbA1c level. RESULTS We identified 196 incident bladder cancers in the metformin cohort and 66 cancers in the SU cohort. Use of metformin was not associated with decreased bladder cancer risk (HR 0.81 [95% CI 0.60-1.09]). This association did not differ by sex (P for interaction = 0.20). We observed no association with duration of metformin relative to SU use (3 to <4 years of use: 0.57 [0.25-1.34]; 4 to <5 years of use: 0.93 [0.30-2.85; ≥5 years of use: 1.18 [0.44-3.19]; P for trend = 0.26). CONCLUSIONS Use of metformin is not associated with a decreased incidence of bladder cancer. Similar methods should be used to study other cancers that have previously been identified as potentially preventable with metformin.
Collapse
Affiliation(s)
- Ronac Mamtani
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Nick Pfanzelter
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Kevin Haynes
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA
| | - Brian S Finkelman
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA
| | - Xingmei Wang
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA
| | - Stephen M Keefe
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Naomi B Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - David J Vaughn
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - James D Lewis
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
37
|
Pinto-Leite R, Arantes-Rodrigues R, Ferreira R, Palmeira C, Oliveira PA, Santos L. Treatment of muscle invasive urinary bladders tumors: A potential role of the mTOR inhibitors. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.biomag.2014.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
38
|
Moon DG, Lee SE, Oh MM, Lee SC, Jeong SJ, Hong SK, Yoon CY, Byun SS, Park HS, Cheon J. NVP-BEZ235, a dual PI3K/mTOR inhibitor synergistically potentiates the antitumor effects of cisplatin in bladder cancer cells. Int J Oncol 2014; 45:1027-35. [PMID: 24969552 PMCID: PMC4121414 DOI: 10.3892/ijo.2014.2505] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/19/2014] [Indexed: 11/28/2022] Open
Abstract
The PI3K/Akt/mTOR pathway is a prototypic survival pathway and constitutively activated in many malignant conditions. Moreover, activation of the PI3K/Akt/mTOR pathway confers resistance to various cancer therapies and is often associated with a poor prognosis. In this study, we explored the antitumor effect of NVP-BEZ235, a dual PI3K/mTOR inhibitor in cisplatin-resistant human bladder cancer cells and its synergistic interaction with cisplatin. A human bladder cancer cell line with cisplatin resistance was exposed to escalating doses of NVP-BEZ235 alone or in combination with cisplatin and antitumor effects was determined by the CCK-8 assay. Based on a dose-response study, synergistic interaction between NVP-BEZ235 and cisplatin was evaluated by combination index (CI), three-dimensional model and clonogenic assay. The combination of NVP-BEZ235 and cisplatin caused significant synergistic antitumor effect in cisplatin-resistant bladder cancer cells over a wide dose range and reduced the IC50 of NVP-BEZ235 and cisplatin by 5.6- and 3.6-fold, respectively. Three-dimensional synergy analysis resulted in a synergy volume of 388.25 μM/ml2% indicating a strong synergistic effect of combination therapy. The combination therapy caused cell cycle arrest and caspase-dependent apoptosis. Although NVP-BEZ235 suppressed PI3K/mTOR signaling without any paradoxical induction of Akt activity, it caused MEK/ERK pathway activation. The present study demonstrated that the PI3K/mTOR dual inhibitor NVP-BEZ235 can synergistically potentiate the antitumor effects of cisplatin in cisplatin-resistant bladder cancer cells though the suppression of cell cycle progression and the survival pathway as well as induction of caspase-dependent apoptosis.
