1
|
Che Y, Yuan J, Tang D, Guo J. Lipid traits and lipid-lowering drug target genes and risk of melanoma: a mendelian randomization study. Arch Dermatol Res 2024; 316:301. [PMID: 38819656 DOI: 10.1007/s00403-024-03100-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 02/19/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024]
Abstract
Our study aimed to investigate the role of lipids in melanoma risk and the effect of lipid-lowering drug targets on melanoma. Using Mendelian Randomization analysis, we examined the genetic agents of nine lipid-lowering drugs and their association with melanoma risk. We found that genetically proxied inhibition of HMGCR, ABCG5/ABCG8, and ANGPTL3 was associated with a reduced risk of melanoma. On the other hand, inhibition of LPL and Apo-B100 was significantly associated with an increased risk of melanoma. Sensitivity analyses did not reveal any statistical evidence of bias from pleiotropy or genetic confounding. We did not find a robust association between lipid traits NPC1L1, PCSK9, APOC3 inhibition, and melanoma risk. These findings were validated using two independent lipid datasets. Our analysis also revealed that HMGCR, ANGPTL3, and ABCG5/ABCG8 inhibitors reduced melanoma risk independent of their effects on lipids. This suggests that these targets may have potential for melanoma prevention or treatment. In conclusion, our study provides evidence for a causal role of lipids in melanoma risk and highlights specific lipid-lowering drug targets that may be effective in reducing the risk of melanoma. These findings contribute to the understanding of the underlying mechanisms of melanoma development and provide potential avenues for further research and therapeutic interventions.
Collapse
Affiliation(s)
- Yuhui Che
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinyao Yuan
- West China Second Hospital of Sichuan University, Chengdu, China
| | - Dadong Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
2
|
L’Orphelin JM, Dollalille C, Akroun J, Alexandre J, Dompmartin A. Cardiovascular Immunotoxicity Associated with Immune Checkpoint Inhibitors in Metastatic Melanoma. Cancers (Basel) 2023; 15:cancers15072170. [PMID: 37046831 PMCID: PMC10093552 DOI: 10.3390/cancers15072170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
Background: Checkpoint inhibitors, such as PD-1 inhibitors (nivolumab, pembrolizumab) and anti-CTLA-4 (CD152) (ipilimumab), are widely used in metastatic melanoma, and most immune-related adverse events are known. Several cardiovascular AEs (CVAEs) associated with immune checkpoint inhibitor exposure have been reported in post-marketing surveillance studies and represent major issues for patients with melanoma during and after cancer treatment. Data on CVAES induced by immune checkpoint inhibitors in melanoma, especially incidence and risk factors, are lacking. Methods: A systematic review of the literature up to 31 August 2020 was performed in Medline, the Cochrane Central Register of Controlled Trials (CENTRAL), and the ClinicalTrials.gov register according to prespecified selection criteria from inception to 7 April 2020. Statistics were performed on 3289 patients from five randomized clinical trials on melanoma. Results: Patients with melanoma treated with immune checkpoint inhibitors had a significant risk of presenting dyslipidemia (Peto OR: 4.74, 95% CI: 2.16–10.41, p < 0.01, I2 = 0%, p = 0.94). The Peto OR was numerically significant for pericarditis, myocarditis, heart failure, myocardial infarction, cerebral ischemia, high pulmonary pressure, blood high pressure, arrhythmias, endocarditis, and conduction disturbances, but the confidence interval was not significant. The risk of CVAEs was not statistically different between melanoma treated with immune checkpoint inhibitors and other tumors treated with immune checkpoint inhibitors (range of p-value from 0.13 to 0.95). No interaction between follow-up length and CVAE reporting was found. Conclusions: Our study underlines that checkpoint inhibitors used for melanoma increase CVAEs, especially dyslipidemia, which could pave the way to chronic inflammatory processes, atherosclerosis, and, finally, ischemic cardiopathy. These cardiovascular adverse events could be acute or delayed, justifying the monitoring of lipidic biology and a baseline cardiology consultation.
Collapse
Affiliation(s)
| | - Charles Dollalille
- Department of Cardiology, Caen-Normandie University Hospital, 14033 Caen CEDEX, France
| | - Julia Akroun
- Department of Dermatology, Caen-Normandie University Hospital, 14033 Caen CEDEX, France
| | - Joachim Alexandre
- Department of Cardiology, Caen-Normandie University Hospital, 14033 Caen CEDEX, France
| | - Anne Dompmartin
- Department of Dermatology, Caen-Normandie University Hospital, 14033 Caen CEDEX, France
| |
Collapse
|
3
|
Pflugfelder A, Yong XLH, Jagirdar K, Eigentler TK, Soyer HP, Sturm RA, Flatz L, Duffy DL. Genome-Wide Association Study Suggests the Variant rs7551288*A within the DHCR24 Gene Is Associated with Poor Overall Survival in Melanoma Patients. Cancers (Basel) 2022; 14:cancers14102410. [PMID: 35626014 PMCID: PMC9139953 DOI: 10.3390/cancers14102410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary The aim of this work was to investigate prognostic genetic factors in melanoma patients. Phenotypic and disease data as well as biomaterial were collected after informed consent from patients followed up in a Skin Cancer Center of a University clinic. Genome-wide analysis (GWAS) was performed with survival data of 556 melanoma patients and genetic data including more than 300,000 common polymorphisms. The SNP rs7551288 reached suggestive genome-wide significance (p = 2 × 10−6). This intronic variant of the DHCR24 gene is involved in the cholesterol synthesis pathway. Further analyses and a literature review suggest an important role of this locus for the clinical course of disease in melanoma patients. Abstract Melanoma incidence rates are high among individuals with fair skin and multiple naevi. Established prognostic factors are tumour specific, and less is known about prognostic host factors. A total of 556 stage I to stage IV melanoma patients from Germany with phenotypic and disease-specific data were analysed; 64 of these patients died of melanoma after a median follow-up time of 8 years. Germline DNA was assessed by the HumanCoreExome BeadChip and data of 356,384 common polymorphisms distributed over all 23 chromosomes were used for a genome-wide analysis. A suggestive genome-wide significant association of the intronic allele rs7551288*A with diminished melanoma-specific survival was detected (p = 2 × 10−6). The frequency of rs7551288*A was 0.43 and was not associated with melanoma risk, hair and eye colour, tanning and total naevus count. Cox regression multivariate analyses revealed a 5.31-fold increased risk of melanoma-specific death for patients with the rs7551288 A/A genotype, independent of tumour thickness, ulceration and stage of disease at diagnoses. The variant rs7551288 belongs to the DHCR24 gene, which encodes Seladin-1, an enzyme involved in the biosynthesis of cholesterol. Further investigations are needed to confirm this genetic variant as a novel prognostic biomarker and to explore whether specific treatment strategies for melanoma patients might be derived from it.
Collapse
Affiliation(s)
- Annette Pflugfelder
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Center of Dermatooncology, Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany;
- Correspondence:
| | - Xuan Ling Hilary Yong
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kasturee Jagirdar
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Biochemistry and Molecular Biology Department, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Thomas K. Eigentler
- Department of Dermatology, Venereology and Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10177 Berlin, Germany;
| | - H. Peter Soyer
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Richard A. Sturm
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
| | - Lukas Flatz
- Center of Dermatooncology, Department of Dermatology, University of Tübingen, 72076 Tübingen, Germany;
| | - David L. Duffy
- The University of Queensland Diamantina Institute, The University of Queensland, Dermatology Research Centre, Brisbane, QLD 4102, Australia; (X.L.H.Y.); (K.J.); (H.P.S.); (R.A.S.); (D.L.D.)
- Genetic Epidemiology, QIMR Berghofer Institute of Medical Research, Herston, QLD 4006, Australia
| |
Collapse
|
4
|
Beyond Lipid-Lowering: Effects of Statins on Cardiovascular and Cerebrovascular Diseases and Cancer. Pharmaceuticals (Basel) 2022; 15:ph15020151. [PMID: 35215263 PMCID: PMC8877351 DOI: 10.3390/ph15020151] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, also known as statins, are administered as first-line therapy for hypercholesterolemia, both as primary and secondary prevention. Besides the lipid-lowering effect, statins have been suggested to inhibit the development of cardiovascular disease through anti-inflammatory, antioxidant, vascular endothelial function-improving, plaque-stabilizing, and platelet aggregation-inhibiting effects. The preventive effect of statins on atherothrombotic stroke has been well established, but statins can influence other cerebrovascular diseases. This suggests that statins have many neuroprotective effects in addition to lowering cholesterol. Furthermore, research suggests that statins cause pro-apoptotic, growth-inhibitory, and pro-differentiation effects in various malignancies. Preclinical and clinical evidence suggests that statins inhibit tumor growth and induce apoptosis in specific cancer cell types. The pleiotropic effects of statins on cardiovascular and cerebrovascular diseases have been well established; however, the effects of statins on cancer patients have not been fully elucidated and are still controversial. This review discusses the recent evidence on the effects of statins on cardiovascular and cerebrovascular diseases and cancer. Additionally, this study describes the pharmacological action of statins, focusing on the aspect of ‘beyond lipid-lowering’.
Collapse
|
5
|
Photosensitizing Medications and Skin Cancer: A Comprehensive Review. Cancers (Basel) 2021; 13:cancers13102344. [PMID: 34066301 PMCID: PMC8152064 DOI: 10.3390/cancers13102344] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
(1) The incidence of skin cancer is increasing in the United States (US) despite scientific advances in our understanding of skin cancer risk factors and treatments. In vitro and in vivo studies have provided evidence that suggests that certain photosensitizing medications (PSMs) increase skin cancer risk. This review summarizes current epidemiological evidence on the association between common PSMs and skin cancer. (2) A comprehensive literature search was conducted to identify meta-analyses, observational studies and clinical trials that report on skin cancer events in PSM users. The associated risks of keratinocyte carcinoma (squamous cell carcinoma and basal cell carcinoma) and melanoma are summarized, for each PSM. (3) There are extensive reports on antihypertensives and statins relative to other PSMs, with positive and null findings, respectively. Fewer studies have explored amiodarone, metformin, antimicrobials and vemurafenib. No studies report on the individual skin cancer risks in glyburide, naproxen, piroxicam, chlorpromazine, thioridazine and nalidixic acid users. (4) The research gaps in understanding the relationship between PSMs and skin cancer outlined in this review should be prioritized because the US population is aging. Thus the number of patients prescribed PSMs is likely to continue to rise.
