51
|
Mocellin S, Goodwin A, Pasquali S. Risk-reducing medications for primary breast cancer: a network meta-analysis. Cochrane Database Syst Rev 2019; 4:CD012191. [PMID: 31032883 PMCID: PMC6487387 DOI: 10.1002/14651858.cd012191.pub2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Breast cancer is the most frequently occurring malignancy and the second cause of death for cancer in women. Cancer prevention agents (CPAs) are a promising approach to reduce the burden of breast cancer. Currently, two main types of CPAs are available: selective estrogen receptor modulators (SERMs, such as tamoxifen and raloxifene) and aromatase inhibitors (AIs, such as exemestane and anastrozole). OBJECTIVES To assess the efficacy and acceptability of single CPAs for the prevention of primary breast cancer, in unaffected women, at an above-average risk of developing breast cancer.Using a network meta-analysis, to rank single CPAs, based on their efficacy and acceptability (an endpoint that is defined as the inverse of CPA-related toxicity). SEARCH METHODS We searched the Cochrane Breast Cancer Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, World Health Organization's International Clinical Trials Registry Platform (WHO ICTRP), and ClinicalTrials.gov on 17 August 2018. We handsearched reference lists to identify additional relevant studies. SELECTION CRITERIA We included randomized controlled trials (RCTs) that enrolled women without a personal history of breast cancer but with an above-average risk of developing a tumor. Women had to be treated with a CPA and followed up to record the occurrence of breast cancer and adverse events. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and conducted risk of bias assessments of the included studies, and assessed the certainty of the evidence using GRADE. Outcome data included incidence of breast carcinoma (both invasive and in situ carcinoma) and adverse events (both overall and severe toxicity). We performed a conventional meta-analysis (for direct comparisons of a single CPA with placebo or a different CPA) and network meta-analysis (for indirect comparisons). MAIN RESULTS We included six studies enrolling 50,927 women randomized to receive one CPA (SERMs: tamoxifen or raloxifene, or AIs: exemestane or anastrozole) or placebo. Three studies compared tamoxifen and placebo, two studies compared AIs (exemestane or anastrozole) versus placebo, and one study compared tamoxifen versus raloxifene. The risk of bias was low for all RCTs.For the tamoxifen versus placebo comparison, tamoxifen likely resulted in a lower risk of developing breast cancer compared to placebo (risk ratio (RR) 0.68, 95% confidence interval (CI) 0.62 to 0.76; 3 studies, 22,832 women; moderate-certainty evidence). In terms of adverse events, tamoxifen likely increased the risk of severe toxicity compared to placebo (RR 1.28, 95% CI 1.12 to 1.47; 2 studies, 20,361 women; moderate-certainty evidence). In particular, women randomized to receive tamoxifen experienced a higher incidence of both endometrial carcinoma (RR 2.26, 95% CI 1.52 to 3.38; high-certainty evidence) and thromboembolism (RR 2.10, 95% CI 1.14 to 3.89; high-certainty evidence) compared to women who received placebo.For the AIs versus placebo comparison, AIs (exemestane or anastrozole) reduced the risk of breast cancer by 53% (RR 0.47, 95% CI 0.35 to 0.63; 2 studies, 8424 women; high-certainty evidence). In terms of adverse events, AIs increased the risk of severe toxicity by 18% (RR 1.18, 95% CI 1.09 to 1.28; 2 studies, 8352 women; high-certainty evidence). These differences were sustained especially by endocrine (e.g. hot flashes), gastrointestinal (e.g. diarrhea), and musculoskeletal (e.g. arthralgia) adverse events, while there were no differences in endometrial cancer or thromboembolism rates between AIs and placebo.For the tamoxifen versus raloxifene comparison, raloxifene probably performed worse than tamoxifen in terms of breast cancer incidence reduction (RR 1.25, 95% CI 1.09 to 1.43; 1 study, 19,490 women; moderate-certainty evidence), but its use was associated with lower toxicity rates (RR 0.87, 95% CI 0.80 to 0.95; 1 study, 19,490 women; moderate-certainty evidence), particularly relating to incidence of endometrial cancer and thromboembolism.An indirect comparison of treatment effects allowed us to compare the SERMs and AIs in this review. In terms of efficacy, AIs (exemestane or anastrozole) may have reduced breast cancer incidence slightly compared to tamoxifen (RR 0.67, 95% CI 0.46 to 0.98; 5 RCTs, 31,256 women); however, the certainty of evidence was low. A lack of model convergence did not allow us to analyze toxicity data. AUTHORS' CONCLUSIONS For women with an above-average risk of developing breast cancer, CPAs can reduce the incidence of this disease. AIs appear to be more effective than SERMs (tamoxifen) in reducing the risk of developing breast cancer. AIs are not associated with an increased risk of endometrial cancer and thromboembolic events. However, long-term data on toxicities from tamoxifen are available while the follow-up toxicity data on unaffected women taking AIs is relatively short. Additional data from direct comparisons are needed to fully address the issues of breast cancer prevention by risk-reducing medications, with special regards to acceptability (i.e. the benefit/harm ratio).
Collapse
Affiliation(s)
| | | | - Sandro Pasquali
- Fondazione IRCCS Istituto Nazionale dei TumoriSarcoma ServiceVia G. Venezian 1MilanoItaly20133
| |
Collapse
|
52
|
Salem O, Hansen CG. The Hippo Pathway in Prostate Cancer. Cells 2019; 8:E370. [PMID: 31018586 PMCID: PMC6523349 DOI: 10.3390/cells8040370] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 02/07/2023] Open
Abstract
Despite recent efforts, prostate cancer (PCa) remains one of the most common cancers in men. Currently, there is no effective treatment for castration-resistant prostate cancer (CRPC). There is, therefore, an urgent need to identify new therapeutic targets. The Hippo pathway and its downstream effectors-the transcriptional co-activators, Yes-associated protein (YAP) and its paralog, transcriptional co-activator with PDZ-binding motif (TAZ)-are foremost regulators of stem cells and cancer biology. Defective Hippo pathway signaling and YAP/TAZ hyperactivation are common across various cancers. Here, we draw on insights learned from other types of cancers and review the latest advances linking the Hippo pathway and YAP/TAZ to PCa onset and progression. We examine the regulatory interaction between Hippo-YAP/TAZ and the androgen receptor (AR), as main regulators of PCa development, and how uncontrolled expression of YAP/TAZ drives castration resistance by inducing cellular stemness. Finally, we survey the potential therapeutic targeting of the Hippo pathway and YAP/TAZ to overcome PCa.
Collapse
Affiliation(s)
- Omar Salem
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| | - Carsten G Hansen
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
53
|
The effect of metformin on biomarkers associated with breast cancer outcomes: a systematic review, meta-analysis, and dose-response of randomized clinical trials. Clin Transl Oncol 2019; 22:37-49. [PMID: 31006835 DOI: 10.1007/s12094-019-02108-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/01/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Breast cancer is a leading cause of cancer mortality in developed countries. We performed a meta-analysis of randomized clinical trials to investigate the effect of metformin on biomarkers associated with breast cancer outcomes and to explore the dose-response relationship. METHODS A systematic search was performed from onset of the database to January 2019 in MEDLINE/PubMed, SCOPUS, and Cochrane library to identify randomized clinical trials investigating the impact of metformin on insulin, glucose, CRP, leptin, body mass indices (BMI), cholesterol, Ki-67, and Homeostatic Model Assessment for Insulin-Resistance (HOMA-IR). Effect sizes were expressed as weighted mean difference (WMD) and 95% confidence intervals (CI) using a random-effects models. RESULTS Nine studies providing 1,363 participants were included in the meta-analysis. Pooled results showed a significant reduction in insulin (WMD: - 0.99 U/ml, 95% CI - 1.66, - 0.33), glucose (WMD: - 1.78 ml/dl, 95% CI - 2.96, - 0.60), CRP (WMD: - 0.60 mg/l, 95% CI - 0.88, - 0.33), HOMA-IR (WMD: - 0.45, 95% CI - 0.77, - 0.11), leptin (WMD: - 2.44 ng/ml, 95% CI - 3.28, - 1.61), BMI (WMD: - 0.55 kg/m2, 95% CI - 1.00, - 0.11), and Ki-67 (WMD: - 4.06, 95% CI - 7.59, - 0.54). Results of the subgroup analyses showed that insulin, glucose, and BMI decreased more significantly when the duration of administering metformin intervention was above 4 weeks. We did not observe non-linear changes in the dose-response relationship between metformin and biomarkers as outcomes. CONCLUSIONS Breast cancer patients receiving metformin as treatment for diabetes showed significant reduction in levels of insulin, fasting glucose, CRP, HOMA, leptin, BMI, and Ki-67.
Collapse
|
54
|
Hamada T, Nowak JA, Milner DA, Song M, Ogino S. Integration of microbiology, molecular pathology, and epidemiology: a new paradigm to explore the pathogenesis of microbiome-driven neoplasms. J Pathol 2019; 247:615-628. [PMID: 30632609 PMCID: PMC6509405 DOI: 10.1002/path.5236] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/24/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023]
Abstract
Molecular pathological epidemiology (MPE) is an integrative transdisciplinary field that addresses heterogeneous effects of exogenous and endogenous factors (collectively termed 'exposures'), including microorganisms, on disease occurrence and consequences, utilising molecular pathological signatures of the disease. In parallel with the paradigm of precision medicine, findings from MPE research can provide aetiological insights into tailored strategies of disease prevention and treatment. Due to the availability of molecular pathological tests on tumours, the MPE approach has been utilised predominantly in research on cancers including breast, lung, prostate, and colorectal carcinomas. Mounting evidence indicates that the microbiome (inclusive of viruses, bacteria, fungi, and parasites) plays an important role in a variety of human diseases including neoplasms. An alteration of the microbiome may be not only a cause of neoplasia but also an informative biomarker that indicates or mediates the association of an epidemiological exposure with health conditions and outcomes. To adequately educate and train investigators in this emerging area, we herein propose the integration of microbiology into the MPE model (termed 'microbiology-MPE'), which could improve our understanding of the complex interactions of environment, tumour cells, the immune system, and microbes in the tumour microenvironment during the carcinogenic process. Using this approach, we can examine how lifestyle factors, dietary patterns, medications, environmental exposures, and germline genetics influence cancer development and progression through impacting the microbial communities in the human body. Further integration of other disciplines (e.g. pharmacology, immunology, nutrition) into microbiology-MPE would expand this developing research frontier. With the advent of high-throughput next-generation sequencing technologies, researchers now have increasing access to large-scale metagenomics as well as other omics data (e.g. genomics, epigenomics, proteomics, and metabolomics) in population-based research. The integrative field of microbiology-MPE will open new opportunities for personalised medicine and public health. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jonathan A Nowak
- Department of Pathology Program in MPE Molecular Pathological Epidemiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Danny A Milner
- American Society for Clinical Pathology, Chicago, Illinois, USA
| | - Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology Program in MPE Molecular Pathological Epidemiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
55
|
Abstract
OBJECTIVE The aim of this study was to explore the relationship between statin use and the risk of pancreatic cancer. METHODS Electronic databases were searched to identify relevant studies published until January 2018. The pooled relative risks (RRs) and 95% confidence intervals (CIs) were calculated with random-effects model. Subgroup analyses and sensitivity analysis were also conducted. Cochran Q test and I(2) statistic were used to evaluate the heterogeneity. RESULTS Twenty-six studies were included that contained more than 3 million participants and 170,000 pancreatic cancer patients. The overall result demonstrated a significant decrease in pancreatic cancer risk with statin use (RR, 0.84; 95% CI, 0.73-0.97; P = 0.000; I(2) = 84.4%). In subgroup analyses, nonsignificant association was detected between long-term statin use and the risk of pancreatic cancer (RR, 0.98; 95% CI, 0.86-1.11; P = 0.718; I(2) = 0.0%). Meanwhile, there was nonsignificant association between the use of lipophilic statins and the risk of pancreatic cancer (RR, 0.98; 95% CI, 0.84-1.15; P = 0.853; I(2) = 27.2%). No publication bias was found in this meta-analysis. CONCLUSIONS The overall result of this meta-analysis supports the hypothesis that statins have a protective effect on pancreatic cancer. Furthermore, high-quality randomized clinical trials and cohort studies are needed to confirm these findings.