Collapse
Affiliation(s)
- Du G Moon
- Department of Urology, College of Medicine, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Sang E Lee
- Department of Urology, School of Medicine, Seoul National University, Bundang Hospital, Gyeonggi‑do, Seoul, Republic of Korea
| | - Mi M Oh
- Department of Urology, College of Medicine, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Sang C Lee
- Department of Urology, School of Medicine, Seoul National University, Bundang Hospital, Gyeonggi‑do, Seoul, Republic of Korea
| | - Seong J Jeong
- Department of Urology, School of Medicine, Seoul National University, Bundang Hospital, Gyeonggi‑do, Seoul, Republic of Korea
| | - Sung K Hong
- Department of Urology, School of Medicine, Seoul National University, Bundang Hospital, Gyeonggi‑do, Seoul, Republic of Korea
| | - Cheol Y Yoon
- Department of Urology, College of Medicine, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Seok S Byun
- Department of Urology, School of Medicine, Seoul National University, Bundang Hospital, Gyeonggi‑do, Seoul, Republic of Korea
| | - Hong S Park
- Department of Urology, College of Medicine, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Jun Cheon
- Department of Urology, College of Medicine, Korea University, Anam Hospital, Seoul, Republic of Korea
| |
Collapse
|
39
|
Houédé N, Pourquier P. Targeting the genetic alterations of the PI3K-AKT-mTOR pathway: its potential use in the treatment of bladder cancers. Pharmacol Ther 2014; 145:1-18. [PMID: 24929024 DOI: 10.1016/j.pharmthera.2014.06.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 02/07/2023]
Abstract
Urothelial carcinoma of the bladder is the most frequent tumor of the urinary tract and represents the fifth cause of death by cancer worldwide. The current first line chemotherapy is a combination of cisplatin and gemcitabine with median survival not exceeding 15months. Vinflunine is the only drug approved by EMEA as second-line treatment and few progresses have been made for the past 20years to increase the survival of metastatic patients, especially those who are not eligible for cisplatin-based regimen. The recent studies characterizing the genetic background of urothelial cancers of the bladder, revealed chromosomal alterations that are not seen at the same level in other types of cancers. This is especially the case for mutations of genes involved in the PI3K/AKT/mTOR signaling pathway that occupies a major place in the etiology of these tumors. Here, we describe the mutations leading to constitutive activation of the PI3K/AKT/mTOR pathway and discuss the potential use of the different classes of PI3K/AKT/mTOR inhibitors in the treatment of urothelial bladder cancers. Despite the recent pivotal study evidencing specific mutations of TSC1 in bladder cancer patients responding to everolimus and the encouraging results obtained with other derivatives than rapalogs, few clinical trials are ongoing in bladder cancers. Because of the genetic complexity of these tumors, the cross-talks of the PI3K/AKT/mTOR pathway with other pathways, and the small number of eligible patients, it will be of utmost importance to carefully choose the drugs or drug combinations to be further tested in the clinic.
Collapse
Affiliation(s)
- Nadine Houédé
- Service d'Oncologie Médicale, Centre Hospitalier Universitaire de Nîmes, France; INSERM U896, Institut de Recherche en Cancérologie de Montpellier & Université de Montpellier 1, France.
| | - Philippe Pourquier
- Service d'Oncologie Médicale, Centre Hospitalier Universitaire de Nîmes, France; INSERM U896, Institut de Recherche en Cancérologie de Montpellier & Université de Montpellier 1, France
| |
Collapse
|
40
|
Oliveira PA, Arantes-Rodrigues R, Vasconcelos-Nóbrega C. Animal models of urinary bladder cancer and their application to novel drug discovery. Expert Opin Drug Discov 2014; 9:485-503. [DOI: 10.1517/17460441.2014.902930] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
41
|
Grivas PD, Day KC, Karatsinides A, Paul A, Shakir N, Owainati I, Liebert M, Kunju LP, Thomas D, Hussain M, Day ML. Evaluation of the antitumor activity of dacomitinib in models of human bladder cancer. Mol Med 2013; 19:367-76. [PMID: 24166682 DOI: 10.2119/molmed.2013.00108] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 10/23/2013] [Indexed: 12/18/2022] Open
Abstract
Members of the human epidermal growth factor receptor (HER) family play a significant role in bladder cancer progression and may underlie the development of chemotherapy resistance. Dacomitinib is an irreversible tyrosine kinase inhibitor with structural specificity for the catalytic domains of epidermal growth factor receptor (EGFR), HER2 and HER4 that has exhibited vigorous efficacy against other solid tumors. We evaluated the antitumor activity of dacomitinib in human bladder cancer cell lines expressing varying levels of HER family receptors. These cell lines also were established as bladder cancer xenografts in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice to assess dacomitinib activity in vivo. Significant cytotoxic and cytostatic effects were noted in cells expressing elevated levels of the dacomitinib target receptors with apoptosis and cell cycle arrest being the predominant mechanisms of antitumor activity. Cells expressing lower levels of HER receptors were much less sensitive to dacomitinib. Interestingly, dacomitinib was more active than either trastuzumab or cetuximab in vitro, and exhibited increased growth inhibition of bladder tumor xenografts compared with lapatinib. Pharmacodynamic effects of dacomitinib included decreased E-cadherin (E-cad) expression, reduction of EGFR and extracellular signal-regulated kinase (ERK) phosphorylation and reduced mitotic count. Dacomitinib also inhibited tumor growth in a chemotherapy-resistant xenograft and, when combined with chemotherapy in a sensitive xenograft, exhibited superior antitumor effects compared with individual treatments. Evaluation in xenograft-bearing mice revealed that this combination was broadly feasible and well tolerated. In conclusion, dacomitinib exhibited pronounced activity both as a single agent and when combined with chemotherapy in human bladder cancer models. Further investigation of dacomitinib in the preclinical and clinical trial settings is being pursued.