Collapse
|
6
|
Yu WY, Hill ST, Chan ER, Pink JJ, Cooper K, Leachman S, Lund AW, Kulkarni R, Bordeaux JS. Computational Drug Repositioning Identifies Statins as Modifiers of Prognostic Genetic Expression Signatures and Metastatic Behavior in Melanoma. J Invest Dermatol 2021; 141:1802-1809. [PMID: 33417917 DOI: 10.1016/j.jid.2020.12.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/02/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022]
Abstract
Despite advances in melanoma treatment, more than 70% of patients with distant metastasis die within 5 years. Proactive treatment of early melanoma to prevent metastasis could save lives and reduce overall healthcare costs. Currently, there are no treatments specifically designed to prevent early melanoma from progressing to metastasis. We used the Connectivity Map to conduct an in silico drug screen and identified 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins) as a drug class that might prevent melanoma metastasis. To confirm the in vitro effect of statins, RNA sequencing was completed on A375 cells after treatment with fluvastatin to describe changes in the melanoma transcriptome. Statins induced differential expression in genes associated with metastasis and are used in commercially available prognostic tests for melanoma metastasis. Finally, we completed a chart review of 475 patients with melanoma. Patients taking statins were less likely to have metastasis at the time of melanoma diagnosis in both univariate and multivariate analyses (24.7% taking statins vs. 37.6% not taking statins, absolute risk reduction = 12.9%, P = 0.038). These findings suggest that statins might be useful as a treatment to prevent melanoma metastasis. Prospective trials are required to verify our findings and to determine the mechanism of metastasis prevention.
Collapse
Affiliation(s)
- Wesley Y Yu
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA.
| | - Sheena T Hill
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - E Ricky Chan
- Institute for Computational Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - John J Pink
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kevin Cooper
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Sancy Leachman
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA; Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Rajan Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Jeremy S Bordeaux
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Lipid metabolic Reprogramming: Role in Melanoma Progression and Therapeutic Perspectives. Cancers (Basel) 2020; 12:cancers12113147. [PMID: 33121001 PMCID: PMC7692067 DOI: 10.3390/cancers12113147] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Melanoma is a devastating skin cancer characterized by an impressive metabolic plasticity. Melanoma cells are able to adapt to the tumor microenvironment by using a variety of fuels that contribute to tumor growth and progression. In this review, the authors summarize the contribution of the lipid metabolic network in melanoma plasticity and aggressiveness, with a particular attention to specific lipid classes such as glycerophospholipids, sphingolipids, sterols and eicosanoids. They also highlight the role of adipose tissue in tumor progression as well as the potential antitumor role of drugs targeting critical steps of lipid metabolic pathways in the context of melanoma. Abstract Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid β-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.
Collapse
|
8
|
Jeong GH, Lee KH, Kim JY, Eisenhut M, Kronbichler A, van der Vliet HJ, Hong SH, Shin JI, Gamerith G. Effect of Statin on Cancer Incidence: An Umbrella Systematic Review and Meta-Analysis. J Clin Med 2019; 8:jcm8060819. [PMID: 31181789 PMCID: PMC6617015 DOI: 10.3390/jcm8060819] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
Statins are reported to reduce the risk of cancer, but the results of various published studies have been contradictory. We carried out an umbrella review to provide an overview and understand the strength of evidence, extent of potential biases, and validity of claimed associations between the use of statins and cancer incidence. We comprehensively re-analyzed the data of meta-analyses of randomized controlled trials (RCTs) and observational studies on associations between statin use and cancer incidence. We also assessed the strength of evidence of the re-analyzed outcomes, which were determined from the criteria including statistical significance of the p-value of random-effects, as well as fixed-effects meta-analyses, small study effects, between-study heterogeneity, and a 95% prediction interval. Using a conventional method to assess the significance of meta-analysis (p-value < 0.05), statins had a statistically significant effect on reducing cancer incidence in 10 of 18 types of cancer. When we graded the level of evidence, no cancer type showed convincing evidence, and four cancers (esophageal cancer, hematological cancer, leukemia, and liver cancer) showed suggestive evidence of a preventive effect. There was weak evidence of an association with six cancers, and no significance for the remaining eight cancers. None of the meta-analyses of RCTs on the association of statin and cancer incidence showed a statistical significance. Although there was a preventive effect of statin on cancer incidence in 10 of the 18 cancer types, the evidence supporting the use of statins to reduce cancer incidence was low. Therefore, the associations between statin use and cancer incidence should be carefully considered by clinicians.
Collapse
Affiliation(s)
- Gwang Hun Jeong
- College of Medicine, Gyeongsang National University, Jinju 52727, Korea.
| | - Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, Yonsei-ro 50, Seodaemun-gu, C.P.O. Box 8044, Seoul 03722, Korea.
- Division of Pediatric Nephrology, Severance Children's Hospital, Seoul 03722, Korea.
| | - Jong Yeob Kim
- Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Michael Eisenhut
- Luton & Dunstable University Hospital NHS Foundation Trust, Lewsey Road, Luton LU4 ODZ, UK.
| | - Andreas Kronbichler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, 6020 Innsbruck, Austria.
| | - Hans J van der Vliet
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, VU University, 1081 HV Amsterdam, The Netherlands.
| | - Sung Hwi Hong
- Yonsei University College of Medicine, Seoul 03722, Korea.
- Department of Global Health and Population, Harvard TH Chan School of Public Health, 67 Huntington Avenue, Boston, MA 02115, USA.
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Yonsei-ro 50, Seodaemun-gu, C.P.O. Box 8044, Seoul 03722, Korea.
- Division of Pediatric Nephrology, Severance Children's Hospital, Seoul 03722, Korea.
- Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, 03722, Korea.
| | - Gabriele Gamerith
- Department of Medical Oncology, Medical University Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
9
|
Jeter JM, Bowles TL, Curiel-Lewandrowski C, Swetter SM, Filipp FV, Abdel-Malek ZA, Geskin LJ, Brewer JD, Arbiser JL, Gershenwald JE, Chu EY, Kirkwood JM, Box NF, Funchain P, Fisher DE, Kendra KL, Marghoob AA, Chen SC, Ming ME, Albertini MR, Vetto JT, Margolin KA, Pagoto SL, Hay JL, Grossman D, Ellis DL, Kashani-Sabet M, Mangold AR, Markovic SN, Meyskens FL, Nelson KC, Powers JG, Robinson JK, Sahni D, Sekulic A, Sondak VK, Wei ML, Zager JS, Dellavalle RP, Thompson JA, Weinstock MA, Leachman SA, Cassidy PB. Chemoprevention agents for melanoma: A path forward into phase 3 clinical trials. Cancer 2019; 125:18-44. [PMID: 30281145 PMCID: PMC6860362 DOI: 10.1002/cncr.31719] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/10/2018] [Accepted: 07/12/2018] [Indexed: 12/12/2022]
Abstract
Recent progress in the treatment of advanced melanoma has led to unprecedented improvements in overall survival and, as these new melanoma treatments have been developed and deployed in the clinic, much has been learned about the natural history of the disease. Now is the time to apply that knowledge toward the design and clinical evaluation of new chemoprevention agents. Melanoma chemoprevention has the potential to reduce dramatically both the morbidity and the high costs associated with treating patients who have metastatic disease. In this work, scientific and clinical melanoma experts from the national Melanoma Prevention Working Group, composed of National Cancer Trials Network investigators, discuss research aimed at discovering and developing (or repurposing) drugs and natural products for the prevention of melanoma and propose an updated pipeline for translating the most promising agents into the clinic. The mechanism of action, preclinical data, epidemiological evidence, and results from available clinical trials are discussed for each class of compounds. Selected keratinocyte carcinoma chemoprevention studies also are considered, and a rationale for their inclusion is presented. These data are summarized in a table that lists the type and level of evidence available for each class of agents. Also included in the discussion is an assessment of additional research necessary and the likelihood that a given compound may be a suitable candidate for a phase 3 clinical trial within the next 5 years.
Collapse
Affiliation(s)
- Joanne M Jeter
- Department of Medicine, Divisions of Genetics and Oncology, The Ohio State University, Columbus, Ohio
| | - Tawnya L Bowles
- Department of Surgery, Intermountain Health Care, Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | | | - Susan M Swetter
- Department of Dermatology, Pigmented Lesion and Melanoma Program, Stanford University Medical Center Cancer Institute, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Fabian V Filipp
- Systems Biology and Cancer Metabolism, Program for Quantitative Systems Biology, University of California Merced, Merced, California
| | | | - Larisa J Geskin
- Department of Dermatology, Cutaneous Oncology Center, Columbia University Medical Center, New York, New York
| | - Jerry D Brewer
- Department of Dermatologic Surgery, Mayo Clinic Minnesota, Rochester, Minnesota
| | - Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia
- Division of Dermatology, Veterans Affairs Medical Center, Atlanta, Georgia
| | - Jeffrey E Gershenwald
- Departments of Surgical Oncology and Cancer Biology, Melanoma and Skin Cancer Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emily Y Chu
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - John M Kirkwood
- Melanoma and Skin Cancer Program, Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Neil F Box
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Dermatology Service, U.S. Department of Veterans Affairs, Eastern Colorado Health Care System, Denver, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Kari L Kendra
- Department of Internal Medicine, Medical Oncology Division, The Ohio State University, Columbus, Ohio
| | - Ashfaq A Marghoob
- Memorial Sloan Kettering Skin Cancer Center and Department of Dermatology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Suephy C Chen
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia
- Division of Dermatology, Veterans Affairs Medical Center, Atlanta, Georgia
| | - Michael E Ming
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark R Albertini
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, University of Wisconsin Carbone Cancer Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - John T Vetto
- Division of Surgical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Kim A Margolin
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Sherry L Pagoto
- Department of Allied Health Sciences, UConn Institute for Collaboration in Health, Interventions, and Policy, University of Connecticut, Storrs, Connecticut
| | - Jennifer L Hay
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Douglas Grossman
- Departments of Dermatology and Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Darrel L Ellis
- Department of Dermatology, Vanderbilt University Medical Center and Division of Dermatology, Vanderbilt Ingram Cancer Center, Nashville, Tennessee
- Department of Medicine, Tennessee Valley Healthcare System, Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | - Mohammed Kashani-Sabet
- Center for Melanoma Research and Treatment, California Pacific Medical Center, San Francisco, California
| | | | | | | | - Kelly C Nelson
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - June K Robinson
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Debjani Sahni
- Department of Dermatology, Boston Medical Center, Boston, Massachusetts
| | | | - Vernon K Sondak
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
- Departments of Oncologic Sciences and Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Maria L Wei
- Department of Dermatology, University of California, San Francisco, San Francisco, California
- Dermatology Service, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Jonathan S Zager
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
- Department of Sarcoma, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Robert P Dellavalle
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Dermatology Service, U.S. Department of Veterans Affairs, Eastern Colorado Health Care System, Denver, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - John A Thompson
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Martin A Weinstock
- Center for Dermatoepidemiology, Veterans Affairs Medical Center, Providence, Rhode Island
- Department of Dermatology, Brown University, Providence, Rhode Island
- Department of Epidemiology, Brown University, Providence, Rhode Island
- Department of Dermatology, Rhode Island Hospital, Providence, Rhode Island
| | - Sancy A Leachman
- Department of Dermatology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Pamela B Cassidy
- Department of Dermatology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
10
|
Vitamin D signaling and melanoma: role of vitamin D and its receptors in melanoma progression and management. J Transl Med 2017; 97:706-724. [PMID: 28218743 PMCID: PMC5446295 DOI: 10.1038/labinvest.2017.3] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/19/2022] Open
Abstract
Ultraviolet B (UVB), in addition to having carcinogenic activity, is required for the production of vitamin D3 (D3) in the skin which supplies >90% of the body's requirement. Vitamin D is activated through hydroxylation by 25-hydroxylases (CYP2R1 or CYP27A1) and 1α-hydroxylase (CYP27B1) to produce 1,25(OH)2D3, or through the action of CYP11A1 to produce mono-di- and trihydroxy-D3 products that can be further modified by CYP27B1, CYP27A1, and CYP24A1. The active forms of D3, in addition to regulating calcium metabolism, exert pleiotropic activities, which include anticarcinogenic and anti-melanoma effects in experimental models, with photoprotection against UVB-induced damage. These diverse effects are mediated through an interaction with the vitamin D receptor (VDR) and/or as most recently demonstrated through action on retinoic acid orphan receptors (ROR)α and RORγ. With respect to melanoma, low levels of 25(OH)D are associated with thicker tumors and reduced patient survival. Furthermore, single-nucleotide polymorphisms of VDR and the vitamin D-binding protein (VDP) genes affect melanomagenesis or disease outcome. Clinicopathological analyses have shown positive correlation between low or undetectable expression of VDR and/or CYP27B1 in melanoma with tumor progression and shorter overall (OS) and disease-free survival (DFS) times. Paradoxically, this correlation was reversed for CYP24A1 (inactivating 24-hydroxylase), indicating that this enzyme, while inactivating 1,25(OH)2D3, can activate other forms of D3 that are products of the non-canonical pathway initiated by CYP11A1. An inverse correlation has been found between the levels of RORα and RORγ expression and melanoma progression and disease outcome. Therefore, we propose that defects in vitamin D signaling including D3 activation/inactivation, and the expression and activity of the corresponding receptors, affect melanoma progression and the outcome of the disease. The existence of multiple bioactive forms of D3 and alternative receptors affecting the behavior of melanoma should be taken into consideration when applying vitamin D management for melanoma therapy.
