1
|
Zhang J, Yu H, Zheng X, Ming WK, Lak YS, Tom KC, Lee A, Huang H, Chen W, Lyu J, Deng L. Deep-learning-based survival prediction of patients with lower limb melanoma. Discov Oncol 2023; 14:218. [PMID: 38030951 PMCID: PMC10686915 DOI: 10.1007/s12672-023-00823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND For the purpose to examine lower limb melanoma (LLM) and its long-term survival rate, we used data from the Surveillance, Epidemiology and End Results (SEER) database. To estimate the prognosis of LLM patients and assess its efficacy, we used a powerful deep learning and neural network approach called DeepSurv. METHODS We gathered data on those who had an LLM diagnosis between 2000 and 2019 from the SEER database. We divided the people into training and testing cohorts at a 7:3 ratio using a random selection technique. To assess the likelihood that LLM patients would survive, we compared the results of the DeepSurv model with those of the Cox proportional-hazards (CoxPH) model. Calibration curves, the time-dependent area under the receiver operating characteristic curve (AUC), and the concordance index (C-index) were all used to assess how accurate the predictions were. RESULTS In this study, a total of 26,243 LLM patients were enrolled, with 7873 serving as the testing cohort and 18,370 as the training cohort. Significant correlations with age, gender, AJCC stage, chemotherapy status, surgery status, regional lymph node removal and the survival outcomes of LLM patients were found by the CoxPH model. The CoxPH model's C-index was 0.766, which signifies a good degree of predicted accuracy. Additionally, we created the DeepSurv model using the training cohort data, which had a higher C-index of 0.852. In addition to calculating the 3-, 5-, and 8-year AUC values, the predictive performance of both models was evaluated. The equivalent AUC values for the CoxPH model were 0.795, 0.767, and 0.847, respectively. The DeepSurv model, in comparison, had better AUC values of 0.872, 0.858, and 0.847. In comparison to the CoxPH model, the DeepSurv model demonstrated greater prediction performance for LLM patients, as shown by the AUC values and the calibration curve. CONCLUSION We created the DeepSurv model using LLM patient data from the SEER database, which performed better than the CoxPH model in predicting the survival time of LLM patients.
Collapse
Affiliation(s)
- Jinrong Zhang
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, 510630, China
| | - Hai Yu
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, 510630, China
| | - Xinkai Zheng
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, 510630, China
| | - Wai-Kit Ming
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China
| | - Yau Sun Lak
- Centro de Hospitalar Conde de Januario, Macau, China
| | | | - Alice Lee
- Hong Kong Medical and Education, Hong Kong, China
| | - Hui Huang
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, 510630, China
| | - Wenhui Chen
- Shanghai Aige Medical Beauty Clinic Co., Ltd. (Agge), Shanghai, China.
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, China.
| | - Liehua Deng
- Department of Dermatology, The First Affiliated Hospital of Jinan University and Jinan University Institute of Dermatology, Guangzhou, 510630, China.
- Department of Dermatology, The Fifth Affiliated Hospital of Jinan University, Heyuan, China.
| |
Collapse
|
2
|
Gajón JA, Juarez-Flores A, De León Rodríguez SG, Aguilar Flores C, Mantilla A, Fuentes-Pananá EM, Bonifaz LC. Immunotherapy Options for Acral Melanoma, A fast-growing but Neglected Malignancy. Arch Med Res 2022; 53:794-806. [PMID: 36460547 DOI: 10.1016/j.arcmed.2022.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/31/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022]
Abstract
Melanoma is the deadliest form of skin cancer. It is classified as cutaneous and non-cutaneous, with the former characterized by developing in sun-exposed areas of the skin, UV-light radiation being its most important risk factor and ordinarily affecting fair skin populations. In recent years, the incidence of melanoma has been increasing in populations with darker complexion, for example, Hispanics, in which acral melanoma is highly prevalent. The WHO estimates that the incidence and mortality of melanoma will increase by more than 60% by 2040, particularly in low/medium income countries. Acral melanoma appears in the palms, soles and nails, and because of these occult locations, it is often considered different from other cutaneous melanomas even though it also originates in the skin. Acral melanoma is very rare in Caucasian populations and is often not included from genetic analysis and clinical trials. In this review, we present the worldwide epidemiology of acral melanoma; we summarize its genetic characterization and point out important signaling pathways for targeted therapy. We also discuss how genetic analyses have shown that acral melanoma carries a sufficient mutational load and neoantigen formation to be targeted by the immune system, arguing for a potential benefit with novel immunotherapeutic strategies, alone or combined with targeted therapy. This is important because chemotherapy remains the first-line treatment in non-developed nations despite a disheartening response. In summary, the increased incidence and mortality of acral melanoma in low/medium income countries calls for increasing our knowledge about its nature and therapeutic options and leveling off the asymmetric research conducted primarily on Caucasian populations.