Collapse
Affiliation(s)
- Petros D Grivas
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kathleen C Day
- Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andreas Karatsinides
- Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Alyssa Paul
- Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Nazia Shakir
- Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Iya Owainati
- Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Monica Liebert
- Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lakshmi P Kunju
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Dafydd Thomas
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Maha Hussain
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mark L Day
- Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
42
|
Temsirolimus improves cytotoxic efficacy of cisplatin and gemcitabine against urinary bladder cancer cell lines. Urol Oncol 2013; 32:41.e11-22. [PMID: 24035472 DOI: 10.1016/j.urolonc.2013.04.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 04/25/2013] [Accepted: 04/26/2013] [Indexed: 12/26/2022]
Abstract
OBJECTIVES To analyze the cytotoxic action of temsirolimus using 3 established human bladder cancer cell lines and to assess whether temsirolimus potentiates the anticancer activity of gemcitabine and cisplatin. METHODS Temsirolimus (500, 1,000, 2,000, and 4,000 nM), in isolation, and combined with gemcitabine (100 nM) and cisplatin (2.5 µg/ml), was given to 5637, T24, and HT1376 bladder cancer cell lines. Cell proliferation, autophagy, early apoptosis, and cell cycle distribution were analyzed after a 72-hour period. The expression of mammalian target of rapamycin baseline, Akt, and their phosphorylated forms, before and after treatment with temsirolimus, was evaluated by immunoblotting. RESULTS Temsirolimus slightly decreased the bladder cancer cell proliferation in all 3 cell lines. No significant differences in the expression of mammalian target of rapamycin, Akt, and their phosphorylated forms because of temsirolimus exposure were found in the 3 cell lines. As part of a combined regime along with gemcitabine, and especially with cisplatin, there was a more pronounced antiproliferative effect. This pattern of response was similar to the other parameters analyzed (increased autophagy and apoptosis). Also, in the combined regime, an enhanced cell cycle arrest in the G0/G1 phase was observed. The non-muscle invasive 5637 bladder cancer cell line was most sensitive to both combinations. CONCLUSIONS Temsirolimus makes a moderate contribution in terms of cell proliferation, apoptosis, and autophagy. However, it does potentiate the activity of gemcitabine and particularly cisplatin. Therefore, cisplatin- or gemcitabine-based chemotherapy regimen used in combination with temsirolimus to treat bladder cancer represents a novel and valuable treatment option, which should be tested for future studies in urinary bladder xenograft models.