Collapse
|
11
|
Undela K, Shah CS, Mothe RK. Statin use and risk of cancer: An overview of meta-analyses. World J Meta-Anal 2017; 5:41-53. [DOI: 10.13105/wjma.v5.i2.41] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/06/2016] [Accepted: 02/20/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To conduct an overview of meta-analyses to critically appraise the evidence and present a comprehensive evaluation of the association between statin use and risk of site specific cancers.
METHODS MEDLINE, EMBASE, the Cochrane Database of Systematic Reviews and Web of Science databases were searched from inception until 31st May 2016. The electronic database search was supplemented by a hand search in PROSPERO and relevant journals which are not indexed in above databases. Meta-analyses that examined the association between statin use and risk of site specific cancers were included. Two reviewers independently screened the literature, abstracted data, and assessed study quality using the Assessment of Multiple Systematic Reviews (AMSTAR) tool.
RESULTS Overall, 38 meta-analyses covered 13 site specific cancers were included. More than half (68%) of the meta-analyses were moderate in quality with an AMSTAR score 4-7 out of a possible 11. Based on current evidence from meta-analyses, use of statin decreases the risk of certain cancers, such as colorectal (8%-12%), gastric (27%-44%), hematological (19%), liver (37%-42%), oesophageal (14%-28%), ovarian (21%) and prostate cancer (7%). On the other side, evidence from meta-analyses also suggests that there is no association between statin use and risk of bladder, breast, endometrial, kidney, lung, pancreatic and skin cancers.
CONCLUSION This overview of meta-analyses with variable quality has been shown that the statins may have a potential role in cancer chemoprevention and reduce the risk of some site specific cancers, but not all.
Collapse
|
12
|
Mounessa J, Buntinx-Krieg T, Qin R, Dunnick CA, Dellavalle RP. Primary and Secondary Chemoprevention of Malignant Melanoma. Am J Clin Dermatol 2016; 17:625-634. [PMID: 27665299 DOI: 10.1007/s40257-016-0221-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The incidence of malignant melanoma (MM) continues to rise in the United States. While sun protection and full body skin examinations remain the mainstay of preventative care, chemoprevention of the deadly disease has become an increasingly popular field of study. In this focused review, we discuss current findings and analyze the risks and benefits of various agents investigated for the primary and secondary chemoprevention of MM. Such agents include topical retinoids, vitamins, and supplements, Polypodium leucotomas extracts, non-steroidal anti-inflammatory agents (NSAIDs), statins, sunscreens, and field therapy with topical imiquimod for primary and secondary chemoprevention. We further identify a need for expanded high quality human research on the topic.
Collapse
Affiliation(s)
- Jessica Mounessa
- Stony Brook University School of Medicine, 101 Nicolls Road, Stony Brook, NY, 11794, USA
- Department of Dermatology, University of Colorado Hospital, 1665 Aurora Ct, Aurora, CO, 80045, USA
| | - Talayesa Buntinx-Krieg
- University of Central Florida College of Medicine, 6850 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Rosie Qin
- Department of Medicine, University of San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Cory A Dunnick
- Department of Dermatology, University of Colorado Hospital, 1665 Aurora Ct, Aurora, CO, 80045, USA
- Department of Dermatology, Denver VA Medical Center, 1055 Clermont St. #165, Denver, CO, 80220, USA
| | - Robert P Dellavalle
- Department of Dermatology, University of Colorado Hospital, 1665 Aurora Ct, Aurora, CO, 80045, USA.
- Department of Dermatology, Denver VA Medical Center, 1055 Clermont St. #165, Denver, CO, 80220, USA.
| |
Collapse
|
13
|
Belbasis L, Stefanaki I, Stratigos AJ, Evangelou E. Non-genetic risk factors for cutaneous melanoma and keratinocyte skin cancers: An umbrella review of meta-analyses. J Dermatol Sci 2016; 84:330-339. [PMID: 27663092 DOI: 10.1016/j.jdermsci.2016.09.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Skin cancers have a complex disease mechanism, involving both genetic and non-genetic risk factors. Numerous meta-analyses have been published claiming statistically significant associations between non-genetic risk factors and skin cancers without applying a thorough methodological assessment. OBJECTIVE The present study maps the literature on the non-genetic risk factors of skin cancers, assesses the presence of statistical biases and identifies the associations with robust evidence. METHODS We searched PubMed up to January 20, 2016 to identify systematic reviews and meta-analyses of observational studies that examined associations between non-genetic factors and skin cancers. For each meta-analysis, we estimated the summary effect size by random-effects and fixed-effects models, the 95% confidence interval and the 95% prediction interval. We also assessed the between-study heterogeneity (I2 metric), evidence for small-study effects and excess significance bias. RESULTS Forty-four eligible papers were identified and included a total of 85 associations. Twenty-one associations were significant at P<10-6. Fifty-two associations had large or very large heterogeneity. Evidence for small-study effects and excess significance bias was found in fifteen and thirteen associations, respectively. Overall, thirteen associations (actinic keratosis, serum vitamin D, sunburns, and hair color for basal cell carcinoma and density of freckles, eye color, hair color, history of melanoma, skin type, sunburns, premalignant skin lesions, common and atypical nevi for melanoma) presented high level of credibility. CONCLUSION The majority of meta-analyses on non-genetic risk factors for skin cancers suffered from large between-study heterogeneity and small-study effects or excess significance bias. The associations with convincing and highly suggestive evidence were mainly focused on skin photosensitivity and phenotypic characteristics.