Collapse
|
56
|
Eriksson L, He W, Eriksson M, Humphreys K, Bergh J, Hall P, Czene K. Adjuvant Therapy and Mammographic Density Changes in Women With Breast Cancer. JNCI Cancer Spectr 2019; 2:pky071. [PMID: 31360886 PMCID: PMC6649795 DOI: 10.1093/jncics/pky071] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/23/2018] [Accepted: 11/15/2018] [Indexed: 12/11/2022] Open
Abstract
Background Tamoxifen decreases mammographic density. Whether compliance affects this relationship is unclear as is the relationship between other types of adjuvant treatment and changes in mammographic density. Methods This prospective cohort study included 2490 women diagnosed with breast cancer during 2001-2015 in Sweden. Mammographic density was assessed within 3 months of diagnosis and 6-36 months post diagnosis. Logistic regression was performed to study the association between each respective adjuvant treatment and mammographic density reduction (annual dense area decrease >15%). Results Intention-to-treat analyses using treatment information from the regional cancer registries showed that tamoxifen-treated patients more frequently experienced mammographic density reductions compared with nontreated patients (odds ratio [OR] = 1.58, 95% confidence interval [CI] = 1.25 to 1.99), as did chemotherapy-treated patients (OR = 1.28, 95% CI = 1.06 to 1.54). For chemotherapy, the association was mainly seen in premenopausal women. Neither aromatase inhibitors nor radiotherapy was associated with density change. Tamoxifen use based on prescription and dispensation data from the Swedish Prescribed Drug Register showed that users were more likely to have density reductions compared with nonusers (adjusted OR = 2.24, 95% CI = 1.40 to 3.59). Moreover, among tamoxifen users, tamoxifen continuers were more likely than discontinuers to experience density reductions (adjusted OR = 1.50, 95% CI = 1.04 to 2.17). Conclusions Our results indicate that adherence influences the association between tamoxifen and mammographic density reduction. We further found that chemotherapy was associated with density reductions and propose that this is largely secondary to chemotherapy-induced ovarian failure.
Collapse
Affiliation(s)
| | - Wei He
- Correspondence to: Wei He, PhD, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12 A, Stockholm 171 77, Sweden (e-mail: )
| | | | | | | | | | | |
Collapse
|
57
|
The anticancer potential of metformin on prostate cancer. Prostate Cancer Prostatic Dis 2019; 22:351-361. [PMID: 30651580 DOI: 10.1038/s41391-018-0085-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 06/29/2018] [Accepted: 07/05/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is characterized as the most frequent type of cancer in males. Recent research has suggested patients who have diabetes mellitus taking metformin (MF) have a lower risk of PCa. MF has antineoplastic effects such as adenosine monophosphate-activated protein kinase (AMPK)-dependent and independent mechanisms, suppression of androgen signaling pathway, and alterations of insulin-like growth factor-1 (IGF-1) signaling pathways that cause the growth and proliferation of PCa. Based on epidemiological factors, patients with diabetes mellitus may have a protective effect on PCa. METHODS A literature search on MEDLINE® was conducted using a combined query of "prostate cancer" and "metformin" to yield publications unveiling the mechanisms of action, biological effects, epidemiological evidence, and research advances of MF with respect to PCa. RESULTS Evidence has shown that MF has multiple antineoplastic effects through AMPK-dependent and independent mechanisms, the alteration of IGF-1 signaling pathways, suppression of the androgen receptor pathway, inhibition of the mTOR pathway, and lipogenesis. Conduction of meta-analysis suggests mortality benefit to patients who exhibit PCa when taking MF. Clinical trials have shown evidence, demonstrating MF to improving significantly. CONCLUSIONS Herewith we review the literature regarding the numerous mechanisms of action of MF on PCa in order to decrease or repress the growth, proliferation, and differentiation of PCa cells. We analyze the molecular impacts of MF as well as adjunct therapies such as androgen deprivation therapy, aspirin, statin, or chemotherapy, proposing that MF may have a future role in the treatment protocol of PCa whether as a monotherapy or in combination with other drugs.
Collapse
|
58
|
Puzianowska-Kuznicka M, Kuryłowicz A, Walkiewicz D, Borkowska J, Owczarz M, Olszanecka-Glinianowicz M, Wieczorowska-Tobis K, Skalska A, Szybalska A, Mossakowska M. Obesity Paradox in Caucasian Seniors: Results of the PolSenior Study. J Nutr Health Aging 2019; 23:796-804. [PMID: 31641728 PMCID: PMC6800404 DOI: 10.1007/s12603-019-1257-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/19/2019] [Indexed: 01/29/2023]
Abstract
OBJECTIVES To investigate the influence of overweight and obesity on general performance and mortality in seniors. DESIGN Cross-sectional multidisciplinary study on ageing of the Polish population. SETTING Community-dwelling individuals aged 65 years or older, selected using three-stage stratified, proportional draw. PARTICIPANTS 4944 Polish Caucasian seniors, aged 65 years or older recruited between October 2007 and October 2010. MEASUREMENTS All study subjects underwent measurement of body mass index (BMI), waist circumference (WC), and arm circumference (AC). The physical and cognitive performance was evaluated using the Katz Activities of Daily Living (ADL) score and Mini-Mental State Examination (MMSE), respectively. Morbidity data were obtained from a medical questionnaire. Mortality data were obtained from the Population Register of Poland between October 2015 and October 2018. RESULTS Increasing age was associated with a decreased prevalence of obesity (all p<0.001). Higher BMI, WC and AC values were associated with higher ADL and MMSE scores (all p<0.001). On multivariate analysis, all three body measurements in women remained independent predictors of the ADL score (BMI p=0.002, WC p=0.005, AC p<0.001) and MMSE score (p<0.001, p=0.003, p<0.001). In men, physical functioning was associated with AC (p=0.003), and cognitive status was associated with AC (p<0.001) and BMI (p=0.013). There was no association between general obesity, abdominal obesity, or AC with several aging-related adverse conditions. Kaplan-Meier survival curves showed that overweight and obesity were associated with the lowest mortality. On multivariate analysis, BMI and AC values remained independent predictors of mortality. In successfully aging individuals, neither BMI, WC, nor AC remained such predictors. CONCLUSIONS Overweight and obesity in Caucasian seniors are not associated with deterioration of physical and cognitive function or with increased mortality.
Collapse
Affiliation(s)
- M Puzianowska-Kuznicka
- Monika Puzianowska-Kuznicka, MD, PhD, Department of Human Epigenetics, Mossakowski Medical Research Centre, PAS, Pawinskiego 5, 02-106 Warsaw, Poland; phone/fax: +48 22 6086591;
| | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Cyclopeptide RA-V Inhibits Organ Enlargement and Tumorigenesis Induced by YAP Activation. Cancers (Basel) 2018; 10:cancers10110449. [PMID: 30453531 PMCID: PMC6265802 DOI: 10.3390/cancers10110449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022] Open
Abstract
The Hippo pathway restricts organ size during development and its inactivation plays a crucial role in cancer. Yes-associated protein (YAP) and its paralog transcriptional coactivator with PSD-95/Dlg/ZO-1 (PDZ)-binding motif (TAZ) are transcription co-activators and effectors of the Hippo pathway mediating aberrant enlargement of organs and tumor growth upon Hippo pathway inactivation. It has been demonstrated that genetic inactivation of YAP could be an effective approach to inhibit tumorigenesis. In order to identify pharmacological inhibitors of YAP, we screened a library of 52,683 compounds using a YAP-specific reporter assay. In this screen we identified cyclopeptide RA-V (deoxybouvardin) as a specific inhibitor of YAP and TAZ but not other reporters. Unexpectedly, later experiments demonstrated that RA-V represses the protein but not mRNA levels of YAP target genes. Nevertheless, RA-V strongly blocks liver enlargement induced by Mst1/2 knockout. Furthermore, RA-V not only inhibits liver tumorigenesis induced by YAP activation, but also induces regression of established tumors. We found that RA-V inhibits dedifferentiation and proliferation, while inducing apoptosis of hepatocytes. Furthermore, RA-V also induces apoptosis and inhibits proliferation of macrophages in the microenvironment, which are essential for YAP-induced tumorigenesis. RA-V is thus a drug candidate for cancers involving YAP/TAZ activation.
Collapse
|
60
|
Wen DT, Xu Z, Xuan ML, Liang GR, Zheng WL, Liang XF, Xiao J, Wang XY. Prognostic Effect of Bisphosphonate Exposure for Patients With Diagnosed Solid Cancer: A Systematic Review With Meta-Analysis of Observational Studies. Front Oncol 2018; 8:495. [PMID: 30420942 PMCID: PMC6215818 DOI: 10.3389/fonc.2018.00495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023] Open
Abstract
Background: Bisphosphonates are widely prescribed for the prevention and treatment of osteoporosis. Recent epidemiological studies indicate that people with bisphosphonate use may have lower cancer risk and have improved survival. The aim of this study is to determine the association between bisphosphonate use and survival outcomes in solid cancer patients using systematic review and meta-analysis. Methods: A systematic literature search was performed using the PubMed, Embase, and Cochrane databases. Original articles published until April, 2018 were selected. The survival outcome measures assessed included overall survival (OS), cancer-specific survival (CSS) and recurrence-free survival (RFS). Pooled hazard ratio (HR) and their 95% confidence interval (95% CI) were derived using a random-effects model. Results: Out of 9,742 retrieved citations, six cohort studies and two nested case-control studies satisfying the inclusion criteria were included for analyses. Bisphosphonate use was significantly associated with improved OS (HR 0.84, 95% CI 0.76–0.93), CSS (HR 0.73, 95% CI 0.58–0.90) and RFS (HR 0.72, 95% CI 0.53–0.96). The results of subgroup analyses stratified by major study characteristics were generally consistent with the main findings. For individual cancer type, we found that bisphosphonate use was significantly associated with longer OS for patients with gastroesophageal cancer (HR 0.62, 95% CI 0.40–0.98), as well as longer CSS for patients with breast cancer (HR 0.73, 95% CI 0.55–0.95). Conclusions: Current evidence indicates that bisphosphonate use is significantly associated with improved survival for patients with solid cancer. However, the prognostic effects in specific solid tumors remains to be confirmed by further large prospective cohort studies.
Collapse
Affiliation(s)
- Dan-Ting Wen
- Department of Gynecology, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Postdoctoral Research Station, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Gynecology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Zheng Xu
- General Office of Multiple Functional Chinese Medications, Bao'an TCM Hospital Group, Shenzhen, China
| | - Mei-Ling Xuan
- Department of Gynecology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Guo-Rong Liang
- Department of Gynecology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Wei-Ling Zheng
- Department of Gynecology, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue-Fang Liang
- Department of Gynecology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Jing Xiao
- Department of Gynecology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xiao-Yun Wang
- Department of Gynecology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
61
|
Giannakeas V, Cadarette SM, Ban JK, Lipscombe L, Narod SA, Kotsopoulos J. Denosumab and breast cancer risk in postmenopausal women: a population-based cohort study. Br J Cancer 2018; 119:1421-1427. [PMID: 30420611 PMCID: PMC6265331 DOI: 10.1038/s41416-018-0225-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/03/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Denosumab inhibits the receptor activator of nuclear factor κB (RANK) pathway and is used to treat osteoporosis. Emerging evidence suggests RANK-blockade may play a role in mammary tumourigenesis. Thus, we undertook a population-based study of denosumab use and breast cancer risk in a large cohort of postmenopausal women. METHODS We included women 67+ years with prior bisphosphonate use who filled a first prescription for denosumab. They were matched on age, date, cumulative prior use of and time since last use of a bisphosphonate to women with no history of denosumab. Cox proportional hazards was used to estimate the hazard ratio (HR) of breast cancer with denosumab use. RESULTS A total of 100,368 women were included in the analysis with 1271 incident breast cancer events. Denosumab use was associated with a 13% decreased breast cancer risk (HR = 0.87; 95% CI 0.76-1.00). There was no relationship between increasing number of denosumab doses and breast cancer risk (P-trend = 0.15). CONCLUSION These findings suggest a potential protective effect of ever denosumab use on breast cancer risk in a cohort of older women previously treated with bisphosphonates.