Collapse
|
43
|
Milowsky MI, Iyer G, Regazzi AM, Al-Ahmadie H, Gerst SR, Ostrovnaya I, Gellert LL, Kaplan R, Garcia-Grossman IR, Pendse D, Balar AV, Flaherty AM, Trout A, Solit DB, Bajorin DF. Phase II study of everolimus in metastatic urothelial cancer. BJU Int 2013; 112:462-70. [PMID: 23551593 DOI: 10.1111/j.1464-410x.2012.11720.x] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
UNLABELLED What's known on the subject? and what does the study add?: No recent advances have been made in the treatment of patients with advanced bladder cancer and, to date, targeted therapies have not resulted in an improvement in outcome. The mammalian target of rapamycin pathway has been shown to be up-regulated in bladder cancer and represents a rational target for therapeutic intervention. In the present phase II study of everolimus, one near-complete response, one partial response and several minor responses suggest that everolimus possesses biological activity in a subset of patients with bladder cancer. To maximize benefit from targeted agents such as everolimus, the preselection of patients based on molecular phenotype is required. OBJECTIVE To assess the efficacy and tolerability of everolimus in advanced urothelial carcimoma (UC). PATIENTS AND METHODS The present study comprised a single-arm, non-randomized study in which all patients received everolimus 10 mg orally once daily continuously (one cycle = 4 weeks). In total, 45 patients with metastatic UC progressing after one to four cytotoxic agents were enrolled between February 2009 and November 2010 at the Memorial Sloan-Kettering Cancer Center. The primary endpoints were 2-month progression-free survival (PFS) and the safety of everolimus, with the secondary endpoint being the response rate. A Simon minimax two-stage design tested the null hypothesis that the true two month PFS rate was ≤ 50%, as opposed to the alternative hypothesis of ≥ 70%. RESULTS The most common grade 3/4 toxicities were fatigue, infection, anaemia, lymphopaenia, hyperglycaemia and hypophosphataemia. There were two partial responses in nodal metastases, with one patient achieving a 94% decrease in target lesions and remaining on drug at 26 months. An additional 12 patients exhibited minor tumour regression. There were 23 of 45 (51%) patients who were progression-free at 2 months with a median (95% CI) PFS of 2.6 (1.8-3.5) months and a median (95% CI) overall survival of 8.3 (5.5-12.1) months. No clear association was observed between mammalian target of rapamycin pathway marker expression and 2-month PFS. CONCLUSIONS Although everolimus did not meet its primary endpoint, one partial response, one near-complete response and twelve minor regressions were observed. Everolimus possesses meaningful anti-tumour activity in a subset of patients with advanced UC. Studies aiming to define the genetic basis of everolimus activity in individual responders are ongoing.
Collapse
Affiliation(s)
- Matthew I Milowsky
- Genitourinary Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vogelzang NJ. Antiangiogenic Agents, Chemotherapy, and the Treatment of Metastatic Transitional Cell Carcinoma. J Clin Oncol 2013; 31:670-5. [DOI: 10.1200/jco.2012.44.4349] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A 69-year-old man with a 100 pack-year history of smoking developed gross hematuria. His medical history included hypertension, a silent myocardial infarction, and a cerebrovascular accident complicated by seizures. Cystoscopy and biopsy showed a 4-cm mass at the right ureteral orifice positive for a high-grade papillary transitional cell carcinoma (TCC) with muscularis propria invasion (Fig 1). The computed tomography (CT)/positron emission tomography (PET) scan of the chest, abdomen, and pelvis showed hydronephrosis and hydroureter with marked cortical thinning and multiple bilateral PET-avid pulmonary nodules, with the largest in the left upper lung measuring 3.0 × 2.5 cm (Figs 2A, 3A), biopsy of which showed invasive high-grade urothelial carcinoma. The patient consented to join a clinical trial for metastatic TCC (USON [US Oncology Network study] 06040) involving treatment with gemcitabine, cisplatin, and sunitinib (GCS) 37.5 mg per day.1Four days later, he experienced a 10-day hospitalization for acute pancreatitis and neutropenia. Sunitinib was discontinued, and he completed four additional cycles of GC. CT/PET showed that the right ureteral mass and all lung nodules had regressed or disappeared (Figs 2B, 3B). The largest remaining lung nodule at 1.4 cm showed no metabolic activity. He underwent a radical cystoprostatectomy and right nephroureterectomy, disclosing residual high-grade urothelial carcinoma infiltrating the full thickness of the ureteral wall. There was carcinoma in situ of the bladder, and 42 nodes were negative for cancer. The surgery was followed by a small, uncomplicated myocardial infarction. A scheduled left thoracotomy to remove the remaining nodule was cancelled. No additional chemotherapy was administered, and the patient remains free of recurrence 2 years from initiation of chemotherapy. The 1.4-cm nodule has calcified and remains stable and metabolically inactive. He has no sequelae of chemotherapy or surgery, with a creatinine level of 1.35 mg/dL.