Collapse
Affiliation(s)
- Julian A Gajón
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México; Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Angel Juarez-Flores
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Ciudad de México, México
| | - Saraí G De León Rodríguez
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Cristina Aguilar Flores
- Unidad de Investigación Médica en Inmunología Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Alejandra Mantilla
- Servicio de Patología, Hospital de Oncología Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Ezequiel M Fuentes-Pananá
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Ciudad de México, México.
| | - Laura C Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México; Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| |
Collapse
|
3
|
Paradoxical Radiosensitizing Effect of Carnosic Acid on B16F10 Metastatic Melanoma Cells: A New Treatment Strategy. Antioxidants (Basel) 2022; 11:antiox11112166. [PMID: 36358539 PMCID: PMC9686564 DOI: 10.3390/antiox11112166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Carnosic acid (CA) is a phenolic diterpene characterized by its high antioxidant activity; it is used in industrial, cosmetic, and nutritional applications. We evaluated the radioprotective capacity of CA on cells directly exposed to X-rays and non-irradiated cells that received signals from X-ray treated cells (radiation induced bystander effect, RIBE). The genoprotective capacity was studied by in vivo and in vitro micronucleus assays. Radioprotective capacity was evaluated by clonogenic cell survival, MTT, apoptosis and intracellular glutathione assays comparing radiosensitive cells (human prostate epithelium, PNT2) with radioresistant cells (murine metastatic melanoma, B16F10). CA was found to exhibit a genoprotective capacity in cells exposed to radiation (p < 0.001) and in RIBE (p < 0.01). In PNT2 cells, considered as normal cells in our study, CA achieved 97% cell survival after exposure to 20 Gy of X-rays, eliminating 67% of radiation-induced cell death (p < 0.001), decreasing apoptosis (p < 0.001), and increasing the GSH/GSSH ratio (p < 0.01). However, the administration of CA to B16F10 cells decreased cell survival by 32%, increased cell death by 200% (p < 0.001) compared to irradiated cells, and increased cell death by 100% (p < 0.001) in RIBE bystander cells (p < 0.01). Furthermore, it increased apoptosis (p < 0.001) and decreased the GSH/GSSG ratio (p < 0.01), expressing a paradoxical radiosensitizing effect in these cells. Knowing the potential mechanisms of action of substances such as CA could help to create new applications that would protect healthy cells and exclusively damage neoplastic cells, thus presenting a new desirable strategy for cancer patients in need of radiotherapy.
Collapse
|
4
|
Abstract
In the Western population, 1 out of every 50 individuals will develop melanoma. The incidence of melanoma is increasing faster than any other malignancy. The development of melanoma is multifactorial arising from an interaction between genetic susceptibility and environmental exposures. Sixty to seventy percent of melanomas are thought to be caused by ultraviolet radiation. Most cutaneous melanomas are of increased risk. Prevention strategies involve mitigating the environmental risk factors and identifying individuals with phenotypic risk factors for increased surveillance.