Collapse
Affiliation(s)
- Vasily Giannakeas
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada
| | - Suzanne M Cadarette
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.,Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Joann K Ban
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Lorraine Lipscombe
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada.,Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada. .,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
62
|
Akinwunmi B, Vitonis AF, Titus L, Terry KL, Cramer DW. Statin therapy and association with ovarian cancer risk in the New England Case Control (NEC) study. Int J Cancer 2018; 144:991-1000. [PMID: 30006925 DOI: 10.1002/ijc.31758] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/25/2018] [Accepted: 07/04/2018] [Indexed: 12/17/2022]
Abstract
Statins are widely used to lower blood cholesterol and reduce risk for cardiovascular diseases, but attention has recently focused on a role in cancer prevention or therapy. Here we present data from a large case-control study addressing whether statin use can lower the risk for epithelial ovarian cancer (EOC). Between 1992 and 2008, data including medications used for at least 6 months were collected from 2,040 cases with EOC and 2,100 frequency-matched controls without the disease who participated in the New England Case Control study. We used unconditional logistic regression controlling for matching factors and potential confounders to examine the association between statin use and the risk for EOC. Overall, women who used statins had 32% lower risk of ovarian cancer compared to non-users (Odds ratio (OR) 0.68, 95% Confidence Interval (CI): 0.54-0.85), adjusting for the matching factors and other covariates. The reduced risk was most apparent in women taking a lipophilic statin who began use after age 49, and who had used them 2-4.9 years. Statin use was associated with lower risks for both serous and non-serous histologic subtypes with the strongest effect seen for mucinous and mixed epithelial subtypes. The association became apparent about a decade after the introduction of statins and did not appear to be confounded by indications for use of statins or medications used concomitantly. In this case-control study, statins were found to lower the risk for both serous and non-serous EOC and especially mucinous EOC.
Collapse
Affiliation(s)
- Babatunde Akinwunmi
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital, Boston, MA.,Department of Global Education, Harvard Medical School, Boston, MA
| | - Allison F Vitonis
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital, Boston, MA
| | - Linda Titus
- Departments of Epidemiology and Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Kathryn L Terry
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA
| | - Daniel W Cramer
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA
| |
Collapse
|
63
|
Li TC, Li CI, Liu CS, Lin WY, Lin CH, Yang SY, Lin CC. Risk score system for the prediction of hepatocellular carcinoma in patients with type 2 diabetes: Taiwan Diabetes Study. Semin Oncol 2018; 45:264-274. [PMID: 30342872 DOI: 10.1053/j.seminoncol.2018.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 01/10/2023]
Abstract
This study aims to develop a risk score system for hepatocellular carcinoma (HCC) in patients with type 2 diabetes using the Taiwan National Diabetes Care Management Program database. This retrospective cohort study included 31,723 Chinese patients who had type 2 diabetes, aged 30-84 years. Participants were randomly grouped into derivation and validation sets in 2:1 ratio. Cox proportional hazard regression models were used to identify the risk factors of HCC in the derivation set. Discrimination ability of the model was assessed by means of a receiver operating characteristic curve and performance was expressed as the c statistic, assessed internally on validation data sets. The average follow-up was 8.33 years with 748 HCC incident cases in the derivation set. The final HCC risk score system included age (-2 to 8 points), gender (0-2 points), smoking (0-2 points), variation in hemoglobin A1c (0-1 point), serum glutamic-pyruvic transaminase (0-6 points), liver cirrhosis (9 points), hepatitis B (4 points), hepatitis C (3 points), antidiabetes medications (0-3 points), and antihyperlipidemia medications and total/high-density lipoprotein cholesterol ratio (-4 to 2 points). The HCC risk score was the sum of these individual scores (range -6 to 40). The area under the receiver operating characteristic curve for 3-, 5-, and 10-year HCC risks was 0.81, 0.80, and 0.77 for the derivation set, respectively. This HCC risk score system has good prediction accuracy and discriminatory ability, and serves a simple tool for HCC risk prediction.
Collapse
Affiliation(s)
- Tsai-Chung Li
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan; Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chia-Ing Li
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chiu-Shong Liu
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Yuan Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsueh Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Sing-Yu Yang
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan
| | - Cheng-Chieh Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
64
|
Abstract
Statin drugs have been used for more than two decades to treat hypercholesterolemia and as cardio-preventive drugs, resulting in a marked decrease in cardiovascular morbidity and mortality worldwide. Statins halt hepatic cholesterol biosynthesis by inhibiting the rate-limiting enzyme in the mevalonate pathway, hydroxymethylglutaryl-coenzyme A reductase (HMGCR). The mevalonate pathway regulates a host of biochemical processes in addition to cholesterol production. Attenuation of these pathways is likely responsible for the myriad benefits of statin therapy beyond cholesterol reduction - the so-called pleiotropic effects of statins. Chief amongst these purported effects is anti-cancer activity. A considerable body of preclinical, epidemiologic and clinical evidence shows that statins impair proliferation of breast cancer cells and reduce the risk of breast cancer recurrence. Potential mechanisms for this effect have been explored in laboratory models, but remain poorly understood and require further investigation. The number of clinical trials assessing the putative clinical benefit of statins in breast cancer is increasing. Currently, a total of 30 breast cancer/statin trials are listed at the global trial identifier website clinicaltrials.gov. Given the compelling evidence from performed trials in a variety of clinical settings, there have been calls for a clinical trial of statins in the adjuvant breast cancer setting. It would be imperative for such a trial to incorporate tumour biomarkers predictive of statin response in its design and analysis plan. Ongoing translational clinical trials aimed at biomarker discovery will help identify, which breast cancer patients are most likely to benefit from adjuvant statin therapy, and will add valuable clinical knowledge to the field.
Collapse
Affiliation(s)
- S. Borgquist
- Department of OncologyAarhus University HospitalAarhus CDenmark
- Division of Oncology and PathologyLund UniversityLundSweden
| | | | - S. Kimbung
- Division of Oncology and PathologyLund UniversityLundSweden
| | - T. P. Ahern
- Departments of Surgery and BiochemistryThe Robert Larner, MD College of MedicineThe University of VermontBurlingtonVTUSA
| |
Collapse
|
65
|
Xia Y, Xie Y, Yu Z, Xiao H, Jiang G, Zhou X, Yang Y, Li X, Zhao M, Li L, Zheng M, Han S, Zong Z, Meng X, Deng H, Ye H, Fa Y, Wu H, Oldfield E, Hu X, Liu W, Shi Y, Zhang Y. The Mevalonate Pathway Is a Druggable Target for Vaccine Adjuvant Discovery. Cell 2018; 175:1059-1073.e21. [PMID: 30270039 DOI: 10.1016/j.cell.2018.08.070] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/13/2018] [Accepted: 08/30/2018] [Indexed: 01/02/2023]
Abstract
Motivated by the clinical observation that interruption of the mevalonate pathway stimulates immune responses, we hypothesized that this pathway may function as a druggable target for vaccine adjuvant discovery. We found that lipophilic statin drugs and rationally designed bisphosphonates that target three distinct enzymes in the mevalonate pathway have potent adjuvant activities in mice and cynomolgus monkeys. These inhibitors function independently of conventional "danger sensing." Instead, they inhibit the geranylgeranylation of small GTPases, including Rab5 in antigen-presenting cells, resulting in arrested endosomal maturation, prolonged antigen retention, enhanced antigen presentation, and T cell activation. Additionally, inhibiting the mevalonate pathway enhances antigen-specific anti-tumor immunity, inducing both Th1 and cytolytic T cell responses. As demonstrated in multiple mouse cancer models, the mevalonate pathway inhibitors are robust for cancer vaccinations and synergize with anti-PD-1 antibodies. Our research thus defines the mevalonate pathway as a druggable target for vaccine adjuvants and cancer immunotherapies.
Collapse
Affiliation(s)
- Yun Xia
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yonghua Xie
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Zhengsen Yu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Hongying Xiao
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Guimei Jiang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Xiaoying Zhou
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Yunyun Yang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Xin Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Meng Zhao
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Liping Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Mingke Zheng
- Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Shuai Han
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Zhaoyun Zong
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Huahu Ye
- Laboratory Animal Center, Academy of Military Medical Sciences, 100071 Beijing, China
| | - Yunzhi Fa
- Laboratory Animal Center, Academy of Military Medical Sciences, 100071 Beijing, China
| | - Haitao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, 100850 Beijing, China
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiaoyu Hu
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China; Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China.
| | - Yan Shi
- Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China; Institute Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, AB, Canada.
| | - Yonghui Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China.
| |
Collapse
|
66
|
Yarmolinsky J, Wade KH, Richmond RC, Langdon RJ, Bull CJ, Tilling KM, Relton CL, Lewis SJ, Davey Smith G, Martin RM. Causal Inference in Cancer Epidemiology: What Is the Role of Mendelian Randomization? Cancer Epidemiol Biomarkers Prev 2018; 27:995-1010. [PMID: 29941659 PMCID: PMC6522350 DOI: 10.1158/1055-9965.epi-17-1177] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/15/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023] Open
Abstract
Observational epidemiologic studies are prone to confounding, measurement error, and reverse causation, undermining robust causal inference. Mendelian randomization (MR) uses genetic variants to proxy modifiable exposures to generate more reliable estimates of the causal effects of these exposures on diseases and their outcomes. MR has seen widespread adoption within cardio-metabolic epidemiology, but also holds much promise for identifying possible interventions for cancer prevention and treatment. However, some methodologic challenges in the implementation of MR are particularly pertinent when applying this method to cancer etiology and prognosis, including reverse causation arising from disease latency and selection bias in studies of cancer progression. These issues must be carefully considered to ensure appropriate design, analysis, and interpretation of such studies. In this review, we provide an overview of the key principles and assumptions of MR, focusing on applications of this method to the study of cancer etiology and prognosis. We summarize recent studies in the cancer literature that have adopted a MR framework to highlight strengths of this approach compared with conventional epidemiological studies. Finally, limitations of MR and recent methodologic developments to address them are discussed, along with the translational opportunities they present to inform public health and clinical interventions in cancer. Cancer Epidemiol Biomarkers Prev; 27(9); 995-1010. ©2018 AACR.