Collapse
Affiliation(s)
- Nicholas J. Vogelzang
- University of Nevada School of Medicine and US Oncology Research/Comprehensive Cancer Centers of Nevada, Las Vegas, NV
| |
Collapse
|
45
|
Pinto-Leite R, Arantes-Rodrigues R, Palmeira C, Colaço B, Lopes C, Colaço A, Costa C, da Silva VM, Oliveira P, Santos L. Everolimus combined with cisplatin has a potential role in treatment of urothelial bladder cancer. Biomed Pharmacother 2012; 67:116-21. [PMID: 23433853 DOI: 10.1016/j.biopha.2012.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 11/23/2012] [Indexed: 12/26/2022] Open
Abstract
Cisplatin (CDDP)-based chemotherapy is a commonly treatment for advanced urothelial carcinoma. However, episodes of cisplatin resistance have been referenced. Recently it has been reported that everolimus (RAD001) could have an important role to play in bladder-cancer treatment and that mTOR inhibitors may restore chemosensitivity in resistant tumours. The aim of this study was to assess RAD001 in vitro ability to enhance CDDP cytotoxicity in three human bladder-cancer cell lines. Over the course of 72h, the cells were exposed to different concentrations of CDDP and RAD001, isolated or combined. Treatment with CDDP statistically (P<0.05) decreased cell proliferation in cell lines in a dose-dependent manner. The anti-proliferative activity of CDDP used in combination with RAD001 was statistically significant (P<0.05) in the cell lines at all concentrations tested. RAD001 had a therapeutic effect when used in combination with CDDP and could therefore be a useful anti-cancer drug combination for patients with bladder cancer.
Collapse
Affiliation(s)
- Rosário Pinto-Leite
- Genetic Service, Cytogenetic Laboratory, Hospital Center of Trás-os-Montes and Alto Douro, 5000-508, Vila Real, Portugal.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dickstein RJ, Nitti G, Dinney CP, Davies BR, Kamat AM, McConkey DJ. Autophagy limits the cytotoxic effects of the AKT inhibitor AZ7328 in human bladder cancer cells. Cancer Biol Ther 2012; 13:1325-38. [PMID: 22895070 DOI: 10.4161/cbt.21793] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mutations that activate the PI3K/AKT/mTOR pathway are relatively common in urothelial (bladder) cancers, but how these pathway mutations affect AKT dependency is not known. We characterized the relationship between AKT pathway mutational status and sensitivity to the effects of the selective AKT kinase inhibitor AZ7328 using a panel of 12 well-characterized human bladder cancer cell lines. METHODS Sequenome DNA sequencing was performed to identify mutations in a panel of 12 urothelial cancer cell lines. Drug-induced proliferative inhibition and apoptosis were quantified using MTT assays and propidium iodide staining with FACS analyses. Protein activation via phosphorylation was measured by immunoblotting. Autophagy was measured by LC3 immunofluorescence and immunoblotting. RESULTS AZ7328 inhibited proliferation and AKT substrate phosphorylation in a concentration-dependent manner but had minimal effects on apoptosis. Proliferative inhibition correlated loosely with the presence of activating PIK3CA mutations and was strengthened in combination with the mTOR inhibitor rapamycin. AZ7328 induced autophagy in some of the lines, and in the cells exposed to a combination of AZ7328 and chemical autophagy inhibitors apoptosis was induced. CONCLUSIONS The cytostatic effects of AZ7328 correlate with PIK3CA mutations and are greatly enhanced by dual pathway inhibition using an mTOR inhibitor. Furthermore, AZ7328 can interact with autophagy inhibitors to induce apoptosis in some cell lines. Overall, our results support the further evaluation of combinations of PI3K/AKT/mTOR pathway and autophagy inhibitors in pre-clinical in vivo models and ultimately in patients with PIK3CA mutant bladder cancers.