Collapse
Affiliation(s)
- Lazaros Belbasis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Irene Stefanaki
- Department of Dermatology, Andreas Sygros Hospital, University of Athens Medical School, Athens, Greece
| | - Alexander J Stratigos
- Department of Dermatology, Andreas Sygros Hospital, University of Athens Medical School, Athens, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
14
|
Lanterna C, Musumeci A, Raccosta L, Corna G, Moresco M, Maggioni D, Fontana R, Doglioni C, Bordignon C, Traversari C, Russo V. The administration of drugs inhibiting cholesterol/oxysterol synthesis is safe and increases the efficacy of immunotherapeutic regimens in tumor-bearing mice. Cancer Immunol Immunother 2016; 65:1303-1315. [PMID: 27520505 PMCID: PMC11029546 DOI: 10.1007/s00262-016-1884-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 08/06/2016] [Indexed: 01/15/2023]
Abstract
Tumor-derived metabolites dampen tumor-infiltrating immune cells and antitumor immune responses. Among the various metabolites produced by tumors, we recently showed that cholesterol oxidized products, namely oxysterols, favor tumor growth through the inhibition of DC migration toward lymphoid organs and by promoting the recruitment of pro-tumor neutrophils within the tumor microenvironment. Here, we tested different drugs capable of blocking cholesterol/oxysterol formation. In particular, we tested efficacy and safety of different administration schedules, and of immunotherapy-based combination of a class of compounds, namely zaragozic acids, which inhibit cholesterol pathway downstream of mevalonate formation, thus leaving intact the formation of the isoprenoids, which are required for the maturation of proteins involved in the immune cell function. We show that zaragozic acids inhibit the in vivo growth of the RMA lymphoma and the Lewis lung carcinoma (LLC) without inducing side effects. Tumor growth inhibition requires an intact immune system, as immunodeficient tumor-bearing mice do not respond to zaragozic acid treatment. Of note, the effect of zaragozic acids is accompanied by a marked reduction in the LXR target genes Abcg1, Mertk, Scd1 and Srebp-1c in the tumor microenvironment. On the other hand, zoledronate, which blocks also isoprenoid formation, did not control the LLC tumor growth. Finally, we show that zaragozic acids potentiate the antitumor effects of active and adoptive immunotherapy, significantly prolonging the overall survival of tumor-bearing mice treated with the combo zaragozic acids and TAA-loaded DCs. This study identifies zaragozic acids as new antitumor compounds exploitable for the treatment of cancer patients.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/therapy
- Cholesterol/metabolism
- Combined Modality Therapy
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immunotherapy, Adoptive/methods
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Oxysterols/metabolism
- Tricarboxylic Acids/therapeutic use
- Tumor Escape
- Tumor Microenvironment
Collapse
Affiliation(s)
- Claudia Lanterna
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
- MolMed S.p.A., Milan, Italy
| | - Andrea Musumeci
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
- Institute for Immunology, Munich, Germany
| | - Laura Raccosta
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Gianfranca Corna
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Marta Moresco
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Daniela Maggioni
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Raffaella Fontana
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Claudio Doglioni
- Department of Pathology, IRCCS Scientific Institute San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Claudio Bordignon
- Università Vita-Salute San Raffaele, Milan, Italy
- MolMed S.p.A., Milan, Italy
| | | | - Vincenzo Russo
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, IRCCS Scientific Institute San Raffaele, Via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
15
|
Tocilizumab unmasks a stage-dependent interleukin-6 component in statin-induced apoptosis of metastatic melanoma cells. Melanoma Res 2016; 25:284-94. [PMID: 26020489 PMCID: PMC4492793 DOI: 10.1097/cmr.0000000000000172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The interleukin (IL)-6 inhibits the growth of early-stage melanoma cells, but not metastatic cells. Metastatic melanoma cells are susceptible to statin-induced apoptosis, but this is not clear for early-stage melanoma cells. This study aimed to investigate the IL-6 susceptibility of melanoma cells from different stages in the presence of simvastatin to overcome loss of growth arrest. ELISA was used to detect secreted IL-6 in human melanoma cells. The effects of IL-6 were measured by western blots for STAT3 and Bcl-2 family proteins. Apoptosis and proliferation were measured by caspase 3 activity, Annexin V staining, cell cycle analysis, and a wound-healing assay. Human metastatic melanoma cells A375 and 518A2 secrete high amounts of IL-6, in contrast to early-stage WM35 cells. Canonical IL-6 signaling is intact in these cells, documented by transient phosphorylation of STAT3. Although WM35 cells are highly resistant to simvastatin-induced apoptosis, coadministration with IL-6 enhanced the susceptibility to undergo apoptosis. This proapoptotic effect of IL-6 might be explained by a downregulation of Bcl-XL, observed only in WM35 cells. Furthermore, the IL-6 receptor blocking antibody tocilizumab was coadministered and unmasked an IL-6-sensitive proportion in the simvastatin-induced caspase 3 activity of metastatic melanoma cells. These results confirm that simvastatin facilitates apoptosis in combination with IL-6. Although endogenous IL-6 secretion is sufficient in metastatic melanoma cells, exogenously added IL-6 is needed for WM35 cells. This effect may explain the failure of simvastatin to reduce melanoma incidence in clinical trials and meta-analyses.
Collapse
|
16
|
Bonovas S. Statins: do they have a potential role in cancer prevention and modifying cancer-related outcomes? Drugs 2015; 74:1841-1848. [PMID: 25288321 DOI: 10.1007/s40265-014-0309-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins) are currently among the most commonly prescribed pharmaceutical agents worldwide. Apart from their well-established therapeutic value in cardiovascular disease, there is a long-standing debate on their potential association with cancer. To obtain and discuss the existing clinical evidence, an overview of meta-analysis articles addressing this issue was carried out. As of today, the accumulated evidence does not support the hypothesis that statins affect the risk of developing cancer, when they are taken at low doses for managing hypercholesterolaemia. However, current data cannot exclude an increased cancer risk in elderly patients associated with hydrophilic statin use, or decreases in the risks of certain cancers, such as gastric, oesophageal, liver, colorectal and advanced/aggressive prostate cancer. On the other hand, some recent observational studies have provided evidence that statins might be useful in modifying the prognosis of patients diagnosed with malignancy. Until a definitive benefit is demonstrated in randomized controlled trials, statins cannot be recommended either for cancer prevention or for modifying cancer-related outcomes. Further research is warranted to clarify the potential role(s) of statins in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Stefanos Bonovas
- Department of Pharmacology, School of Medicine, University of Athens, Athens, Greece. .,Laboratory of Drug Regulatory Policies, IRCCS Mario Negri Institute for Pharmacological Research, Via La Masa 19, 20156, Milan, Italy.
| |
Collapse
|
17
|
Wasinger C, Künzl M, Minichsdorfer C, Höller C, Zellner M, Hohenegger M. Autocrine secretion of 15d-PGJ2 mediates simvastatin-induced apoptotic burst in human metastatic melanoma cells. Br J Pharmacol 2015; 171:5708-27. [PMID: 25091578 DOI: 10.1111/bph.12871] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 07/14/2014] [Accepted: 07/29/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Despite new therapeutic approaches, metastatic melanomas still have a poor prognosis. Statins reduce low-density lipoprotein cholesterol and exert anti-inflammatory and anti-proliferative actions. We have recently shown that simvastatin triggers an apoptotic burst in human metastatic melanoma cells by the synthesis of an autocrine factor. EXPERIMENTAL APPROACH The current in vitro study was performed in human metastatic melanoma cell lines (A375, 518a2) and primary human melanocytes and melanoma cells. The secretome of simvastatin-stressed cells was analysed with two-dimensional difference gel electrophoresis and MS. The signalling pathways involved were analysed at the protein and mRNA level using pharmacological approaches and siRNA technology. KEY RESULTS Simvastatin was shown to activate a stress cascade, leading to the synthesis of 15-deoxy-12,14-PGJ2 (15d-PGJ2 ), in a p38- and COX-2-dependent manner. Significant concentrations of 15d-PGJ2 were reached in the medium of melanoma cells, which were sufficient to activate caspase 8 and the mitochondrial pathway of apoptosis. Inhibition of lipocalin-type PGD synthase, a key enzyme for 15d-PGJ2 synthesis, abolished the apoptotic effect of simvastatin. Moreover, 15d-PGJ2 was shown to bind to the fatty acid-binding protein 5 (FABP5), which was up-regulated and predominantly detected in the secretome of simvastatin-stressed cells. Knockdown of FABP5 abolished simvastatin-induced activation of PPAR-γ and amplified the apoptotic response. CONCLUSIONS AND IMPLICATIONS We characterized simvastatin-induced activation of the 15d-PGJ2 /FABP5 signalling cascades, which triggered an apoptotic burst in melanoma cells but did not affect primary human melanocytes. These data support the rationale for the pharmacological targeting of 15d-PGJ2 in metastatic melanoma.
Collapse
Affiliation(s)
- Christine Wasinger
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Melanoma is a common cancer in the Western world with an increasing incidence. Sun exposure is still considered to be the major risk factor for melanoma. The prognosis of patients with malignant (advanced-stage) melanoma differs widely between countries, but public campaigns advocating early detection have led to significant reductions in mortality rates. As well as sun exposure, distinct genetic alterations have been identified as associated with melanoma. For example, families with melanoma who have germline mutations in CDKN2A are well known, whereas the vast majority of sporadic melanomas have mutations in the mitogen-activated protein kinase cascade, which is the pathway with the highest oncogenic and therapeutic relevance for this disease. BRAF and NRAS mutations are typically found in cutaneous melanomas, whereas KIT mutations are predominantly observed in mucosal and acral melanomas. GNAQ and GNA11 mutations prevail in uveal melanomas. Additionally, the PI3K-AKT-PTEN pathway and the immune checkpoint pathways are important. The finding that programmed cell death protein 1 ligand 1 (PDL1) and PDL2 are expressed by melanoma cells, T cells, B cells and natural killer cells led to the recent development of programmed cell death protein 1 (PD1)-specific antibodies (for example, nivolumab and pembrolizumab). Alongside other new drugs - namely, BRAF inhibitors (vemurafenib and dabrafenib) and MEK inhibitors (trametinib and cobimetinib) - these agents are very promising and have been shown to significantly improve prognosis for patients with advanced-stage metastatic disease. Early signs are apparent that these new treatment modalities are also improving long-term clinical benefit and the quality of life of patients. This Primer summarizes the current understanding of melanoma, from mechanistic insights to clinical progress. For an illustrated summary of this Primer, visit: http://go.nature.com/vX2N9s.
Collapse
|
19
|
Sertznig P, Reichrath J. Peroxisome proliferator-activated receptors (PPARs) in dermatology. DERMATO-ENDOCRINOLOGY 2014. [DOI: 10.4161/derm.15025] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
20
|
Livingstone E, Hollestein LM, van Herk-Sukel MPP, van de Poll-Franse L, Joosse A, Schilling B, Nijsten T, Schadendorf D, de Vries E. Statin use and its effect on all-cause mortality of melanoma patients: a population-based Dutch cohort study. Cancer Med 2014; 3:1284-93. [PMID: 24935402 PMCID: PMC4302678 DOI: 10.1002/cam4.285] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/15/2014] [Accepted: 04/16/2014] [Indexed: 01/01/2023] Open
Abstract
Preclinical data showed anticancer effects of statins in melanoma, but meta-analyses could not demonstrate a reduced melanoma incidence in statin users. Rather than preventing occurrence, statins might reduce growth and metastatic spread of melanomas and ultimately improve survival. In this population-based study, we investigated the relationship between statin use and survival of melanoma patients. Patients ≥18 years who were diagnosed with cutaneous melanoma (Breslow thickness >1 mm) and registered in the Eindhoven Cancer Registry and in PHARMO Database Network between 1 January 1998 and 31 December 2010 were eligible. The hazard ratio (HR) of all-cause mortality was calculated by employing adjusted time-dependent and time-fixed Cox proportional hazard models. Disease-specific survival was estimated by means of 3-year relative survival rates (RSR). A control cohort of randomly selected patients using statins from PHARMO Database Network matched on age and gender was used to compare RSR of statin users to the general population. After melanoma diagnosis, 171 of 709 patients used statins. Use of statins showed a nonsignificantly decreased hazard of death (adjusted HR 0.76, 95% confidence interval [CI] 0.50–1.61). After stratification for gender, male but not female statin users showed a favorable outcome compared to nonusers (HR 0.57, 95% CI 0.32–0.99; HR 1.22, 95% CI 0.62–2.38, respectively). Three-year RSR for male statin users tended to be higher than for nonusers (91% vs. 80.5%, P = 0.06), no differences were observed in women (87.1% vs. 92.5%, P = 0.76). Statin use was not associated with an improved survival of melanoma patients. The trend for better survival of male in contrast to female statin users warrants further research.