Collapse
Affiliation(s)
- James Yarmolinsky
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ryan J Langdon
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Caroline J Bull
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kate M Tilling
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Sarah J Lewis
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Richard M Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
67
|
Hamada T, Khalaf N, Yuan C, Babic A, Morales-Oyarvide V, Qian ZR, Nowak JA, Ng K, Kraft P, Rubinson DA, Stampfer MJ, Giovannucci EL, Fuchs CS, Ogino S, Wolpin BM. Statin use and pancreatic cancer risk in two prospective cohort studies. J Gastroenterol 2018; 53:959-966. [PMID: 29362938 PMCID: PMC7609961 DOI: 10.1007/s00535-018-1430-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/05/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Statins, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, are common lipid-lowering agents and may reduce the risk of several cancer types including pancreatic cancer. However, the association between statin use and pancreatic cancer risk has not been fully evaluated in prospective studies. METHODS We studied the association between statin use and incident pancreatic cancer in 113,059 participants from the prospective Nurses' Health Study and Health Professionals Follow-up Study. Statin use was self-reported via study questionnaires and updated biennially. Hazard ratios (HRs) and 95% confidence intervals (CIs) for incidence of pancreatic cancer were estimated using multivariable Cox proportional hazards models with adjustment for potential confounders. RESULTS In total, 583 participants developed incident pancreatic cancer during 1.4 million person-years of follow-up. No difference was identified in pancreatic cancer risk for regular versus non-regular statin users (multivariable-adjusted HR 0.98; 95% CI 0.82-1.16). There was no significant heterogeneity in the association of statin use with pancreatic cancer risk between the cohorts. Similarly, longer duration of regular statin use was not associated with decreased risk of pancreatic cancer (Ptrend = 0.65). The results remained similar when we examined statin use status at baseline or accounting for 4-year latency period. We observed no statistically significant effect modification for the association of statin use with pancreatic cancer risk by body mass index, smoking status, or diabetes mellitus status (all Pinteraction > 0.21). CONCLUSIONS Regular statin use was not associated with pancreatic cancer risk in two large prospective cohort studies in the U.S.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Natalia Khalaf
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Vicente Morales-Oyarvide
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Zhi Rong Qian
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jonathan A. Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Douglas A. Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Meir J. Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Charles S. Fuchs
- Yale Cancer Center, New Haven, CT, USA,Department of Medicine, Yale School of Medicine, New Haven, CT, USA,Smilow Cancer Hospital, New Haven, CT, USA
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA,Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
68
|
Hamada T, Khalaf N, Yuan C, Morales-Oyarvide V, Babic A, Nowak JA, Qian ZR, Ng K, Rubinson DA, Kraft P, Giovannucci EL, Stampfer MJ, Fuchs CS, Ogino S, Wolpin BM. Prediagnosis Use of Statins Associates With Increased Survival Times of Patients With Pancreatic Cancer. Clin Gastroenterol Hepatol 2018; 16:1300-1306.e3. [PMID: 29474971 PMCID: PMC6056316 DOI: 10.1016/j.cgh.2018.02.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/29/2018] [Accepted: 02/11/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Statin medications, most commonly prescribed to reduce lipid levels and prevent cardiovascular disease, may be associated with longer survival times of patients with cancer. However, the association of statins with outcomes of patients with pancreatic adenocarcinoma is not clear. METHODS We analyzed the association of statin use before a diagnosis of pancreatic cancer with survival times of 648 participants in the Nurses' Health Study and Health Professionals Follow-up Study who were diagnosed with pancreatic adenocarcinoma from 2000 through 2013. We estimated hazard ratios (HRs) for overall mortality using Cox proportional hazards models with adjustment for potential confounders. We assessed the temporal association between prediagnosis statin use and cancer survival by 2-year lag periods to account for a possible latency period between statin use and cancer survival. RESULTS Regular statin use before diagnosis of pancreatic cancer was associated with modestly prolonged survival compared with nonregular use (adjusted HR, 0.82; 95% CI, 0.69-0.97; P = .02). A 1-month longer median survival was observed in regular statin users compared with nonregular users. Regular statin use within the 2 years prior to cancer diagnosis was most strongly associated with longer survival. We observed no statistically significant effect modification by smoking status, body mass index, diabetes, or cancer stage (all Pinteraction > .53). Regular statin use before diagnosis was similarly associated with survival in the Nurses' Health Study (HR, 0.79; 95% CI, 0.64-0.97) and Health Professionals Follow-up Study (HR, 0.86; 95% CI, 0.63-1.15). CONCLUSIONS Regular statin use before diagnosis of pancreatic cancer was associated with modest increases in survival times in 2 large prospective cohort studies.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Natalia Khalaf
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Vicente Morales-Oyarvide
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zhi Rong Qian
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Douglas A Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Peter Kraft
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts; Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Meir J Stampfer
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, Connecticut; Department of Medicine, Yale School of Medicine, New Haven, Connecticut; Smilow Cancer Hospital, New Haven, Connecticut
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
69
|
Jian-Yu E, Graber JM, Lu SE, Lin Y, Lu-Yao G, Tan XL. Effect of Metformin and Statin Use on Survival in Pancreatic Cancer Patients: a Systematic Literature Review and Meta-analysis. Curr Med Chem 2018; 25:2595-2607. [PMID: 28403788 DOI: 10.2174/0929867324666170412145232] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Current epidemiological studies report conflicting results for the effect of statin or metformin on pancreatic cancer overall survival. This literature review and meta-analysis summarize the studies reporting an association between statin or metformin use and overall survival of pancreatic cancer patients. METHODS We systematically searched for studies about the association between statin or metformin use and pancreatic cancer overall survival in electronic databases (PubMed, ISI Web of Science, MEDLINE, Cochrane, Scopus, Google Scholar). A meta-analysis based on hazard ratios (HRs) and 95% confidence intervals (CIs) was performed using random effect models. Heterogeneity between the studies was examined using I2 statistics, and sensitivity analyses were conducted to assess the robustness of the findings. RESULTS Of 116 statin-related articles identified, 6 retrospective cohort studies representing 12,057 patients were included. There was significant heterogeneity between studies. Statin use was associated with improved survival among pancreatic cancer patients (meta-HR = 0.75; 95% CI: 0.59, 0.90; P < 0.001). Of 311 metformin-related articles, 8 retrospective cohort studies and 2 randomized clinical trials, representing 3,042 patients were identified. Metformin use was associated with better overall survival among pancreatic cancer patients (meta-HR = 0.79; 95% CI: 0.70, 0.92, P < 0.001), and significant heterogeneity was observed between studies. CONCLUSION Our findings suggest that the improved survival time of pancreatic cancer patients are associated with statin or metformin use. Due to the multiple sources of heterogeneity of the original studies, these findings should be considered cautiously, and confirmed with larger prospective individual-level studies.
Collapse
Affiliation(s)
- Jian-Yu E
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States.,Department of Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States
| | - Judith M Graber
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States.,Department of Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States.,Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, United States
| | - Shou-En Lu
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States.,Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States
| | - Yong Lin
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States.,Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States
| | - Grace Lu-Yao
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States.,Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States
| | - Xiang-Lin Tan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States.,Department of Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States.,Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08904, United States
| |
Collapse
|
70
|
Zhang XS, Zhang YM, Li B, Fan B, Zhao Y, Yang SJ. Risk reduction of endometrial and ovarian cancer after bisphosphonates use: A meta-analysis. Gynecol Oncol 2018; 150:509-514. [PMID: 29960711 DOI: 10.1016/j.ygyno.2018.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Recent epidemiological studies have investigated the associations between the use of bisphosphonates and the development of endometrial cancer and ovarian cancer; these studies have shown controversial results. Hence, this meta-analysis was conducted to evaluate the changes in the risks of developing endometrial and ovarian cancers after using bisphosphonates based on current evidence. METHODS A comprehensive search was performed in the MEDLINE, EMBASE, and Web of Science databases through January 2017. The summary relative risk (RR) estimates for the effects of the use of bisphosphonates on the risks of developing endometrial and ovarian cancers were calculated using a random-effects model. RESULTS Seven studies were included with a total of 6471 endometrial cancer cases (7 studies with 213,920 participants) and 6783 ovarian cancer cases (4 studies with 105,507 participants). This meta-analysis suggested that any use of bisphosphonates was associated with a significant 27% reduction in the risk of endometrial cancer (RR = 0.73, 95% CI: 0.58-0.93, P = 0.012), but the reduction in the risk of ovarian cancer (RR = 0.81, 95% CI: 0.58-1.14, P = 0.227) was not significant. The protective effects of the use of bisphosphonates against endometrial cancer are mainly found in postmenopausal women (RR = 0.53, 95% CI: 0.34-0.93, P = 0.012) or in those who have taken bisphosphonates for longer than 1 year (RR = 0.57, 95% CI: 0.35-0.93, P = 0.024). CONCLUSION This meta-analysis suggests that the use of bisphosphonates is associated with a reduction in the risk of endometrial cancer but not ovarian cancer.
Collapse
Affiliation(s)
- Xiao-San Zhang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yi-Ming Zhang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Bin Li
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Bo Fan
- Department of Internal Medicine, Luoshan County Traditional Chinese Medicine Hospital, Luoshan County, Xinyang, Henan 464200, China
| | - Yan Zhao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Shu-Jun Yang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, China.
| |
Collapse
|
71
|
Rivas M, del Valle LJ, Armelin E, Bertran O, Turon P, Puiggalí J, Alemán C. Hydroxyapatite with Permanent Electrical Polarization: Preparation, Characterization, and Response against Inorganic Adsorbates. Chemphyschem 2018; 19:1746-1755. [DOI: 10.1002/cphc.201800196] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Manuel Rivas
- Department of Chemical EngineeringUniversitat Politècnica de Catalunya EEBE, C/Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
| | - Luis J. del Valle
- Department of Chemical EngineeringUniversitat Politècnica de Catalunya EEBE, C/Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
- Barcelona Research Center for Multiscale Science and EngineeringUniversitat Politècnica de Catalunya Institution EEBE, C/ Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
| | - Elaine Armelin
- Department of Chemical EngineeringUniversitat Politècnica de Catalunya EEBE, C/Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
- Barcelona Research Center for Multiscale Science and EngineeringUniversitat Politècnica de Catalunya Institution EEBE, C/ Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
| | - Oscar Bertran
- Department of PhysicsUniversitat Politècnica de Catalunya EEI, Av. Pla de la Massa, 8 08700 Igualada Spain
| | - Pau Turon
- Department of Chemical EngineeringUniversitat Politècnica de Catalunya EEBE, C/Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
- B. Braun Surgical, S.A Carretera de Terrassa 121 08191 Rubí (Barcelona) Spain
| | - Jordi Puiggalí
- Department of Chemical EngineeringUniversitat Politècnica de Catalunya EEBE, C/Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
- Barcelona Research Center for Multiscale Science and EngineeringUniversitat Politècnica de Catalunya Institution EEBE, C/ Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
| | - Carlos Alemán
- Department of Chemical EngineeringUniversitat Politècnica de Catalunya EEBE, C/Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
- Barcelona Research Center for Multiscale Science and EngineeringUniversitat Politècnica de Catalunya Institution EEBE, C/ Eduard Maristany 10–14, Ed. I2 08019 Barcelona Spain
| |
Collapse
|
72
|
Abstract
Inhibitors of mTOR, including clinically available rapalogs such as rapamycin (Sirolimus) and Everolimus, are gerosuppressants, which suppress cellular senescence. Rapamycin slows aging and extends life span in a variety of species from worm to mammals. Rapalogs can prevent age-related diseases, including cancer, atherosclerosis, obesity, neurodegeneration and retinopathy and potentially rejuvenate stem cells, immunity and metabolism. Here, I further suggest how rapamycin can be combined with metformin, inhibitors of angiotensin II signaling (Losartan, Lisinopril), statins (simvastatin, atorvastatin), propranolol, aspirin and a PDE5 inhibitor. Rational combinations of these drugs with physical exercise and an anti-aging diet (Koschei formula) can maximize their anti-aging effects and decrease side effects.
Collapse
|
73
|
Lin KC, Park HW, Guan KL. Deregulation and Therapeutic Potential of the Hippo Pathway in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kimberly C. Lin
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
74
|
Park JH, Shin JE, Park HW. The Role of Hippo Pathway in Cancer Stem Cell Biology. Mol Cells 2018; 41:83-92. [PMID: 29429151 PMCID: PMC5824027 DOI: 10.14348/molcells.2018.2242] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/19/2017] [Accepted: 01/08/2018] [Indexed: 02/08/2023] Open
Abstract
The biological significance and deregulation of the Hippo pathway during organ growth and tumorigenesis have received a surge of interest in the past decade. The Hippo pathway core kinases, MST1/2 and LATS1/2, are tumor suppressors that inhibit the oncogenic nuclear function of YAP/TAZ and TEAD. In addition to earlier studies that highlight the role of Hippo pathway in organ size control, cell proliferation, and tumor development, recent evidence demonstrates its critical role in cancer stem cell biology, including EMT, drug resistance, and self-renewal. Here we provide a brief overview of the regulatory mechanisms of the Hippo pathway, its role in cancer stem cell biology, and promising therapeutic interventions.
Collapse
Affiliation(s)
- Jae Hyung Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Ji Eun Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| |
Collapse
|
75
|
Liu JC, Hao WR, Hsu YP, Sung LC, Kao PF, Lin CF, Wu ATH, Yuan KSP, Wu SY. Statins dose-dependently exert a significant chemopreventive effect on colon cancer in patients with chronic obstructive pulmonary disease: A population-based cohort study. Oncotarget 2018; 7:65270-65283. [PMID: 27542242 PMCID: PMC5323154 DOI: 10.18632/oncotarget.11263] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/27/2016] [Indexed: 12/21/2022] Open
Abstract
Purpose We evaluated the chemopreventive effect of statins on colon cancer in patients with chronic obstructive pulmonary disease (COPD) and identified the statin exerting the strongest chemopreventive effect. Methods Using the National Health Insurance Research Database, we identified patients who received a COPD diagnosis in Taiwan between January 1, 2001, and December 31, 2012, and included them in the study cohort. Each patient was followed to assess the colon cancer risk and protective factors. A propensity score was derived using a logistic regression model to estimate the effect of statins by accounting for covariates predicted during the intervention (statins). To examine the dose–response relationship, we categorized statin doses into four groups in each cohort [<28, 28–90, 91–365, and >365 cumulative defined daily dose]. Results Compared with the statin nonusers, the adjusted hazard ratio (aHR) for colon cancer decreased in the statin users (aHR = 0.52, 95% confidence interval = 0.44, 0.62). Hydrophilic statins exerted a stronger preventive effect against colon cancer. Regarding the statin type, lovastatin, pravastatin, and fluvastatin nonsignificantly reduced the colon cancer risk in the patients with COPD. Compared with the statin nonusers, the aHRs for colon cancer decreased in the individual statin users (rosuvastatin, simvastatin, and atorvastatin: aHRs = 0.28, 0.64, and 0.65, respectively). In the sensitivity analysis, statins dose-dependently reduced the colon cancer risk. Conclusions Statins dose-dependently exert significant chemopreventive effects on colon cancer in patients with COPD, with rosuvastatin exerting the largest chemopreventive effect.