Collapse
Affiliation(s)
- Rian J Dickstein
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
47
|
Altmeyer A, Josset E, Denis JM, Gueulette J, Slabbert J, Mutter D, Noël G, Bischoff P. The mTOR inhibitor RAD001 augments radiation-induced growth inhibition in a hepatocellular carcinoma cell line by increasing autophagy. Int J Oncol 2012; 41:1381-6. [PMID: 22895785 DOI: 10.3892/ijo.2012.1583] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 05/09/2012] [Indexed: 11/06/2022] Open
Abstract
Treatment of hepatocellular carcinoma (HCC) is a major concern for physicians as its response to chemotherapy and radiotherapy remains generally poor, due, in part, to intrinsic resistance to either form of treatment. We previously reported that an irradiation with fast neutrons, which are high-linear energy transfer (LET) particles, massively induced autophagic cell death in the human HCC SK-Hep1 cell line. In the present study, we tested the capacity of the mammalian target of rapamycin (mTOR) inhibitor RAD001 to augment the cytotoxicity of low and high-LET radiation in these cells. As mTOR is a key component in a series of pathways involved in tumor growth and development, it represents a potential molecular target for cancer treatment. Results indicate that RAD001, at clinically relevant nanomolar concentrations, enhances the efficacy of both high- and low-LET radiation in SK-Hep1 cells, and that the induction of autophagy may account for this effect. However, fast neutrons were found to be more efficient at reducing tumor cell growth than low-LET radiation.
Collapse
Affiliation(s)
- Anaïs Altmeyer
- EA 3430, University of Strasbourg, Centre Paul Strauss, Strasbourg, France
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Wei H, Kamat A, Chen M, Ke HL, Chang DW, Yin J, Grossman HB, Dinney CP, Wu X. Association of polymorphisms in oxidative stress genes with clinical outcomes for bladder cancer treated with Bacillus Calmette-Guérin. PLoS One 2012; 7:e38533. [PMID: 22701660 PMCID: PMC3373532 DOI: 10.1371/journal.pone.0038533] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 05/06/2012] [Indexed: 12/11/2022] Open
Abstract
Genetic polymorphisms in oxidative stress pathway genes may contribute to carcinogenesis, disease recurrence, treatment response, and clinical outcomes. We applied a pathway-based approach to determine the effects of multiple single nucleotide polymorphisms (SNPs) within this pathway on clinical outcomes in non-muscle-invasive bladder cancer (NMIBC) patients treated with Bacillus Calmette-Guérin (BCG). We genotyped 276 SNPs in 38 genes and evaluated their associations with clinical outcomes in 421 NMIBC patients. Twenty-eight SNPs were associated with recurrence in the BCG-treated group (P<0.05). Six SNPs, including five in NEIL2 gene from the overall and BCG group remained significantly associated with recurrence after multiple comparison adjustments (q<0.1). Cumulative unfavorable genotype analysis showed that the risk of recurrence increased with increasing number of unfavorable genotypes. In the analysis of risk factors associated with progression to disease, rs3890995 in UNG, remained significant after adjustment for multiple comparison (q<0.1). These results support the hypothesis that genetic variations in host oxidative stress genes in NMIBC patients may affect response to therapy with BCG.
Collapse
Affiliation(s)
- Hua Wei
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ashish Kamat
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Meng Chen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hung-Lung Ke
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - David W. Chang
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jikai Yin
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - H. Barton Grossman
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Colin P. Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
49
|
Jana BRP, Galsky MD, Hahn NM, Milowsky MI, Sonpavde G. Novel molecular targets for the therapy of urothelial carcinoma. Expert Opin Ther Targets 2012; 16:499-513. [PMID: 22510032 DOI: 10.1517/14728222.2012.677441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION First-line platinum-based combinations are active in locally advanced and metastatic urothelial carcinoma; however, long-term outcomes including disease-specific and overall survival remain suboptimal. In addition, approximately 40 - 50% of patients with advanced urothelial carcinoma have coexisting medical issues that preclude the use of cisplatin-based therapy. Improvements in our understanding of the molecular mechanisms of urothelial tumorigenesis have led to first-generation clinical trials evaluating novel agents targeting molecular pathways. These are particularly relevant in regard to subpopulations. Novel trial designs warrant consideration to accelerate accrual. AREAS COVERED In this review, novel molecular targets for the therapy of urothelial carcinoma, as well as recently completed and ongoing clinical trials utilizing novel targeted agents, are discussed. A Medline search with key words, abstracts reported at national conferences on urothelial carcinoma and NCI clinical trial identifiers was used for this review. EXPERT OPINION Improved understanding of molecular biology has identified a number of new molecular targets, but there is a seeming absence of one dominant molecular driver for urothelial cancer. An adaptive and biomarker-derived strategy may be warranted. Clinical trials utilizing targeted agents are ongoing and results are awaited.