Collapse
|
21
|
Salvo F, Bazin F, Kostrzewa A, Bandre C, Robinson P, Moore N, Bégaud B, Pariente A. Fibrates and Risk of Cancer in Tissues with High PPAR-α Concentration: A Nested Case–Control Study. Drug Saf 2014; 37:361-8. [DOI: 10.1007/s40264-014-0157-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
22
|
Linden KG, Leachman SA, Zager JS, Jakowatz JG, Viner JL, McLaren CE, Barr RJ, Carpenter PM, Chen WP, Elmets CA, Tangrea JA, Lim SJ, Cochran AJ, Meyskens FL. A randomized, double-blind, placebo-controlled phase II clinical trial of lovastatin for various endpoints of melanoma pathobiology. Cancer Prev Res (Phila) 2014; 7:496-504. [PMID: 24614012 DOI: 10.1158/1940-6207.capr-13-0189] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
On the basis of large cardiovascular clinical trials of lipid-lowering agents that showed a considerable decrease in the incidence of primary melanomas in the active agent arm, we have carried out a randomized, double-blind clinical trial examining the impact of lovastatin on various biomarkers of melanoma pathogenesis. Subjects with at least two clinically atypical nevi were randomized to receive oral lovastatin or placebo for a 6-month period. Clinical, histopathologic, and molecular biomarkers were evaluated for change in the two groups. Eighty subjects were randomized, evaluable, and included in the analyses. Lovastatin showed no benefit in comparison with placebo in the primary endpoint of decreasing the level of histopathologic atypia, nor in any of the secondary endpoints of decreasing clinical atypia, impact on nevus number, nor in showing significant changes in any of the molecular biomarkers. There were no significant differences in adverse event profiles for lovastatin compared with placebo. The lovastatin arm did show a significant and considerable decrease in total serum cholesterol and serum low-density lipoprotein (LDL) levels compared with placebo, an expected result. This finding bolsters confidence in subject compliance. Given the results of this trial, it is concluded that if lovastatin were to lower the incidence of melanoma, it would appear not to be doing so by reversing atypia of precursor atypical nevi over the 6-month time frame studied. Further research into the pathogenesis of melanoma and in other potential chemopreventive agents is needed.
Collapse
Affiliation(s)
- Kenneth G Linden
- Department of Dermatology and The Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, 101 The City Drive, Orange, CA 92868.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ardila JA, Oliveira GG, Medeiros RA, Fatibello-Filho O. Square-wave adsorptive stripping voltammetric determination of nanomolar levels of bezafibrate using a glassy carbon electrode modified with multi-walled carbon nanotubes within a dihexadecyl hydrogen phosphate film. Analyst 2014; 139:1762-8. [DOI: 10.1039/c3an02016a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A glassy carbon electrode modified with carbon nanotubes is applied for bezafibrate sensing.
Collapse
Affiliation(s)
| | | | | | - Orlando Fatibello-Filho
- Departamento de Química
- Universidade Federal de São Carlos
- São Carlos, Brazil
- Instituto Nacional de Ciência e Tecnologia de Bioanalítica (INCT de Bioanalítica)
- Campinas, Brazil
| |
Collapse
|
24
|
Statin use is not associated with reduced risk of skin cancer: a meta-analysis. Br J Cancer 2013; 110:802-7. [PMID: 24366301 PMCID: PMC3915126 DOI: 10.1038/bjc.2013.762] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/31/2013] [Accepted: 11/12/2013] [Indexed: 01/10/2023] Open
Abstract
Background: There is contradictory evidence about the association between statin and skin cancer. Methods: Literature search in PubMed and Web of Science was undertaken up to June 2013. Pooled relative risk (RR) estimates and 95% confidence intervals (CIs) were calculated. Result: A total of 21 articles with 29 studies were identified. No association was found between statin and skin cancer among neither melanoma (RR, 0.94; 95% CI, 0.85–1.04) nor non-melanoma skin cancer (RR, 1.03; 95% CI, 0.90–1.19). Conclusion: Our meta-analysis does not support a potential role of statin use in the prevention of skin cancer.
Collapse
|
25
|
Ioannidis JPA, Zhou Y, Chang CQ, Schully SD, Khoury MJ, Freedman AN. Potential increased risk of cancer from commonly used medications: an umbrella review of meta-analyses. Ann Oncol 2013; 25:16-23. [PMID: 24310915 DOI: 10.1093/annonc/mdt372] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Several commonly used medications have been associated with increased cancer risk in the literature. Here, we evaluated the strength and consistency of these claims in published meta-analyses. We carried out an umbrella review of 74 meta-analysis articles addressing the association of commonly used medications (antidiabetics, antihyperlipidemics, antihypertensives, antirheumatics, drugs for osteoporosis, and others) with cancer risk where at least one meta-analysis in the medication class included some data from randomized trials. Overall, 51 articles found no statistically significant differences, 13 found some decreased cancer risk, and 11 found some increased risk (one reported both increased and decreased risks). The 11 meta-analyses that found some increased risks reported 16 increased risk estimates, of which 5 pertained to overall cancer and 11 to site-specific cancer. Six of the 16 estimates were derived from randomized trials and 10 from observational data. Estimates of increased risk were strongly inversely correlated with the amount of evidence (number of cancer cases) (Spearman's correlation coefficient = -0.77, P < 0.001). In 4 of the 16 topics, another meta-analysis existed that was larger (n = 2) or included better controlled data (n = 2) and in all 4 cases there was no statistically significantly increased risk of malignancy. No medication or class had substantial and consistent evidence for increased risk of malignancy. However, for most medications we cannot exclude small risks or risks in population subsets. Such risks are unlikely to be possible to document robustly unless very large, collaborative studies with standardized analyses and no selective reporting are carried out.
Collapse
Affiliation(s)
- J P A Ioannidis
- Stanford Prevention Research Center, Department of Medicine and Department of Health Research and Policy, Stanford University School of Medicine, Stanford
| | | | | | | | | | | |
Collapse
|
26
|
Grabacka M, Pierzchalska M, Reiss K. Peroxisome proliferator activated receptor α ligands as anticancer drugs targeting mitochondrial metabolism. Curr Pharm Biotechnol 2013; 14:342-56. [PMID: 21133850 DOI: 10.2174/1389201011314030009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 07/15/2010] [Accepted: 09/17/2010] [Indexed: 12/15/2022]
Abstract
Tumor cells show metabolic features distinctive from normal tissues, with characteristically enhanced aerobic glycolysis, glutaminolysis and lipid synthesis. Peroxisome proliferator activated receptor α (PPAR α) is activated by nutrients (fatty acids and their derivatives) and influences these metabolic pathways acting antagonistically to oncogenic Akt and c-Myc. Therefore PPAR α can be regarded as a candidate target molecule in supplementary anticancer pharmacotherapy as well as dietary therapeutic approach. This idea is based on hitting the cancer cell metabolic weak points through PPAR α mediated stimulation of mitochondrial fatty acid oxidation and ketogenesis with simultaneous reduction of glucose and glutamine consumption. PPAR α activity is induced by fasting and its molecular consequences overlap with the effects of calorie restriction and ketogenic diet (CRKD). CRKD induces increase of NAD+/NADH ratio and drop in ATP/AMP ratio. The first one is the main stimulus for enhanced protein deacetylase SIRT1 activity; the second one activates AMP-dependent protein kinase (AMPK). Both SIRT1 and AMPK exert their major metabolic activities such as fatty acid oxidation and block of glycolysis and protein, nucleotide and fatty acid synthesis through the effector protein peroxisome proliferator activated receptor gamma 1 α coactivator (PGC-1α). PGC-1α cooperates with PPAR α and their activities might contribute to potential anticancer effects of CRKD, which were reported for various brain tumors. Therefore, PPAR α activation can engage molecular interplay among SIRT1, AMPK, and PGC-1α that provides a new, low toxicity dietary approach supplementing traditional anticancer regimen.
Collapse
Affiliation(s)
- Maja Grabacka
- Department of Food Biotechnology, Faculty of Food Technology, University of Agriculture, Krakow 30- 149, ul. Balicka 122, Poland.
| | | | | |
Collapse
|
27
|
Huang YC, Liu KC, Chiou YL, Yang CH, Chen TH, Li TT, Liu LL. Fenofibrate suppresses melanogenesis in B16-F10 melanoma cells via activation of the p38 mitogen-activated protein kinase pathway. Chem Biol Interact 2013; 205:157-64. [PMID: 23872139 DOI: 10.1016/j.cbi.2013.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 06/17/2013] [Accepted: 07/11/2013] [Indexed: 12/13/2022]
Abstract
Fenofibrate and ciglitazone belong to the classes of fibrates and thiazolidinediones, respectively. Their pharmacological actions on peroxisome proliferator-activated receptors (PPARs) present a potential therapy for hyperlipidemia and hyperglycemia. However, the melanogenesis affected by PPAR ligands in melanocytes has not been well investigated. By determining the melanin content of cells treated with PPAR agonists, we showed that fenofibrate significantly reduced melanin synthesis, but its major active metabolite, fenofibric acid, did not. Notably, the suppression of melanogenesis by fenofibrate could not be prevented by the PPARα specific antagonist GW6471. In addition, T0901317, a liver X receptor (LXR) agonist, restored the antimelanogenic activity of fenofibrate. Accordingly, fenofibrate may suppress melanogenesis through a PPARα-independent pathway. Treatment of cells with fenofibrate led to the down-regulated gene expression of melanocortin 1 receptor (MC1R). Fenofibrate also attenuated the dihydroxyphenylalanine (DOPA)-staining activity and expression of tyrosinase as well as the expression of microphthalmia-associated transcription factor (MITF). The phosphorylation of p38 mitogen-activated protein kinase (MAPK) was stimulated by fenofibrate. Furthermore, the p38 MAPK inhibitor SB203580 prevented the repressive effects of fenofibrate on the melanin production. Taken together, the results of the present study suggest that fenofibrate inhibits melanin synthesis via the down-regulation of MC1R, the up-regulation of p38 MAPK, and interference with LXR signaling pathways to decrease the expression of tyrosinase in B16-F10 melanoma cells.
Collapse
Affiliation(s)
- Yu-Chun Huang
- Department of Cosmetic Science, Providence University, No. 200, Sec. 7, Taiwan Boulevard, Shalu Dist., Taichung City 43301, Taiwan, ROC.