Collapse
Affiliation(s)
- Ju-Chi Liu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Rui Hao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| | - Yi-Ping Hsu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| | - Li-Chin Sung
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pai-Feng Kao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Feng Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| | - Alexander T H Wu
- Ph.D. Program for Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kevin Sheng-Po Yuan
- Department of Otorhinolaryngology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Szu-Yuan Wu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Biotechnology, Hungkuang University, Taichung, Taiwan
| |
Collapse
|
76
|
Lipid-lowering drugs, dyslipidemia, and breast cancer risk in a Medicare population. Breast Cancer Res Treat 2018; 169:607-614. [PMID: 29450675 DOI: 10.1007/s10549-018-4680-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/18/2018] [Indexed: 01/25/2023]
Abstract
PURPOSE We sought to disentangle the effects of statins and other lipid-lowering drugs and the underlying dyslipidemia for which they are prescribed on breast cancer risk. METHODS We conducted a case-control study within the linked Surveillance, Epidemiology, and End results (SEER)-Medicare data. Cases were women with invasive breast cancer aged 66 + years (N = 30,004) identified by SEER registries (years 2007-2011). Controls were women (N = 198,969) identified from a 5% random sample of Medicare recipients alive and breast cancer free in year of selection. Participants had a minimum of 13 months of Part A, Part B non-health maintenance organization Medicare and Part D Medicare coverage at least 13 months preceding cancer diagnosis/selection. Exposures were assessed until 12 months before diagnosis/control selection. Odds ratios (OR) and 99.9% confidence intervals (CI) were estimated using adjusted unconditional and multinomial logistic regression. RESULTS ORs of invasive breast cancer associated with dyslipidemia, statins, and non-statin lipid-lowering drugs were 0.86 (99.9% CI 0.81-0.90), 1.07 (99.9% CI 1.03-1.13) and 1.03 (99.9% CI 0.95-1.11), respectively. Risk reductions with dyslipidemia were slightly greater when untreated than treated and did not vary much by time between dyslipidemia and breast cancer diagnosis. Whether treated or untreated, dyslipidemia was associated with greater reductions in risk for later stage than earlier stage breast cancer (p-heterogeneity < 0.0001). CONCLUSIONS Lipid-lowering drugs did not account for the lower breast cancer risk associated with dyslipidemia. Our data do not support using statins or other lipid-lowering drugs to prevent breast cancer.
Collapse
|
77
|
Understanding the Progression of Bone Metastases to Identify Novel Therapeutic Targets. Int J Mol Sci 2018; 19:ijms19010148. [PMID: 29300334 PMCID: PMC5796097 DOI: 10.3390/ijms19010148] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/22/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022] Open
Abstract
Bone is one of the most preferential target site for cancer metastases, particularly for prostate, breast, kidney, lung and thyroid primary tumours. Indeed, numerous chemical signals and growth factors produced by the bone microenvironment constitute factors promoting cancer cell invasion and aggression. After reviewing the different theories proposed to provide mechanism for metastatic progression, we report on the gene expression profile of bone-seeking cancer cells. We also discuss the cross-talk between the bone microenvironment and invading cells, which impacts on the tumour actions on surrounding bone tissue. Lastly, we detail therapies for bone metastases. Due to poor prognosis for patients, the strategies mainly aim at reducing the impact of skeletal-related events on patients' quality of life. However, recent advances have led to a better understanding of molecular mechanisms underlying bone metastases progression, and therefore of novel therapeutic targets.
Collapse
|
78
|
Ogino S, Jhun I, Mata DA, Soong TR, Hamada T, Liu L, Nishihara R, Giannakis M, Cao Y, Manson JE, Nowak JA, Chan AT. Integration of pharmacology, molecular pathology, and population data science to support precision gastrointestinal oncology. NPJ Precis Oncol 2017; 1. [PMID: 29552640 PMCID: PMC5856171 DOI: 10.1038/s41698-017-0042-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Precision medicine has a goal of customizing disease prevention and treatment strategies. Under the precision medicine paradigm, each patient has unique pathologic processes resulting from cellular genomic, epigenomic, proteomic, and metabolomic alterations, which are influenced by pharmacological, environmental, microbial, dietary, and lifestyle factors. Hence, to realize the promise of precision medicine, multi-level research methods that can comprehensively analyze many of these variables are needed. In order to address this gap, the integrative field of molecular pathology and population data science (i.e., molecular pathological epidemiology) has been developed to enable such multi-level analyses, especially in gastrointestinal cancer research. Further integration of pharmacology can improve our understanding of drug effects, and inform decision-making of drug use at both the individual and population levels. Such integrative research demonstrated potential benefits of aspirin in colorectal carcinoma with PIK3CA mutations, providing the basis for new clinical trials. Evidence also suggests that HPGD (15-PDGH) expression levels in normal colon and the germline rs6983267 polymorphism that relates to tumor CTNNB1 (β-catenin)/WNT signaling status may predict the efficacy of aspirin for cancer chemoprevention. As immune checkpoint blockade targeting the CD274 (PD-L1)/PDCD1 (PD-1) pathway for microsatellite instability-high (or mismatch repair-deficient) metastatic gastrointestinal or other tumors has become standard of care, potential modifying effects of dietary, lifestyle, microbial, and environmental factors on immunotherapy need to be studied to further optimize treatment strategies. With its broad applicability, our integrative approach can provide insights into the interactive role of medications, exposures, and molecular pathology, and guide the development of precision medicine.
Collapse
Affiliation(s)
- Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Iny Jhun
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Douglas A Mata
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thing Rinda Soong
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Li Liu
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Reiko Nishihara
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marios Giannakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yin Cao
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
79
|
Zhou YY, Zhu GQ, Wang Y, Zheng JN, Ruan LY, Cheng Z, Hu B, Fu SW, Zheng MH. Systematic review with network meta-analysis: statins and risk of hepatocellular carcinoma. Oncotarget 2017; 7:21753-62. [PMID: 26943041 PMCID: PMC5008320 DOI: 10.18632/oncotarget.7832] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/21/2016] [Indexed: 02/06/2023] Open
Abstract
Objectives Usage of statins is suggested to decrease the incidence of HCC. When it comes to different statin subtypes, the chemopreventive action remains controversial. We aim to compare the usage of different statins and reduction of HCC risk. Methods We searched PubMed, Embase.com and Cochrane Library database up to August 10, 2015. Duplicated or overlapping reports were eliminated. We performed a traditional pair-wise meta-analysis and a Bayesian network meta-analysis to compare different treatments with a random-effects model. Results We reviewed five observational studies enrolling a total of 87127 patients who received at least two different treatment strategies including rosuvastatin, atorvastatin, simvastatin, pravastatin, fluvastatin, cerivastatin, and lovastatin or observation alone. Direct comparisons showed that usage of atorvastatin (OR 0.63, 95%CI 0.45-0.89) and fluvastatin (OR 0.58, 95%CI 0.40-0.85) could significantly cut the risk of liver cancer. The difference of indirect comparisons between the included regimens is not statistically significant. However, usage of all types of statins, such as fluvastatin (RR 0.55, 95%CI 0.26-1.11), atorvastatin (RR 0.59, 95%CI 0.30-1.16), simvastatin (RR 0.69, 95%CI 0.38-1.25), cerivastatin (RR 0.71, 95%CI 0.19-2.70), pravastatin (RR 0.72, 95%CI 0.37-1.45), lovastatin (RR 0.81, 95%CI 0.34-1.96) and rosuvastatin (RR 0.92, 95%CI 0.44-1.80), appeared to be superior to observation alone. Notably, fluvastatin was hierarchically the best when compared with the six other statins. Conclusions Our analyses indicate the superiority of usage of statins in reduction of liver cancer. Available evidence supports that fluvastatin is the most effective strategy for reducing HCC risk compared with other statin interventions.
Collapse
Affiliation(s)
- Yao-Yao Zhou
- Department of Cardiology, Jinhua Municipal Hospital, Jinhua 321004, China
| | - Gui-Qi Zhu
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,School of The First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yue Wang
- Department of Cardiology, Jinhua Municipal Hospital, Jinhua 321004, China
| | - Ji-Na Zheng
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,School of The First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Lu-Yi Ruan
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,School of The First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Zhang Cheng
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,School of The First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Bin Hu
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,School of The First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Shen-Wen Fu
- Department of Cardiology, Jinhua Municipal Hospital, Jinhua 321004, China
| | - Ming-Hua Zheng
- Department of Infection and Liver Diseases, Liver Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.,Institute of Hepatology, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
80
|
Fournier A, Mesrine S, Gelot A, Fagherazzi G, Baglietto L, Clavel-Chapelon F, Boutron-Ruault MC, Chabbert-Buffet N. Use of Bisphosphonates and Risk of Breast Cancer in a French Cohort of Postmenopausal Women. J Clin Oncol 2017; 35:3230-3239. [DOI: 10.1200/jco.2016.71.4337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose To assess whether bisphosphonate (BP) use is associated with decreased breast cancer incidence in a cohort of postmenopausal women. Methods The study population included 64,438 postmenopausal women participating in the French E3N (Etude Epidémiologique auprès de femmes de la Mutuelle Générale de l'Education Nationale) prospective cohort, with data self-reported in biennial questionnaires matched with data from a drug reimbursement database. Exposure to BPs and the use of other osteoporosis treatments during follow-up were determined using reimbursement data. Other covariates (breast cancer risk factors, clinical risk factors for osteoporotic fractures, and bone mineral density surveillance) originated from the questionnaires. Hazard ratios (HRs) of breast cancer were estimated using Cox proportional hazards models, considering exposure as a time-varying variable. Results Over an average of 7.2 years of follow-up (2004 to 2011), 2,407 first primary breast cancer cases were identified. The HR of breast cancer associated with exposure to BPs was 0.98 (95% CI, 0.85 to 1.12). We found no effect modification by age, body mass index, time since menopause, use of hormone replacement therapy, use of calcium supplements, or use of vitamin D supplements. There was no heterogeneity across BP molecules and no trend according to cumulative dose, duration of use, or time since last use. We observed a decrease in breast cancer risk restricted to the year after treatment initiation (HR, 0.56; 95% CI, 0.36 to 0.87), which was likely explained by healthy screenee bias. Finally, we did not find any variation in HRs across breast carcinomas defined by their estrogen receptor or invasive or in situ status. Conclusion In our observational cohort of postmenopausal women observed from 2004 to 2011, BP use, likely prescribed for the management of osteoporosis, was not associated with decreased breast cancer incidence.
Collapse
Affiliation(s)
- Agnès Fournier
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| | - Sylvie Mesrine
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| | - Amandine Gelot
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| | - Guy Fagherazzi
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| | - Laura Baglietto
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| | - Françoise Clavel-Chapelon
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| | - Marie-Christine Boutron-Ruault
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| | - Nathalie Chabbert-Buffet
- Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Centre for Research in Epidemiology and Population Health (CESP), Inserm, Université Paris-Sud, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Université Paris-Saclay; Agnès Fournier, Sylvie Mesrine, Amandine Gelot, Guy Fagherazzi, Laura Baglietto, Françoise Clavel-Chapelon, and Marie-Christine Boutron-Ruault, Gustave Roussy, Villejuif; Sylvie Mesrine
| |
Collapse
|
81
|
Islam MM, Yang HC, Nguyen PA, Poly TN, Huang CW, Kekade S, Khalfan AM, Debnath T, Li YCJ, Abdul SS. Exploring association between statin use and breast cancer risk: an updated meta-analysis. Arch Gynecol Obstet 2017; 296:1043-1053. [PMID: 28940025 DOI: 10.1007/s00404-017-4533-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/12/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE The benefits of statin treatment for preventing cardiac disease are well established. However, preclinical studies suggested that statins may influence mammary cancer growth, but the clinical evidence is still inconsistent. We, therefore, performed an updated meta-analysis to provide a precise estimate of the risk of breast cancer in individuals undergoing statin therapy. METHODS For this meta-analysis, we searched PubMed, the Cochrane Library, Web of Science, Embase, and CINAHL for published studies up to January 31, 2017. Articles were included if they (1) were published in English; (2) had an observational study design with individual-level exposure and outcome data, examined the effect of statin therapy, and reported the incidence of breast cancer; and (3) reported estimates of either the relative risk, odds ratios, or hazard ratios with 95% confidence intervals (CIs). We used random-effect models to pool the estimates. RESULTS Of 2754 unique abstracts, 39 were selected for full-text review, and 36 studies reporting on 121,399 patients met all inclusion criteria. The overall pooled risks of breast cancer in patients using statins were 0.94 (95% CI 0.86-1.03) in random-effect models with significant heterogeneity between estimates (I 2 = 83.79%, p = 0.0001). However, we also stratified by region, the duration of statin therapy, methodological design, statin properties, and individual stain use. CONCLUSIONS Our results suggest that there is no association between statin use and breast cancer risk. However, observational studies cannot clarify whether the observed epidemiologic association is a causal effect or the result of some unmeasured confounding variable. Therefore, more research is needed.