Collapse
Affiliation(s)
- Bagi R P Jana
- University of Texas Medical Branch , Galveston, TX , USA
| | | | | | | | | |
Collapse
|
50
|
Seront E, Rottey S, Sautois B, Kerger J, D'Hondt LA, Verschaeve V, Canon JL, Dopchie C, Vandenbulcke JM, Whenham N, Goeminne JC, Clausse M, Verhoeven D, Glorieux P, Branders S, Dupont P, Schoonjans J, Feron O, Machiels JP. Phase II study of everolimus in patients with locally advanced or metastatic transitional cell carcinoma of the urothelial tract: clinical activity, molecular response, and biomarkers. Ann Oncol 2012; 23:2663-2670. [PMID: 22473592 DOI: 10.1093/annonc/mds057] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND This phase II study assessed the safety and efficacy of everolimus, an oral mammalian target of rapamycin inhibitor in advanced transitional carcinoma cell (TCC) after failure of platinum-based therapy. PATIENTS AND METHODS Thirty-seven patients with advanced TCC received everolimus 10 mg/day until progressive disease (PD) or unacceptable toxicity. The primary end point was the disease control rate (DCR), defined as either stable disease (SD), partial response (PR), or complete response at 8 weeks. Angiogenesis-related proteins were detected in plasma and changes during everolimus treatment were analyzed. PTEN expression and PIK3CA mutations were correlated to disease control. RESULTS Two confirmed PR and eight SD were observed, resulting in a DCR of 27% at 8 weeks. Everolimus was well tolerated. Compared with patients with noncontrolled disease, we observed in patients with controlled disease a significant higher baseline level of angiopoietin-1 and a significant early plasma decrease in angiopoietin-1, endoglin, and platelet-derived growth factor-AB. PTEN loss was observed only in patients with PD. CONCLUSIONS Everolimus showed clinical activity in advanced TCC. The profile of the plasma angiogenesis-related proteins suggested a role of the everolimus antiangiogenic properties in disease control. PTEN loss might be associated with everolimus resistance.
Collapse
Affiliation(s)
- E Seront
- Department of Medical Oncology, Centre du Cancer, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels; Angiogenesis and Cancer Research Laboratory, Pole of Pharmacology and Therapeutics, Université catholique de Louvain, Brussels
| | - S Rottey
- Department of Medical Oncology, University Hospital Gent, Gent
| | - B Sautois
- Department of Medical Oncology, Centre Hospitalier Universitaire Sart-Tilman, Liège
| | - J Kerger
- Department of Medical Oncology, Centre Hospitalier Universitaire Mont-Godinne, Namur
| | - L A D'Hondt
- Department of Medical Oncology, Centre Hospitalier Universitaire Mont-Godinne, Namur
| | - V Verschaeve
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi
| | - J-L Canon
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi
| | - C Dopchie
- Department of Medical Oncology, Réseau Hospitalier de Médecine Sociale, Tournai
| | - J M Vandenbulcke
- Department of Medical Oncology, Réseau Hospitalier de Médecine Sociale, Tournai
| | - N Whenham
- Department of Medical Oncology, Clinique Saint-Pierre Ottignies, Ottignies
| | - J C Goeminne
- Department of Medical Oncology, Centre de Maternité Saint Elisabeth, Namur
| | - M Clausse
- Department of Medical Oncology, Clinique Saint-Luc, Bouge
| | - D Verhoeven
- Department of Medical Oncology, AZ Klina, Braschaat
| | - P Glorieux
- Department of Medical Oncology, Clinique Saint Joseph, Arlon
| | - S Branders
- Machine Learning Group, Institute of Information and Communication Technologies, Electronics and Applied Mathematics, Université catholique de Louvain, Louvain-la-Neuve
| | - P Dupont
- Machine Learning Group, Institute of Information and Communication Technologies, Electronics and Applied Mathematics, Université catholique de Louvain, Louvain-la-Neuve
| | - J Schoonjans
- Department of radiology, Centre Hospitalier de Jolimont, Haine Saint Paul, Belgium
| | - O Feron
- Angiogenesis and Cancer Research Laboratory, Pole of Pharmacology and Therapeutics, Université catholique de Louvain, Brussels
| | - J-P Machiels
- Department of Medical Oncology, Centre du Cancer, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels.
| |
Collapse
|