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
BACKGROUND Randomized trials have examined short-term effects of lifestyle interventions for diabetes prevention only among high-risk individuals. Prospective studies have examined the associations between lifestyle factors and diabetes in healthy populations but have not characterized the intervention. We estimated the long-term effects of hypothetical lifestyle interventions on diabetes in a prospective study of healthy women, using the parametric g-formula. METHODS Using data from the Nurses' Health Study, we followed 76,402 women from 1984 to 2008. We estimated the risk of type 2 diabetes under eight hypothetical interventions: quitting smoking, losing weight by 5% every 2 years if overweight/obese, exercising at least 30 minutes a day, eating less than three servings a week of red meat, eating at least two servings a day of whole grain, drinking two or more cups of coffee a day, drinking five or more grams of alcohol a day, and drinking less than one serving of soda a week. RESULTS The 24-year risk of diabetes was 9.6% under no intervention and 4.3% when all interventions were imposed (55% lower risk [95% confidence interval = 47 to 63%]). The most effective interventions were weight loss (24% lower risk), physical activity (19%), and moderate alcohol use (19%). Overweight/obese women would benefit the most, with 10.8 percentage point reduction in 24-year risk of diabetes. The validity of these estimates relies on the absence of unmeasured confounding, measurement error, and model misspecification. CONCLUSION A combination of dietary and nondietary lifestyle modifications, begun in midlife or later in relatively healthy women, could have prevented at least half of the cases of type 2 diabetes in this cohort of U.S. women.
Collapse
|
29
|
Bonovas S, Nikolopoulos GK, Bagos PG. Use of fibrates and cancer risk: a systematic review and meta-analysis of 17 long-term randomized placebo-controlled trials. PLoS One 2012; 7:e45259. [PMID: 23028888 PMCID: PMC3446944 DOI: 10.1371/journal.pone.0045259] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/16/2012] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Fibrates comprise a class of well-established antilipidemic agents that significantly reduce cardiovascular events. Given the concerns of cancer with fibrate therapy, we undertook a systematic review and meta-analysis to investigate the effects of fibrates on cancer outcomes. METHODS We systematically searched Medline, Scopus, SCI Expanded, and the Cochrane Library for studies published up to 2012. We included randomized controlled trials (RCTs) that evaluated a fibrate therapy compared with placebo, had a minimum duration of two years, and reported data on the incidence of and/or deaths from cancer during the trial. Reviews of each study were performed and the relative data were abstracted. Pooled relative risk estimates (RR) and 95% confidence intervals (CIs) were calculated using the inverse variance weighted approach. Subgroup, sensitivity and meta-regression analyses were also conducted. RESULTS Seventeen RCTs, involving 44,929 participants with an average follow-up of 5.2 years, contributed to the analysis. The degree of variability between trials was consistent with what would be expected to occur by chance alone. The quantitative synthesis of data retrieved from the RCTs was not indicative of a fibrate effect on cancer incidence (780 [fibrate] vs 814 [control]; RR = 1.02, 95% CI 0.92-1.12) or cancer death (385 [fibrate] vs 377 [control]; RR = 1.06, 95% CI: 0.92-1.22). When the analysis was restricted to major RCTs, the results did not substantially change. Similarly, we found no evidence of differential effects by length of follow-up or type of fibrate. Insignificant results were also obtained for the role of fibrates in cancers of the respiratory tract, breast, colon, gastrointestinal tract, prostate, genitourinary tract, or in melanoma. CONCLUSION Our findings demonstrate that fibrates have a neutral effect on cancer outcomes. However, it is important to continue monitoring their long-term safety profiles.
Collapse
Affiliation(s)
- Stefanos Bonovas
- Department of Pharmacology, School of Medicine, University of Athens, Athens, Greece.
| | | | | |
Collapse
|
30
|
Jagtap D, Rosenberg CA, Martin LW, Pettinger M, Khandekar J, Lane D, Ockene I, Simon MS. Prospective analysis of association between use of statins and melanoma risk in the Women's Health Initiative. Cancer 2012; 118:5124-31. [PMID: 22434400 DOI: 10.1002/cncr.27497] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Revised: 01/18/2012] [Accepted: 01/24/2012] [Indexed: 01/20/2023]
Abstract
BACKGROUND Melanoma is the most lethal form of skin cancer, with an estimated 68,130 new cases and 8700 deaths in the United States in 2010. The increasing incidence and high death rate associated with metastatic disease support the need to focus on prevention. The authors used data from the Women's Health Initiative (WHI) to assess whether 3-hydroxy-3 methylglutaryl coenzyme A inhibitors (statins) are associated with a decreased risk of melanoma. METHODS The study population consisted of 119,726 postmenopausal white women, in which 1099 cases of malignant melanoma were identified over an average (± standard deviation) of 11.6 ± 3.2 years. All diagnoses were confirmed by medical record review and pathology reports. Information on statin use was collected at baseline and during follow-up. Self-administered and interview-administered questionnaires were used to collect information on other risk factors. Cox proportional hazards regression was used to calculate hazard ratios (HRs) with 95% confidence intervals (CIs). Analyses investigated the association of any statin use, type, potency, lipophilic status, and duration of use with melanoma. RESULTS Statins were used by 8824 women (7.4%) at baseline. The annualized rate of melanoma was 0.09% among statin users and 0.09% among nonusers The multivariable adjusted HR for statin users compared with nonusers was 1.14 (95% CI, 0.91-1.43). There were no significant differences in risk based on statin type, potency, category, duration, or in time-dependent models. CONCLUSIONS There was no significant association between statin use and melanoma risk among postmenopausal women in the WHI.
Collapse
Affiliation(s)
- Deepa Jagtap
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Sertznig P, Reichrath J. Peroxisome proliferator-activated receptors (PPARs) in dermatology: Challenge and promise. DERMATO-ENDOCRINOLOGY 2011; 3:130-5. [PMID: 22110772 DOI: 10.4161/derm.3.3.15025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 01/17/2011] [Accepted: 02/01/2011] [Indexed: 02/03/2023]
Abstract
Since their discovery it has become clear that peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors involved in the genetic regulation of the lipid metabolism and energy homoeostasis. Subsequently, accumulating evidence suggests a role of PPARs in genomic pathways including the regulation of cell growth, apoptosis and differentiation. These findings indicate that PPARs and PPAR agonists play an important role in inflammatory responses and tumor promotion. Because of their diverse biologic activities on keratinocytes and other skin cells, PPARs represent a major research target for the understanding and treatment of many skin pathologies, such as hyperproliferative and inflammatory diseases. Overmore recent clinical trials identified PPARs as promising drug targets for the prevention and treatment of various diseases in the field of dermatology. The present review summarizes the current knowledge of PPAR functions in various skin disorders particularly those involving inflammation and epidermal hyperproliferation (i.e., psoriasis, atopic dermatitis, acne, scleroderma, skin malignancies).
Collapse
Affiliation(s)
- Pit Sertznig
- Department of Dermatology; RWTH Aachen University Hospital; Aachen
| | | |
Collapse
|
32
|
Long-Term Use of Nonsteroidal Anti-inflammatory Drugs Decreases the Risk of Cutaneous Melanoma: Results of a United States Case–Control Study. J Invest Dermatol 2011; 131:1460-8. [DOI: 10.1038/jid.2011.58] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
33
|
Abstract
Statins are competitive inhibitors of 3-hydroxy-3-methylyglutaryl-coenzyme A reductase and reduce low-density lipoprotein-C levels. Statins are well-tolerated drugs used for prevention of atherosclerosis and cardiovascular events. Statins possess anti-inflammatory, immunomodulatory, antioxidant, metabolic, and possible anticancer effects. Statins are reported to be effective against psoriasis, dermatitis, graft-versus-host disease, uremic pruritus, vitiligo, and hirsutism. Topical forms of statins are employed in the treatment of acne, seborrhea, rosacea, and rhinophyma. Animal studies show the beneficial effect of statins against contact dermatitis and wound healing. They have promising anti-HIV effects as well. This article succinctly reviews the various cellular and molecular effects of statins, their applications in cutaneous medicine and their side effects.
Collapse
Affiliation(s)
- Farideh Jowkar
- Dermatology Department, Shiraz University of Medical Science, Shiraz, Iran
| | | |
Collapse
|
34
|
Elmore RG, Ioffe Y, Scoles DR, Karlan BY, Li AJ. Impact of statin therapy on survival in epithelial ovarian cancer. Gynecol Oncol 2010; 111:102-5. [PMID: 20698078 DOI: 10.1016/j.ygyno.2008.06.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase inhibitors, or statins, are common therapeutic agents in the management of dyslipidemias. Epidemiologic and pharmacologic data also suggest statins have an effect on cancer biology. We sought to determine the association between use of statins and epithelial ovarian cancer survival, disease progression, and clinico-pathologic factors. METHODS After IRB approval, we performed a retrospective review of all patients with advanced stage (III/IV) epithelial ovarian or primary peritoneal cancer undergoing primary cytoreductive surgery by a gynecologist oncologist between 6/1996 and 6/2001. Abstracted data included statin use at time of initial cytoreductive surgery, as well as clinico-pathologic factors and survival. Statistical analysis included Fisher's exact test, Kaplan-Meier survival, and Cox regression analyses. RESULTS 126 patients met criteria for review. Seventeen (14%) patients were undergoing concurrent statin therapy at time of initial cytoreductive surgery. Statin users were statistically older (median age 67 years, versus 60 years for non-users, p=0.002) and had a greater incidence of diabetes mellitus (18% versus 3% in non-users, p=0.03). Of the entire cohort, 21 patients (17%) were suboptimally cytoreduced after initial surgery, with residual disease N1 cm; statin use did not correlate with incidence of optimal resection (p=0.3). Median progression-free survival for statin users was 24 months, compared to 16 months for statin non-users (p=0.007). Similarly, overall survival was significantly longer for statin users (62 months) compared to statin non-users (46 months, p=0.04). Multivariable analysis identified statin use as an independent positive prognostic factor, after controlling for age, stage, grade, and suboptimal cytoreduction (p=0.02). CONCLUSIONS These data indicate statin use in patients diagnosed with epithelial ovarian cancer is associated with improved survival, and suggest a potential suppressive impact of HMG-CoA reductase inhibitors on tumor biology. Studies are proposed to explore the molecular mechanisms underlying these clinical observations.