Collapse
Affiliation(s)
- Md Mohaimenul Islam
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan.,International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei, Taiwan
| | - Hsuan-Chia Yang
- International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei, Taiwan
| | - Phung-Anh Nguyen
- International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei, Taiwan
| | - Tahmina Nasrin Poly
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan
| | - Chih-Wei Huang
- International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei, Taiwan
| | - Shwetambara Kekade
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan.,International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei, Taiwan
| | | | - Tonmoy Debnath
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chuan Jack Li
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan.,International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei, Taiwan
| | - Shabbir Syed Abdul
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan. .,International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
82
|
Lawson MA, Ebetino FH, Mazur A, Chantry AD, Paton-Hough J, Evans HR, Lath D, Tsoumpra MK, Lundy MW, Dobson RL, Quijano M, Kwaasi AA, Dunford JE, Duan X, Triffitt JT, Jeans G, Russell RGG. The Pharmacological Profile of a Novel Highly Potent Bisphosphonate, OX14 (1-Fluoro-2-(Imidazo-[1,2-α]Pyridin-3-yl)-Ethyl-Bisphosphonate). J Bone Miner Res 2017; 32:1860-1869. [PMID: 28337806 PMCID: PMC5596338 DOI: 10.1002/jbmr.3138] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023]
Abstract
Bisphosphonates are widely used in the treatment of clinical disorders characterized by increased bone resorption, including osteoporosis, Paget's disease, and the skeletal complications of malignancy. The antiresorptive potency of the nitrogen-containing bisphosphonates on bone in vivo is now recognized to depend upon two key properties, namely mineral binding affinity and inhibitory activity on farnesyl pyrophosphate synthase (FPPS), and these properties vary independently of each other in individual bisphosphonates. The better understanding of structure activity relationships among the bisphosphonates has enabled us to design a series of novel bisphosphonates with a range of mineral binding properties and antiresorptive potencies. Among these is a highly potent bisphosphonate, 1-fluoro-2-(imidazo-[1,2 alpha]pyridin-3-yl)-ethyl-bisphosphonate, also known as OX14, which is a strong inhibitor of FPPS, but has lower binding affinity for bone mineral than most of the commonly studied bisphosphonates. The aim of this work was to characterize OX14 pharmacologically in relation to several of the bisphosphonates currently used clinically. When OX14 was compared to zoledronate (ZOL), risedronate (RIS), and minodronate (MIN), it was as potent at inhibiting FPPS in vitro but had significantly lower binding affinity to hydroxyapatite (HAP) columns than ALN, ZOL, RIS, and MIN. When injected i.v. into growing Sprague Dawley rats, OX14 was excreted into the urine to a greater extent than the other bisphosphonates, indicating reduced short-term skeletal uptake and retention. In studies in both Sprague Dawley rats and C57BL/6J mice, OX14 inhibited bone resorption, with an antiresorptive potency equivalent to or greater than the comparator bisphosphonates. In the JJN3-NSG murine model of myeloma-induced bone disease, OX14 significantly prevented the formation of osteolytic lesions (p < 0.05). In summary, OX14 is a new, highly potent bisphosphonate with lower bone binding affinity than other clinically relevant bisphosphonates. This renders OX14 an interesting potential candidate for further development for its potential skeletal and nonskeletal benefits. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Michelle A Lawson
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK
| | - Frank H Ebetino
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK.,Department of Chemistry, University of Rochester, Rochester, NY, USA
| | | | - Andrew D Chantry
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK
| | - Julia Paton-Hough
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK
| | - Holly R Evans
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK
| | - Darren Lath
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK
| | - Maria K Tsoumpra
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK
| | - Mark W Lundy
- Department of Anatomy and Cell Biology, Indiana University, Indianapolis, IN, USA
| | | | | | - Aaron A Kwaasi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, The Oxford University Institute of Musculoskeletal Sciences, The Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | - James E Dunford
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, The Oxford University Institute of Musculoskeletal Sciences, The Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | - Xuchen Duan
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, The Oxford University Institute of Musculoskeletal Sciences, The Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | - James T Triffitt
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, The Oxford University Institute of Musculoskeletal Sciences, The Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| | | | - R Graham G Russell
- Department of Oncology and Metabolism, Medical School, University of Sheffield, UK.,Mellanby Centre for Bone Research, Medical School, University of Sheffield, UK.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, The Oxford University Institute of Musculoskeletal Sciences, The Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK
| |
Collapse
|
83
|
Abstract
Purpose of Review Since epidemiological studies first demonstrated a potential positive effect of metformin in reducing cancer incidence and mortality, there has been an increased interest in not only better understanding metformin’s mechanisms of action but also in exploring its potential anti-cancer effects. In this review, we aim to summarise the current evidence exploring a role for metformin in prostate cancer therapy. Recent Findings Preclinical studies have demonstrated a number of antineoplastic biological effects via a range of molecular mechanisms. Data from retrospective epidemiological studies in prostate cancer has been mixed; however, there are several clinical trials currently underway evaluating metformin’s role as an anti-cancer agent. Early studies have shown benefits of metformin to inhibit cancer cell proliferation and improve metabolic syndrome in prostate cancer patients receiving androgen deprivation therapy (ADT). Summary While the body of evidence to support a role for metformin in prostate cancer therapy is rapidly growing, there is still insufficient data from randomised trials, which are currently still ongoing. However, evidence so far suggests metformin could be a useful adjuvant agent, particularly in patients on ADT.
Collapse
Affiliation(s)
- Jessica Whitburn
- Nuffield Department of Surgical Sciences, University of Oxford, Botnar Research Centre Old Road, Oxford, OX3 7LD, UK.
| | - Claire M Edwards
- Nuffield Department of Surgical Sciences, University of Oxford, Botnar Research Centre Old Road, Oxford, OX3 7LD, UK
| | - Prasanna Sooriakumaran
- Nuffield Department of Surgical Sciences, University of Oxford, Botnar Research Centre Old Road, Oxford, OX3 7LD, UK.,Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
84
|
Yi C, Song Z, Wan M, Chen Y, Cheng X. Statins intake and risk of liver cancer: A dose-response meta analysis of prospective cohort studies. Medicine (Baltimore) 2017; 96:e7435. [PMID: 28682909 PMCID: PMC5502182 DOI: 10.1097/md.0000000000007435] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/10/2017] [Accepted: 06/15/2017] [Indexed: 12/18/2022] Open
Abstract
Previous studies have indicated that statins intake was associated with liver cancer risk, but presented controversial results.Studies in PubMed and EMBASE were searched update to February 2017 to identify and quantify the potential dose-response association between statins intake and liver cancer.Six eligible studies involving a total of 11,8961 participants with 9530 incident cases were included in this meta-analysis. Statistically significant association was observed between increasing statins intake and liver cancer risk reduction (OR = 0.46, 95%CI: 0.24-0.68, P <.001). Furthermore, the summary relative risk of liver cancer for an increase of 50 cumulative defined daily dose per year was 0.86 (95%CI: 0.81-0.90, P <.001). Evidence of a nonlinear dose-response relationship between statins intake and liver cancer risk was found (P for nonlinearity <.01). Subgroups analysis indicated that statins intake was associated with a significantly risk of liver cancer risk reduction in Asia (OR = 0.44, 95%CI: 0.11-0.77, P <.001) and Caucasian (OR = 0.49, 95%CI: 0.36-0.61, P <.001). Subgroup meta-analyses in study design, study quality, number of participants, and number of cases showed consistency with the primary findings.Additional statins intake is associated with liver cancer risk reduction.
Collapse
Affiliation(s)
- Changhong Yi
- Department of Interventional, Jingzhou Central Hospital
- The Second Clinical Medical College Yangtze University, Jingzhou
| | - Zhenggui Song
- Department of Interventional, Zigui County Hospital of traditional Chinese Medicine, Yichang, Hubei Province, People's Republic of China
| | - Maolin Wan
- Department of Interventional, Jingzhou Central Hospital
| | - Ya Chen
- Department of Interventional, Jingzhou Central Hospital
| | - Xiang Cheng
- Department of Hepatobiliary surgery, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan
| |
Collapse
|
85
|
Horiuchi T, Sakata N, Narumi Y, Kimura T, Hayashi T, Nagano K, Liu K, Nishibori M, Tsukita S, Yamada T, Katagiri H, Shirakawa R, Horiuchi H. Metformin directly binds the alarmin HMGB1 and inhibits its proinflammatory activity. J Biol Chem 2017; 292:8436-8446. [PMID: 28373282 PMCID: PMC5437248 DOI: 10.1074/jbc.m116.769380] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/31/2017] [Indexed: 01/26/2023] Open
Abstract
Metformin is the first-line drug in the treatment of type 2 diabetes. In addition to its hypoglycemic effect, metformin has an anti-inflammatory function, but the precise mechanism promoting this activity remains unclear. High mobility group box 1 (HMGB1) is an alarmin that is released from necrotic cells and induces inflammatory responses by its cytokine-like activity and is, therefore, a target of anti-inflammatory therapies. Here we identified HMGB1 as a novel metformin-binding protein by affinity purification using a biotinylated metformin analogue. Metformin directly bound to the C-terminal acidic tail of HMGB1. Both in vitro and in vivo, metformin inhibited inflammatory responses induced by full-length HMGB1 but not by HMGB1 lacking the acidic tail. In an acetaminophen-induced acute liver injury model in which HMGB1 released from injured cells exacerbates the initial injury, metformin effectively reduced liver injury and had no additional inhibitory effects when the extracellular HMGB1 was blocked by anti-HMGB1-neutralizing antibody. In summary, we report for the first time that metformin suppresses inflammation by inhibiting the extracellular activity of HMGB1. Because HMGB1 plays a major role in inflammation, our results suggest possible new ways to manage HMGB1-induced inflammation.
Collapse
Affiliation(s)
- Takahiro Horiuchi
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Natsumi Sakata
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yoshihiro Narumi
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Tomohiro Kimura
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Takashi Hayashi
- Biomedical Technology Research Center, Tokushima Research Institute
| | - Keisuke Nagano
- First Institute of New Drug Discovery, Otsuka Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | - Keyue Liu
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Sohei Tsukita
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Ryutaro Shirakawa
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Hisanori Horiuchi
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| |
Collapse
|
86
|
Kim MH, Kim J. Role of YAP/TAZ transcriptional regulators in resistance to anti-cancer therapies. Cell Mol Life Sci 2017; 74:1457-1474. [PMID: 27826640 PMCID: PMC11107740 DOI: 10.1007/s00018-016-2412-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/15/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022]
Abstract
A diverse range of drug resistance mechanisms in cancer cells and their microenvironment significantly reduces the effectiveness of anti-cancer therapies. Growing evidence suggests that transcriptional effectors of the Hippo pathway, YAP and TAZ, promote resistance to various anti-cancer therapies, including cytotoxic chemotherapy, molecular targeted therapy, and radiation therapy. Here, we overview the role of YAP and TAZ as drug resistance mediators, and also discuss potential upstream regulators and downstream targets of YAP/TAZ in cancer. The widespread involvement of YAP and TAZ in resistance mechanisms suggests that therapeutic targeting of YAP and TAZ may expedite the development of effective anti-resistance therapies.
Collapse
Affiliation(s)
- Min Hwan Kim
- Graduate School of Medical Science and Engineering, KAIST, 291 Daehak-ro, Taejon, 34141, Republic of Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, KAIST, 291 Daehak-ro, Taejon, 34141, Republic of Korea.
| |
Collapse
|
87
|
Nadar RA, Margiotta N, Iafisco M, van den Beucken JJJP, Boerman OC, Leeuwenburgh SCG. Bisphosphonate-Functionalized Imaging Agents, Anti-Tumor Agents and Nanocarriers for Treatment of Bone Cancer. Adv Healthc Mater 2017; 6. [PMID: 28207199 DOI: 10.1002/adhm.201601119] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Indexed: 12/14/2022]
Abstract
Bone metastases result from the invasion of primary tumors to bone. Current treatment modalities include local treatments such as surgery and radiotherapy, while systemic treatments include chemotherapy and (palliative) treatment of skeletal metastases. Nevertheless, once bone metastases have been established they remain incurable leading to morbidity and mortality. Bisphosphonates are a well-established class of drugs, which are increasingly applied in the treatment of bone cancers owing to their effective inhibition of tumor cells and suppression of bone metastases. The increased understanding of the mechanism of action of bisphosphonates on bone and tumor cells has prompted the development of novel bisphosphonate-functionalized imaging and therapeutic agents. This review provides an update on the preclinical efficacy of bisphosphonate-functionalized fluorophore, anti-tumor agents and nanocarriers for the treatment of bone metastases. After an overview of the general characteristics of bisphosphonates and their mechanisms of action, an outline is provided on the various conjugation strategies that have become available to functionalize imaging agents, anti-tumor agents and nanocarriers with bisphosphonates. Finally, the efficacy of these bisphosphonate-modified agents and carriers in preclinical studies is evaluated by reviewing their potential to target tumors and inhibit tumor growth in clinically relevant animal models for the treatment of bone cancer.