Collapse
Affiliation(s)
- R Geoffrey Elmore
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|
35
|
Sertznig P, Seifert M, Tilgen W, Reichrath J. Peroxisome proliferator-activated receptor (PPAR) and vitamin D receptor (VDR) signaling pathways in melanoma cells: promising new therapeutic targets? J Steroid Biochem Mol Biol 2010; 121:383-6. [PMID: 20214982 DOI: 10.1016/j.jsbmb.2010.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/05/2010] [Accepted: 03/01/2010] [Indexed: 11/22/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR) and vitamin D receptor (VDR) signaling pathways regulate a multitude of genes that are of importance for a multitude of cellular functions including cell proliferation, cell differentiation, immune responses and apoptosis. Ligands and other agents influencing the PPAR and VDR signaling pathways have been shown to reveal chemopreventive potential by mediating tumor suppressive activities in a variety of human cancers. Use of these compounds may represent a potential novel strategy to prevent melanoma pathogenesis and to inhibit melanoma progression. We recently showed that 1,25-dihydroxyvitamin D3 and some of the investigated PPAR ligands inhibited proliferation of the human melanoma cell line MeWo. In addition to this, our results gave an indication of an interconnection of the PPAR and VDR signaling pathways at the level of cross-regulation of their respective transcription factor mRNA levels. The provided link between VDR and PPAR may play an important role in treatment and prevention of melanoma. This review summarizes the currently available data on the roles of the PPARs and the VDR in pathogenesis and progression of melanoma as well as their role as promising future therapeutic targets.
Collapse
Affiliation(s)
- Pit Sertznig
- Department of Dermatology, University Hospital Aachen, Pauwelsstr. 30, 52074 Aachen, Germany.
| | | | | | | |
Collapse
|
36
|
Gamble RG, Jensen D, Suarez AL, Hanson AH, McLaughlin L, Duke J, Dellavalle RP. Outpatient Follow-up and Secondary Prevention for Melanoma Patients. Cancers (Basel) 2010; 2:1178-97. [PMID: 24281112 PMCID: PMC3835125 DOI: 10.3390/cancers2021178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 06/02/2010] [Accepted: 06/03/2010] [Indexed: 02/07/2023] Open
Abstract
Health care providers and their patients jointly participate in melanoma prevention, surveillance, diagnosis, and treatment. This paper reviews screening and follow-up strategies for patients who have been diagnosed with melanoma, based on current available evidence, and focuses on methods to assess disease recurrence and second primary occurrence. Secondary prevention, including the roles of behavioral modification and chemoprevention are also reviewed. The role of follow-up dermatologist consultation, with focused physical examinations complemented by dermatoscopy, reflectance confocal microscopy, and/or full-body mapping is discussed. Furthermore, we address the inclusion of routine imaging and laboratory assessment as components of follow-up and monitoring of advanced stage melanoma. The role of physicians in addressing the psychosocial stresses associated with a diagnosis of melanoma is reviewed.
Collapse
Affiliation(s)
- Ryan G. Gamble
- Department of Dermatology, University of Colorado Denver, Aurora, CO, USA; E-Mail: (R.G.G.); (J.D.J.)
| | - Daniel Jensen
- Department of Dermatology, University of Colorado Denver, Aurora, CO, USA; E-Mail: (R.G.G.); (J.D.J.)
| | - Andrea L. Suarez
- Department of Dermatology, University of Colorado Denver, Aurora, CO, USA; E-Mail: (R.G.G.); (J.D.J.)
| | - Anne H. Hanson
- Kansas City University of Medicine and Biosciences, Kansas City, MO, USA; E-Mail:
| | - Lauren McLaughlin
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA; E-Mail:
| | - Jodi Duke
- Department of Dermatology, University of Colorado Denver, Aurora, CO, USA; E-Mail: (R.G.G.); (J.D.J.)
- School of Pharmacy, University of Colorado, Aurora, CO, USA; E-Mail:
| | - Robert P. Dellavalle
- Department of Dermatology, University of Colorado Denver, Aurora, CO, USA; E-Mail: (R.G.G.); (J.D.J.)
- Dermatology Service, Denver Veterans Affairs Medical Center, Denver, CO, USA
- Epidemiology Department, Colorado School of Public Health, Aurora, CO, USA
| |
Collapse
|
37
|
Somerset SM. Reporting of dietary control frameworks in studies on simvastatin efficacy. Clin Nutr 2010; 29:352-7. [DOI: 10.1016/j.clnu.2009.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 10/08/2009] [Accepted: 10/19/2009] [Indexed: 11/26/2022]
|
38
|
Abstract
Gemfibrozil is long known for its ability to reduce the level of triglycerides in the blood circulation and to decrease the risk of hyperlipidemia. However, a number of recent studies reveal that apart from its lipid-lowering effects, gemfibrozil can also regulate many other signaling pathways responsible for inflammation, switching of T-helper cells, cell-to-cell contact, migration, and oxidative stress. In this review, we have made an honest attempt to analyze various biological activities of gemfibrozil and associated mechanisms that may help to consider this drug for different human disorders as primary or adjunct therapy.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | |
Collapse
|
39
|
Guyton KZ, Chiu WA, Bateson TF, Jinot J, Scott CS, Brown RC, Caldwell JC. A reexamination of the PPAR-alpha activation mode of action as a basis for assessing human cancer risks of environmental contaminants. ENVIRONMENTAL HEALTH PERSPECTIVES 2009; 117:1664-72. [PMID: 20049115 PMCID: PMC2801168 DOI: 10.1289/ehp.0900758] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 05/14/2009] [Indexed: 05/17/2023]
Abstract
BACKGROUND Diverse environmental contaminants, including the plasticizer di(2-ethylhexyl)phthalate (DEHP), are hepatocarcinogenic peroxisome proliferators in rodents. Peroxisome proliferator-activated receptor-alpha (PPAR-alpha) activation and its sequelae have been proposed to constitute a mode of action (MOA) for hepatocarcinogenesis by such agents as a sole causative factor. Further, based on a hypothesized lower sensitivity of humans to this MOA, prior reviews have concluded that rodent hepatocarcinogenesis by PPAR-alpha agonists is irrelevant to human carcinogenic risk. DATA SYNTHESIS Herein, we review recent studies that experimentally challenge the PPAR-alpha activation MOA hypothesis, providing evidence that DEHP is hepatocarcinogenic in PPAR-alpha-null mice and that the MOA but not hepatocarcinogenesis is evoked by PPAR-alpha activation in a transgenic mouse model. We further examine whether relative potency for PPAR-alpha activation or other steps in the MOA correlates with tumorigenic potency. In addition, for most PPAR-alpha agonists of environmental concern, available data are insufficient to characterize relative human sensitivity to this rodent MOA or to induction of hepatocarcinogenesis. CONCLUSIONS Our review and analyses raise questions about the hypothesized PPAR-alpha activation MOA as a sole explanation for rodent hepatocarcinogenesis by PPAR-alpha agonists and therefore its utility as a primary basis for assessing human carcinogenic risk from the diverse compounds that activate PPAR-alpha. These findings have broad implications for how MOA hypotheses are developed, tested, and applied in human health risk assessment. We discuss alternatives to the current approaches to these key aspects of mechanistic data evaluation.
Collapse
Affiliation(s)
- Kathryn Z. Guyton
- Address correspondence to K.Z. Guyton, U.S. Environmental Protection Agency, 1200 Pennsylvania Ave. NW, Mail Code: 8623P, Washington, DC 20460 USA. Telephone: (703) 347-8562. Fax: (703) 347-8692. E-mail:
| | | | | | | | | | | | | |
Collapse
|
40
|
Melanoma incidence and exposure to angiotensin-converting enzyme inhibitors and angiotensin receptor blockers. Cancer Epidemiol 2009; 33:391-5. [DOI: 10.1016/j.canep.2009.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 10/07/2009] [Accepted: 10/08/2009] [Indexed: 11/17/2022]
|
41
|
Bonovas S, Nikolopoulos G, Filioussi K, Peponi E, Bagos P, Sitaras NM. Can statin therapy reduce the risk of melanoma? A meta-analysis of randomized controlled trials. Eur J Epidemiol 2009; 25:29-35. [PMID: 19844794 DOI: 10.1007/s10654-009-9396-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 10/07/2009] [Indexed: 12/12/2022]
Abstract
A growing body of literature suggests that statins may have a chemopreventive potential against melanoma through pleiotropic anti-inflammatory, immunomodulatory, and antiangiogenesis mechanisms. Our aim was to examine this association through a detailed meta-analysis of randomized controlled trials (RCTs). A comprehensive search for trials published up to June 2009 was performed, reviews of each study were conducted and data were abstracted. Prior to meta-analysis, the studies were evaluated for publication bias and heterogeneity. Pooled relative risk estimates (RR) and 95% confidence intervals (CIs) were calculated using the fixed- and the random-effects models. Subgroup and sensitivity analyses were also conducted. Sixteen RCTs of statins for cardiovascular outcomes, involving 62,568 individuals with a mean age of 60 years and an average follow-up of nearly 4.7 years, contributed to the analysis. We found no evidence of publication bias (P = 0.47) or heterogeneity among the studies (P = 0.25). Statin use did not significantly affect the risk of developing melanoma assuming either a fixed- (RR = 0.92, 95% CI: 0.67-1.26), or a random-effects model (RR = 0.92, 95% CI: 0.62-1.36). This neutral effect was further supported by the results of subgroup and sensitivity analyses. Our findings do not support a protective effect of statins against melanoma.
Collapse
Affiliation(s)
- Stefanos Bonovas
- Department of Pharmacology, School of Medicine, University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece.
| | | | | | | | | | | |
Collapse
|
42
|
Does use of estrogens decrease the Breslow thickness of melanoma of the skin? Oral contraceptives and hormonal replacement therapy. Melanoma Res 2009; 19:327-32. [DOI: 10.1097/cmr.0b013e32832f159c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
43
|
Minichsdorfer C, Hohenegger M. Autocrine amplification loop in statin-induced apoptosis of human melanoma cells. Br J Pharmacol 2009; 157:1278-90. [PMID: 19563533 DOI: 10.1111/j.1476-5381.2009.00298.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Beside their cholesterol lowering effect, statins exert pleiotropic effects, which include anti-inflammatory, immunosuppressive and anti-proliferative actions. In higher concentrations, statins trigger apoptosis in primary cells and tumour cells. In particular, melanoma cells have been found to be susceptible to statin-induced apoptosis, although only after longer incubation times. The molecular mechanisms behind this delayed drug-induced apoptosis are still unclear. EXPERIMENTAL APPROACH The human melanoma A375 and 518A2 cell lines were exposed to various statins in a time-dependent and dose-dependent manner, and indicators of apoptosis, caspase activity and individual apoptotic pathways were analysed for 3-hydroxy-3-methylglutaryl-coenzyme A reductase dependent and independent effects. KEY RESULTS Kinetic analysis of statin-induced apoptosis revealed an apoptotic burst for exposure times longer than 24 h. While the extrinsic pathway was not activated within 24 h, longer incubation times corroborated amplification of the mitochondrial pathway with significant activation of caspase 8. Continuous refreshing of the simvastatin-containing medium abrogated the mitochondrial amplification loop via caspase 8. Moreover, conditional medium, supplemented with mevalonic acid in order to nullify a possible contamination by statins, significantly triggered caspase 8 activity. Fas ligand was excluded as a possible candidate to account for the statin-induced autocrine amplification loop. CONCLUSIONS AND IMPLICATIONS Simvastatin and atorvastatin are capable of triggering an 'autocrine' suicide factor, which amplifies apoptosis via the extrinsic pathway in human melanoma cells. This pro-apoptotic stimulus implies possible therapeutic potential and may guide feasibility for more potent statins in anti-cancer strategies.
Collapse
Affiliation(s)
- C Minichsdorfer
- Institute of Pharmacology, Center of Biomolecular Medicine and Pharmacology, Medical University of Vienna, Waehringerstrasse 13A, Vienna, Austria
| | | |
Collapse
|
44
|
|
45
|
Fildes JE, Shaw SM, Williams SG, Yonan N. Potential immunologic effects of statins in cancer following transplantation. Cancer Immunol Immunother 2009; 58:461-7. [PMID: 18523769 PMCID: PMC11030733 DOI: 10.1007/s00262-008-0541-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 05/22/2008] [Indexed: 11/29/2022]
Abstract
3-hydroxy-3-methyglutaryl CoA reductase inhibitors (statins) are frequently used following organ transplantation and have well reported pleiotropic effects, including immunomodulation, which may be of benefit in preventing graft rejection. However, the immunomodulatory effects of statins on cell transformation and malignancy, combined with the immunologic processes and administration of immunosuppression are almost completely unknown. The administration of immunosuppression is well recognised as the main cause of cancer following transplantation, so the addition of an immunomodulatory agent should be associated with an increased incidence of cancer, as immune surveillance and response may be suppressed, allowing cellular transformation and proliferation combined with lack of recognition to occur. This hypothetical review attempts to delineate the mode of action of statins in terms of pro/anti-carcinogenic mechanisms, while considering graft rejection and the presence of immunosuppression.
Collapse
Affiliation(s)
- J E Fildes
- The Transplant Centre, University Hospital of South Manchester NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK.
| | | | | | | |
Collapse
|
46
|
Koomen E, Joosse A, Herings R, Casparie M, Guchelaar H, Nijsten T. Estrogens, oral contraceptives and hormonal replacement therapy increase the incidence of cutaneous melanoma: a population-based case–control study. Ann Oncol 2009; 20:358-64. [DOI: 10.1093/annonc/mdn589] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
47
|
Grange F. [Skin cancer: what's new in clinical research?]. Ann Dermatol Venereol 2008; 134 Suppl 1:8S53-63. [PMID: 18675141 DOI: 10.1016/s0151-9638(07)80560-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Every year skin cancer is the subject of many publications. In this review of the recent literature, we selected epidemiological, clinical, and therapeutic data published between October 2006 and September 2007. Some of the studies presented in conferences in 2007 are also mentioned. New epidemiological data have been provided regarding the incidence of different types of skin cancers, the effect of certain risk factors or drug or food protective factors, the overall improvement of survival in melanoma, the epidemiological, clinical, and/or therapeutic particularities of rapidely growing melanomas and thick melanomas, and the relations between sun exposure, skin cancers, and other solid tumors. New prognostic studies in melanoma have improved the evaluation of prognosis in specific situations such as thin melanoma, acrolentiginous melanoma, and melanoma with positive sentinel lymph nodes (depending on the type of positivity) or negative sentinel lymph nodes. Important results on adjuvant interferon in melanoma have been presented. Clinical trials evaluating new therapeutic approaches in stage III and IV melanoma are going on. Several studies confirmed the clinical benefit of adjuvant irradiation on the tumor site in Merkel cell carcinoma. New treatments are being studied or have been approved in refractory forms of cutaneous T-cell lymphoma and in inoperable forms of squamous cell carcinoma and dermatofibrosarcoma protuberans.
Collapse
Affiliation(s)
- F Grange
- Service de Dermatologie, Hôpital Robert-Debré, avenue du Général-Koenig, 51092 Reims cedex, France.
| |
Collapse
|
48
|
Caldieri G, Giacchetti G, Beznoussenko G, Attanasio F, Ayala I, Buccione R. Invadopodia biogenesis is regulated by caveolin-mediated modulation of membrane cholesterol levels. J Cell Mol Med 2008; 13:1728-1740. [PMID: 19175685 DOI: 10.1111/j.1582-4934.2008.00568.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Invadopodia are proteolytically active protrusions formed by invasive tumoural cells when grown on an extracellular matrix (ECM) substratum. Clearly, invadopodia are specialized membrane domains acting as sites of signal transduction and polarized delivery of components required for focalized ECM degradation. For these reasons, invadopodia are a model to study focal ECM degradation by tumour cells. We investigated the features of invadopodia membrane domains and how altering their composition would affect invadopodia biogenesis and function. This was achieved through multiple approaches including manipulation of the levels of cholesterol and other lipids at the plasma membrane, alteration of cholesterol trafficking by acting on caveolin 1 expression and phosphorylation. We show that cholesterol depletion impairs invadopodia formation and persistence, and that invadopodia themselves are cholesterol-rich membranes. Furthermore, the inhibition of invadopodia formation and ECM degradation after caveolin 1 knock-down was efficiently reverted by simple provision of cholesterol. In addition, the inhibitory effect of caveolin 3(DGV) expression, a mutant known to block cholesterol transport to the plasma membrane, was similarly reverted by provision of cholesterol. We suggest that invadopodia biogenesis, function and structural integrity rely on appropriate levels of plasma membrane cholesterol, and that invadopodia display the properties of cholesterol-rich membranes. Also, caveolin 1 exerts its function in invadopodia formation by regulating cholesterol balance at the plasma membrane. These findings support the connection between cholesterol, cancer and caveolin 1, provide further understanding of the role of cholesterol in cancer progression and suggest a mechanistic framework for the proposed anti-cancer activity of statins, tightly related to their blood cholesterol-lowering properties.
Collapse
Affiliation(s)
- Giusi Caldieri
- Tumor Cell Invasion Laboratory, Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Chieti, Italy
| | - Giada Giacchetti
- Tumor Cell Invasion Laboratory, Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Chieti, Italy
| | - Galina Beznoussenko
- Tumor Cell Invasion Laboratory, Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Chieti, Italy
| | - Francesca Attanasio
- Tumor Cell Invasion Laboratory, Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Chieti, Italy
| | - Inmaculada Ayala
- Tumor Cell Invasion Laboratory, Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Chieti, Italy
| | - Roberto Buccione
- Tumor Cell Invasion Laboratory, Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Chieti, Italy
| |
Collapse
|
49
|
Bonovas S, Filioussi K, Sitaras NM. Statins are not associated with a reduced risk of pancreatic cancer at the population level, when taken at low doses for managing hypercholesterolemia: evidence from a meta-analysis of 12 studies. Am J Gastroenterol 2008; 103:2646-51. [PMID: 18684187 DOI: 10.1111/j.1572-0241.2008.02051.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Recent experimental research on a class of pharmacological agents that reduce plasma cholesterol, 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins), has shown promise in pancreatic cancer chemoprevention. While the mechanism remains unclear, several epidemiological studies have also evaluated the relationship between statin use and pancreatic cancer. Our aim was to examine the strength of this association through a detailed meta-analysis of the studies published on the subject in peer-reviewed literature. METHODS A comprehensive search for articles published up to December 2007 was performed, reviews of each study were conducted, and data were abstracted. Prior to meta-analysis, the studies were evaluated for publication bias and heterogeneity. Pooled relative risk (RR) estimates and 95% confidence intervals (CIs) were calculated using the random-effects model. RESULTS Twelve studies (3 randomized placebo-controlled trials [RCTs], 4 cohort, and 5 case-control studies) contributed to the analysis. The studies were grouped on the basis of study design, and separate meta-analyses were conducted. There was no evidence of an association between statin use and pancreatic cancer among either the RCTs (RR 0.99, 95% CI 0.44-2.21) or the observational studies (RR 0.86, 95% CI 0.60-1.24). Similarly, we found no evidence of publication bias. However, a high heterogeneity was detected among the observational studies. CONCLUSION Despite the chemopreventive potential of statins demonstrated in experimental studies, our results do not support the hypothesis that these agents reduce the risk of pancreatic cancer at the population level, when taken at low doses for managing hypercholesterolemia.
Collapse
Affiliation(s)
- Stefanos Bonovas
- Department of Pharmacology, School of Medicine, University of Athens, Athens, Greece
| | | | | |
Collapse
|
50
|
Kuoppala J, Lamminpää A, Pukkala E. Statins and cancer: A systematic review and meta-analysis. Eur J Cancer 2008; 44:2122-32. [PMID: 18707867 DOI: 10.1016/j.ejca.2008.06.025] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2008] [Revised: 05/29/2008] [Accepted: 06/20/2008] [Indexed: 12/23/2022]
Abstract
BACKGROUND Systematic reviews on the association between statin therapy and cancer have focused on randomised trials without assessing the quality of evidence. We aimed to review the overall evidence taking study quality into consideration. METHODS Publications of original studies on the effect of statin treatment on cancer in adult patients were searched on MEDLINE, EMBASE and CENTRAL databases upto October 2007. Our search yielded 37 eligible original studies out of 3607 references. Five studies were additionally found through manual search. Thus, 42 studies were included in the analyses: 17 randomised controlled trials, 10 cohort studies, and 15 case-control studies. FINDINGS Statins had no effect on the overall incidence of cancer (median risk ratio (RR) 0.96, range 0.72 to 1.2), or on the incidence of lung (median RR 0.92, range 0.83 to 3.0), breast (median RR 1.04, range 0.74 to 19) or prostate cancer (median RR 0.96, range 0.33 to 1.7). They seemed to protect from stomach (median RR 0.59, range 0.40 to 0.88) and liver cancer (median RR 0.62, range 0.33 to 1.2), and from lymphoma (median RR 0.74, range 0.28 to 2.2). They increased the incidence of both melanoma (median RR 1.5, range 1.3 to 1.7) and non-melanoma skin cancer (median RR 1.6, range 1.2 to 2.2). The effect varied, yet inconsistently, by statin type. The median follow-up time was 4 years. The strength of evidence was mostly weak. INTERPRETATION The evidence suggests that statins do not have short-term effects on cancer risk. The evidence on potentially protective or harmful effects is inconclusive. High quality cohort studies with long follow-up are needed to resolve the issue.
Collapse
Affiliation(s)
- Jaana Kuoppala
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | | | | |
Collapse
|