Collapse
Affiliation(s)
- Robin A. Nadar
- Department of Biomaterials; Radboud University Medical Center; Philips van Leydenlaan 25 6525 EX Nijmegen The Netherlands
| | - Nicola Margiotta
- Dipartimento di Chimica; Università degli Studi di Bari Aldo Moro; Via E. Orabona 4 70125 Bari Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics (ISTEC); National Research Council (CNR); Via Granarolo 64 48018 Faenza Italy
| | | | - Otto C. Boerman
- Department of Nuclear Medicine; Radboud University Medical Center; Geert Grooteplein Zuid 10 6525 AG Nijmegen The Netherlands
| | - Sander C. G. Leeuwenburgh
- Department of Biomaterials; Radboud University Medical Center; Philips van Leydenlaan 25 6525 EX Nijmegen The Netherlands
| |
Collapse
|
88
|
Gilmour R, Velasco S. Back to the Future: Therapeutic Targeting of Cancer Cell Metabolism. SLAS DISCOVERY 2017; 22:333-337. [PMID: 28328320 DOI: 10.1177/2472555217691265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Raymond Gilmour
- 1 Discovery Oncology Research, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Susana Velasco
- 2 Cell Signaling Therapies, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
89
|
Hosseini M, Kasraian Z, Rezvani HR. Energy metabolism in skin cancers: A therapeutic perspective. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:712-722. [PMID: 28161328 DOI: 10.1016/j.bbabio.2017.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 12/13/2022]
Abstract
Skin cancers are the most common cancers worldwide. The incidence of common skin cancers, including basal cell carcinomas (BCCs), squamous cell carcinomas (SCCs) and melanomas, continues to rise by 5 to 7% per year, mainly due to ultraviolet (UV) exposure and partly because of aging. This suggests an urgent necessity to improve the level of prevention and protection for skin cancers as well as developing new prognostic and diagnostic markers of skin cancers. Moreover, despite innovative therapies especially in the fields of melanoma and carcinomas, new therapeutic options are needed to bypass resistance to targeted therapies or treatment's side effects. Since reprogramming of cellular metabolism is now considered as a hallmark of cancer, some of the recent findings on the role of energy metabolism in skin cancer initiation and progression as well as its effect on the response to targeted therapies are discussed in this review. This article is part of a Special Issue entitled Mitochondria in cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.
Collapse
Affiliation(s)
- Mohsen Hosseini
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Zeinab Kasraian
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Hamid Reza Rezvani
- Inserm U 1035, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre de Référence pour les Maladies Rares de la Peau, CHU de Bordeaux, France.
| |
Collapse
|
90
|
Gong J, Sachdev E, Robbins LA, Lin E, Hendifar AE, Mita MM. Statins and pancreatic cancer. Oncol Lett 2017; 13:1035-1040. [PMID: 28454210 DOI: 10.3892/ol.2017.5572] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/22/2016] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer remains among the most lethal cancers, despite ongoing advances in treatment for all stages of the disease. Disease prevention represents another opportunity to improve patient outcome, with metabolic syndrome and its components, such as diabetes, obesity and dyslipidemia, having been recognized as modifiable risk factors for pancreatic cancer. In addition, statins have been shown to potentially reduce pancreatic cancer risk and to improve survival in patients with a combination of metabolic syndrome and pancreatic cancer. Furthermore, preclinical studies have demonstrated that statins exhibit antitumor effects in pancreatic cancer cell lines in vitro and animal models in vivo, in addition to delaying the progression of pancreatic intraepithelial neoplasia to pancreatic ductal adenocarcinoma (PDAC) and inhibiting PDAC formation in conditional K-Ras mutant mice. The mechanisms by which statins produce anticancer effects remain poorly understood, although appear to involve inhibition of the mevalonate/cholesterol synthesis pathway, thus blocking the synthesis of intermediates important for prenylation and activation of the Ras/mitogen-activated protein kinase 1 signaling pathway. Furthermore, statins have been identified to modulate the phosphoinositide 3-kinase/Akt serine/threonine kinase 1 and inflammation signaling pathways, and to alter the expression of genes involved in lipid metabolism, which are important for PDAC growth and proliferation. In addition, statins have been demonstrated to exhibit further antitumor mechanisms in a number of other cancer types, which are beyond the scope of the present review. In the present review, current evidence highlighting the potential of statins as chemopreventive agents in pancreatic cancer is presented, and the antitumor mechanisms of statins elucidated thus far in this disease are discussed.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Esha Sachdev
- Department of Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lori A Robbins
- Department of Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Emily Lin
- Department of Internal Medicine, Harbor-UCLA Medical Center, Torrance, CA 90509, USA
| | - Andrew E Hendifar
- Department of Internal Medicine, Division of Medical Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Monica M Mita
- Experimental Therapeutics Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
91
|
Huang BZ, Chang JI, Li E, Xiang AH, Wu BU. Influence of Statins and Cholesterol on Mortality Among Patients With Pancreatic Cancer. J Natl Cancer Inst 2016; 109:2898139. [PMID: 28040693 DOI: 10.1093/jnci/djw275] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 09/04/2016] [Accepted: 10/13/2016] [Indexed: 12/17/2022] Open
Abstract
Background Recent studies have suggested associations between statins and enhanced survival among patients with pancreatic ductal adenocarcinoma (PDAC). However, the relationship between statins, cholesterol, and survival remains unclear. Methods We conducted a retrospective cohort study on 2142 PDAC patients in a regional integrated healthcare system from 2006 to 2014. Electronic pharmacy records were used to abstract information on the type, length, and dosage of statin exposures starting in the year prior to diagnosis. The cumulative and individual effects of simvastatin, lovastatin, atorvastatin, pravastatin, and rosuvastatin on mortality were assessed using Cox proportional hazards regression. Statins were evaluated as any use (pre- and postdiagnosis as a time-dependent variable) and baseline use (prediagnosis only). We also evaluated whether low-density lipoprotein (LDL) cholesterol, measured at various time windows prior to diagnosis, had an independent influence on survival. Additional analyses were performed to examine whether cholesterol mediated the relationship between statins and mortality. All models included age, race, stage, surgery, gemcitabine-based chemotherapy, and the Charlson comorbidity index as covariates. Results Any (hazard ratio [HR] = 0.87, 95% confidence interval [CI] = 0.79 to 0.97) and baseline (HR = 0.88, 95% CI = 0.79 to 0.98) statin use were both associated with a decreased risk in mortality. When assessing individual statins, we found reduced mortality among simvastatin (HR = 0.87, 95% CI = 0.77 to 0.98) and atorvastatin (HR = 0.58, 95% CI = 0.46 to 0.72) users. Cholesterol was not associated with mortality and did not mediate any relationships between statins and survival. Conclusions Statin use rather than cholesterol level was associated with lower mortality risk in patients with pancreatic cancer. Statins appear to improve survival through a lipid-independent mechanism.
Collapse
Affiliation(s)
- Brian Z Huang
- Affiliations of authors: Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA (BZH, AHX); Department of Internal Medicine, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (JIC), and Division of Gastroenterology, Center for Pancreatic Care, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (BUW), Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA (BZH); David Geffen School of Medicine at UCLA, Los Angeles, CA (EL)
| | - Jonathan I Chang
- Affiliations of authors: Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA (BZH, AHX); Department of Internal Medicine, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (JIC), and Division of Gastroenterology, Center for Pancreatic Care, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (BUW), Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA (BZH); David Geffen School of Medicine at UCLA, Los Angeles, CA (EL)
| | - Erica Li
- Affiliations of authors: Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA (BZH, AHX); Department of Internal Medicine, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (JIC), and Division of Gastroenterology, Center for Pancreatic Care, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (BUW), Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA (BZH); David Geffen School of Medicine at UCLA, Los Angeles, CA (EL)
| | - Anny H Xiang
- Affiliations of authors: Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA (BZH, AHX); Department of Internal Medicine, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (JIC), and Division of Gastroenterology, Center for Pancreatic Care, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (BUW), Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA (BZH); David Geffen School of Medicine at UCLA, Los Angeles, CA (EL)
| | - Bechien U Wu
- Affiliations of authors: Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA (BZH, AHX); Department of Internal Medicine, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (JIC), and Division of Gastroenterology, Center for Pancreatic Care, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA (BUW), Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA (BZH); David Geffen School of Medicine at UCLA, Los Angeles, CA (EL)
| |
Collapse
|
92
|
McCaw L, Shi Y, Wang G, Li YJ, Spaner DE. Low Density Lipoproteins Amplify Cytokine-signaling in Chronic Lymphocytic Leukemia Cells. EBioMedicine 2016; 15:24-35. [PMID: 27932296 PMCID: PMC5233814 DOI: 10.1016/j.ebiom.2016.11.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 11/26/2016] [Accepted: 11/28/2016] [Indexed: 11/18/2022] Open
Abstract
Recent studies suggest there is a high incidence of elevated low-density lipoprotein (LDL) levels in Chronic Lymphocytic Leukemia (CLL) patients and a survival benefit from cholesterol-lowering statin drugs. The mechanisms of these observations and the kinds of patients they apply to are unclear. Using an in vitro model of the pseudofollicles where CLL cells originate, LDLs were found to increase plasma membrane cholesterol, signaling molecules such as tyrosine-phosphorylated STAT3, and activated CLL cell numbers. The signaling effects of LDLs were not seen in normal lymphocytes or glycolytic lymphoma cell-lines but were restored by transduction with the nuclear receptor PPARδ, which mediates metabolic activity in CLL cells. Breakdown of LDLs in lysosomes was required for the amplification effect, which correlated with down-regulation of HMGCR expression and long lymphocyte doubling times (LDTs) of 53.6 ± 10.4 months. Cholesterol content of circulating CLL cells correlated directly with blood LDL levels in a subgroup of patients. These observations suggest LDLs may enhance proliferative responses of CLL cells to inflammatory signals. Prospective clinical trials are needed to confirm the therapeutic potential of lowering LDL concentrations in CLL, particularly in patients with indolent disease in the “watch-and-wait” phase of management. Slow-growing CLL cells use lysosomal lipase to break low density lipoproteins (LDLs) into free fatty acids and cholesterol. LdL degradation products increase survival of proliferating CLL cells. LDLs decrease oxidative stress and increase plasma membrane cholesterol. LDLs amplify signaling responses to cytokines but not antigens in proliferating CLL cells. Rapidly growing CLL cells, acute leukemia cells, and normal lymphocytes do not exhibit this dependence on LDLs.
Collapse
Affiliation(s)
- Lindsay McCaw
- Biology Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Yonghong Shi
- Biology Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Guizhi Wang
- Biology Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - You-Jun Li
- Biology Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - David E Spaner
- Biology Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, ON M5G 2C4, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada; Sunnybrook Odette Cancer Center, Toronto, ON M4N 3M5, Canada.
| |
Collapse
|
93
|
Todoric J, Antonucci L, Karin M. Targeting Inflammation in Cancer Prevention and Therapy. Cancer Prev Res (Phila) 2016; 9:895-905. [PMID: 27913448 DOI: 10.1158/1940-6207.capr-16-0209] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/03/2016] [Indexed: 12/14/2022]
Abstract
Inflammation is associated with the development and malignant progression of most cancers. As most of the cell types involved in cancer-associated inflammation are genetically stable and thus are not subjected to rapid emergence of drug resistance, the targeting of inflammation represents an attractive strategy both for cancer prevention and for cancer therapy. Tumor-extrinsic inflammation is caused by many factors, including bacterial and viral infections, autoimmune diseases, obesity, tobacco smoking, asbestos exposure, and excessive alcohol consumption, all of which increase cancer risk and stimulate malignant progression. In contrast, cancer-intrinsic or cancer-elicited inflammation can be triggered by cancer-initiating mutations and can contribute to malignant progression through the recruitment and activation of inflammatory cells. Both extrinsic and intrinsic inflammation can result in immunosuppression, thereby providing a preferred background for tumor development. In clinical trials, lifestyle modifications including healthy diet, exercise, alcohol, and smoking cessation have proven effective in ameliorating inflammation and reducing the risk of cancer-related deaths. In addition, consumption of certain anti-inflammatory drugs, including aspirin, can significantly reduce cancer risk, suggesting that common nonsteroidal anti-inflammatory drugs (NSAID) and more specific COX2 inhibitors can be used in cancer prevention. In addition to being examined for their preventative potential, both NSAIDs and more potent anti-inflammatory antibody-based drugs need to be tested for their ability to augment the efficacy of more conventional therapeutic approaches on the basis of tumor resection, radiation, and cytotoxic chemicals. Cancer Prev Res; 9(12); 895-905. ©2016 AACR.
Collapse
Affiliation(s)
- Jelena Todoric
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California.,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, California. .,Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
94
|
Borgquist S, Tamimi RM, Chen WY, Garber JE, Eliassen AH, Ahern TP. Statin Use and Breast Cancer Risk in the Nurses' Health Study. Cancer Epidemiol Biomarkers Prev 2016; 25:201-6. [PMID: 26762806 DOI: 10.1158/1055-9965.epi-15-0654] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Preclinical studies support an anticancer effect of statin drugs, yet epidemiologic evidence remains inconsistent regarding their role in breast cancer primary prevention. Here, we report an updated analysis of the association between statin use and breast cancer incidence in the Nurses' Health Study (NHS) cohort. Postmenopausal NHS participants without a cancer history were followed from 2000 until 2012 (n = 79,518). Data on statin use were retrieved from biennial questionnaires. We fit Cox regression models to estimate associations between longitudinal statin use and breast cancer incidence. Over 823,086 person-years of follow-up, 3,055 cases of invasive breast cancer occurred. Compared with never users, both former and current statin users had similar rates of invasive breast cancer incidence [former users: HRadj, 0.96; 95% confidence interval (CI), 0.82-1.1; current users: HRadj, 1.1; 95% CI, 0.92-1.3]. Associations did not differ by estrogen receptor (ER) status or histology (ductal vs. lobular carcinoma). Statin use was not associated with risk of invasive breast cancer, irrespective of histologic subtype and ER status. Statin drugs do not appear to modify processes involved in breast cancer initiation.
Collapse
Affiliation(s)
- Signe Borgquist
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden.
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Wendy Y Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - A Heather Eliassen
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Thomas P Ahern
- Departments of Surgery and Biochemistry, University of Vermont College of Medicine, Burlington, Vermont
| |
Collapse
|
95
|
Hanly EK, Bednarczyk RB, Tuli NY, Moscatello AL, Halicka HD, Li J, Geliebter J, Darzynkiewicz Z, Tiwari RK. mTOR inhibitors sensitize thyroid cancer cells to cytotoxic effect of vemurafenib. Oncotarget 2016; 6:39702-13. [PMID: 26284586 PMCID: PMC4741856 DOI: 10.18632/oncotarget.4052] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/31/2015] [Indexed: 12/23/2022] Open
Abstract
Treatment options for advanced metastatic thyroid cancer patients are limited. Vemurafenib, a BRAFV600E inhibitor, has shown promise in clinical trials although cellular resistance occurs. Combination therapy that includes BRAFV600E inhibition and avoids resistance is a clinical need. We used an in vitro model to examine combination treatment with vemurafenib and mammalian target of rapamycin (mTOR) inhibitors, metformin and rapamycin. Cellular viability and apoptosis were analyzed in thyroid cell lines by trypan blue exclusion and TUNEL assays. Combination of vemurafenib and metformin decreased cell viability and increased apoptosis in both BCPAP papillary thyroid cancer cells and 8505c anaplastic thyroid cancer cells. This combination was also found to be active in vemurafenib-resistant BCPAP cells. Changes in expression of signaling molecules such as decreased mTOR expression in BCPAP and enhanced inhibition of phospho-MAPK in resistant BCPAP and 8505c were observed. The second combination of vemurafenib and rapamycin amplified cell death in BCPAP cells. We conclude that combination of BRAFV600E and mTOR inhibition forms the basis of a treatment regimen that should be further investigated in in vivo model systems. Metformin or rapamycin adjuvant treatment may provide clinical benefits with minimal side effects to BRAFV600E-positive advanced thyroid cancer patients treated with vemurafenib.
Collapse
Affiliation(s)
- Elyse K Hanly
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Robert B Bednarczyk
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Neha Y Tuli
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Augustine L Moscatello
- Department of Otolaryngology/Head and Neck Surgery, New York Medical College, Valhalla, NY, USA
| | - H Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Jiangwei Li
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Jan Geliebter
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | | | - Raj K Tiwari
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
96
|
Chen CC, Hsu YP, Liu JC, Kao PF, Sung LC, Lin CF, Hao WR, Liu SH, Wu SY. Statins Dose-Dependently Exert Significant Chemopreventive Effects Against Various Cancers in Chronic Obstructive Pulmonary Disease Patients: A Population-Based Cohort Study. J Cancer 2016; 7:1892-1900. [PMID: 27698930 PMCID: PMC5039374 DOI: 10.7150/jca.15779] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/29/2016] [Indexed: 01/14/2023] Open
Abstract
PURPOSE: Chronic obstructive pulmonary disease (COPD) is associated with an increased cancer risk. We evaluated the chemopreventive effect of statins against all cancers in COPD patients and identified the statin with the strongest chemopreventive effect. PATIENTS AND METHODS: All patients diagnosed with COPD at health care facilities in Taiwan (n = 116,017) from January 1, 2001, to December 31, 2012, were recruited. Each patient was followed to assess the following protective and risk factors for all cancers: age; sex; comorbidities (diabetes, hypertension, dyslipidemia) and the Charlson comorbidity index [CCI]); urbanization level; monthly income; and nonstatin drug use. The index date of statins use was the date of COPD confirmation. Propensity scores (PSs) were derived using a logistic regression model to estimate the effect of statins by considering the covariates predicting intervention (statins) receipt. To examine the dose-response relationship, we categorized statin use into four groups in each cohort (<28 [statin nonusers], 28-90, 91-365, and >365 cumulative defined daily dose). RESULTS: After PS adjustment for age, sex, CCI, diabetes, hypertension, dyslipidemia, urbanization level, and monthly income, we analyzed the all-cancer risk. The adjusted hazard ratios (aHRs) for the all-cancer risk were lower among statin users than among statin nonusers (aHR = 0.46, 95% confidence interval: 0.43 to 0.50). The aHRs for the all-cancer risk were lower among patients using rosuvastatin, simvastatin, atorvastatin, pravastatin, and fluvastatin than among statin nonusers (aHRs = 0.42, 0.55, 0.59, 0.66, and 0.78, respectively). Sensitivity analysis indicated that statins dose-dependently reduced the all-cancer risk. CONCLUSION: Statins dose-dependently exert a significant chemopreventive effect against various cancers in COPD patients. In particular, rosuvastatin has the strongest chemopreventive effect.
Collapse
Affiliation(s)
- Chun-Chao Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Ping Hsu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Ju-Chi Liu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pai-Feng Kao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Chin Sung
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Feng Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wen-Rui Hao
- Division of Cardiovascular Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Szu-Yuan Wu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Radiation Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biotechnology, Hungkuang University, Taichung, Taiwan
| |
Collapse
|
97
|
Mocellin S, Goodwin A, Pasquali S. Risk-reducing medication for primary breast cancer: a network meta-analysis. Hippokratia 2016. [DOI: 10.1002/14651858.cd012191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Simone Mocellin
- University of Padova; Dept. Surgery Oncology and Gastroenterology; Via Giustiniani 2 Padova Veneto Italy 35128
- IOV-IRCCS; Istituto Oncologico Veneto; Padova Italy 35100
| | - Annabel Goodwin
- The University of Sydney, Concord Repatriation General Hospital; Concord Clinical School; Concord NSW Australia 2137
- Concord Repatriation General Hospital; Medical Oncology Department; Concord Australia
- Sydney Local Health District and South Western Sydney Local Health District; Cancer Genetics Department; Sydney Australia
| | - Sandro Pasquali
- Veneto Institute of Oncology - IRCCS; Surgical Oncology Unit; Via Gattamelata 64 Padova Italy 35128
| |
Collapse
|
98
|
Göbel A, Thiele S, Browne AJ, Rauner M, Zinna VM, Hofbauer LC, Rachner TD. Combined inhibition of the mevalonate pathway with statins and zoledronic acid potentiates their anti-tumor effects in human breast cancer cells. Cancer Lett 2016; 375:162-171. [DOI: 10.1016/j.canlet.2016.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 12/22/2022]
|
99
|
Lee HS, Lee SH, Lee HJ, Chung MJ, Park JY, Park SW, Song SY, Bang S. Statin Use and Its Impact on Survival in Pancreatic Cancer Patients. Medicine (Baltimore) 2016; 95:e3607. [PMID: 27175667 PMCID: PMC4902509 DOI: 10.1097/md.0000000000003607] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Statins are cholesterol-lowering medications that are associated with a number of signaling pathways involved in carcinogenesis. Recent observational studies raised the possibility that the use of statins may reduce overall mortality in various types of cancer. We investigated whether statins used after pancreatic cancer diagnosis are associated with longer survival in pancreatic cancer patients.We retrospectively analyzed data from 1761 patients newly diagnosed with pancreatic adenocarcinoma between January 1, 2006, and December 31, 2014. We used the time-dependent Cox proportional hazards regression model to estimate mortality among pancreatic cancer patients according to statin use.Among the 1761 pancreatic cancer patients, 118 patients had used statins. During the study period, 1176 patients (66.7%) died. After adjusting for age, sex, location of cancer, cancer stage, diabetes mellitus, hypertension, dyslipidemia, smoking, alcohol use, body mass index, and CA 19-9, statin use was associated with a lower risk of cancer death (hazard ratio [HR], 0.780; 95% confidence interval [CI], 0.617-0.986), especially among simvastatin users (HR, 0.554; 95% CI, 0.312-0.982) and atorvastatin users (HR, 0.636; 95% CI, 0.437-0.927). Subgroup analysis showed that overall survival was statistically significantly longer in patients with nonmetastatic pancreatic cancer (log-rank P = 0.024).We found that the use of simvastatin and atorvastatin after cancer diagnosis is associated with longer survival in patients with nonmetastatic pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Hee Seung Lee
- From the Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Seo YS, Kim YJ, Kim MS, Suh KS, Kim SB, Han CJ, Kim YJ, Jang WI, Kang SH, Tchoe HJ, Park CM, Jo AJ, Kim HJ, Choi JA, Choi HJ, Polak MN, Ko MJ. Association of Metformin Use With Cancer-Specific Mortality in Hepatocellular Carcinoma After Curative Resection: A Nationwide Population-Based Study. Medicine (Baltimore) 2016; 95:e3527. [PMID: 27124061 PMCID: PMC4998724 DOI: 10.1097/md.0000000000003527] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many preclinical reports and retrospective population studies have shown an anticancer effect of metformin in patients with several types of cancer and comorbid type 2 diabetes mellitus (T2DM). In this work, the anticancer effect of metformin was assessed in hepatocellular carcinoma (HCC) patients with T2DM who underwent curative resection.A population-based retrospective cohort design was used. Data were obtained from the National Health Insurance Service and Korea Center Cancer Registry in the Republic of Korea, identifying 5494 patients with newly diagnosed HCC who underwent curative resection between 2005 and 2011. Crude and adjusted hazard ratios (HRs) were calculated using Cox proportional hazard models to estimate effects. In the sensitivity analysis, we excluded patients who started metformin or other oral hypoglycemic agents (OHAs) after HCC diagnosis to control for immortal time bias.From the patient cohort, 751 diabetic patients who were prescribed an OHA were analyzed for HCC-specific mortality and retreatment upon recurrence, comparing 533 patients treated with metformin to 218 patients treated without metformin. In the fully adjusted analyses, metformin users showed a significantly lower risk of HCC-specific mortality (HR 0.38, 95% confidence interval [CI] 0.30-0.49) and retreatment events (HR 0.41, 95% CI 0.33-0.52) compared with metformin nonusers. Risks for HCC-specific mortality were consistently lower among metformin-using groups, excluding patients who started metformin or OHAs after diagnosis.In this large population-based cohort of patients with comorbid HCC and T2DM, treated with curative hepatic resection, metformin use was associated with improvement of HCC-specific mortality and reduced occurrence of retreatment events.
Collapse
Affiliation(s)
- Young-Seok Seo
- From the Department of Radiation Oncology (Y-SS, M-SK, WIJ), Korea Institute of Radiological and Medical Sciences; Division for Healthcare Technology Assessment Research (Y-JK, M-SK, SHK, HJT, CMP, AJJ, HJK, JAC, MJK), National Evidence-based Healthcare Collaborating Agency; Department of General Surgery (K-SS), Seoul National University Hospital; Department of General Surgery (SBK); Department of Internal Medicine (CJH, YJK), Korea Institute of Radiological and Medical Sciences; Department of Anatomy (HJC), Seoul National University College of Medicine, Seoul, Korea; and Department of Oncology (MNP), McGill University, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|