Collapse
Affiliation(s)
- William W Dzwierzynski
- Department of Plastic Surgery, Medical College of Wisconsin, 1155 N. Mayfair Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
5
|
Fakhari S, Nouri A, Jamzad M, Arab‐Salmanabadi S, Falaki F. Investigation of inclusion complex of metformin into selective cyclic peptides as novel drug delivery system: Structure, electronic properties,
AIM,
and
NBO
study via
DFT. J CHIN CHEM SOC-TAIP 2020. [DOI: 10.1002/jccs.202000304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Shabnam Fakhari
- Department of Chemistry, Shahr‐e‐Qods Branch Islamic Azad University Tehran Iran
| | - Azita Nouri
- Department of Chemistry, Shahr‐e‐Qods Branch Islamic Azad University Tehran Iran
| | - Mina Jamzad
- Department of Chemistry, Shahr‐e‐Qods Branch Islamic Azad University Tehran Iran
| | | | - Foujan Falaki
- Department of Chemistry, Shahr‐e‐Qods Branch Islamic Azad University Tehran Iran
| |
Collapse
|
6
|
Gu N, Dai W, Liu H, Ge J, Luo S, Cho E, Amos CI, Lee JE, Li X, Nan H, Yuan H, Wei Q. Genetic variants in TKT and DERA in the nicotinamide adenine dinucleotide phosphate pathway predict melanoma survival. Eur J Cancer 2020; 136:84-94. [PMID: 32659474 DOI: 10.1016/j.ejca.2020.04.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/14/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cutaneous melanoma (CM) is the most lethal type of skin cancers. Nicotinamide adenine dinucleotide phosphate (NADPH) plays an important role in anabolic reactions and tumorigenesis, but many genes are involved in the NADPH system. METHODS We used 10,912 single-nucleotide polymorphisms (SNPs) (2018 genotyped and 8894 imputed) in 134 NADPH-related genes from a genome-wide association study (GWAS) of 858 patients from The University of Texas MD Anderson Cancer Center (MDACC) in a single-locus analysis to predict CM survival. We then replicated the results in another GWAS data set of 409 patients from the Nurses' Health Study (NHS) and the Health Professionals Follow-up Study (HPFS). RESULTS There were 95 of 858 (11.1%) and 48 of 409 (11.7%) patients who died of CM, respectively. In multivariable Cox regression analyses, we identified two independent SNPs (TKT rs9864057 G > A and deoxyribose phosphate aldolase (DERA) rs12297652 A > G) to be significantly associated with CM-specific survival [hazards ratio (HR) of 1.52, 95% confidence interval (CI) = 1.18-1.96, P = 1.06 × 10-3 and 1.51 (1.19-1.91, 5.89 × 10-4)] in the meta-analysis, respectively. Furthermore, an increasing number of risk genotypes of these two SNPs was associated with a higher risk of death in the MDACC, the NHS/HPFS, and their combined data sets (Ptrend<0.001, = 0.004 and <0.001, respectively). In the expression quantitative trait loci analysis, TKT rs9864057 G > A and DERA rs12297652 A > G were also significantly associated with higher mRNA expression levels in sun-exposed lower-leg skin (P = 0.043 and 0.006, respectively). CONCLUSIONS These results suggest that these two potentially functional SNPs may be valuable prognostic biomarkers for CM survival, but larger studies are needed to validate these findings.
Collapse
Affiliation(s)
- Ning Gu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Duke Cancer Institute, Duke University Medical Center, Durham, NC, 27710, USA; Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Wei Dai
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, 27710, USA; Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, 27710, USA; Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jie Ge
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, 27710, USA; Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Eunyoung Cho
- Department of Dermatology, Warren Alpert Medical School, Brown University, Providence, RI, 02912, USA; Department of Epidemiology, Brown University School of Public Health, Providence, RI, 02912, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xin Li
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Hongmei Nan
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Hua Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, 27710, USA; Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
7
|
Abstract
Metformin is a widely used biguanide drug due to its safety and low cost. It has been used for over 60 years to treat type 2 diabetes at the early stages because of its outstanding ability to decrease plasma glucose levels. Over time, different uses of metformin were discovered, and the benefits of metformin for various diseases and even aging were verified. These diseases include cancers (e.g., breast cancer, endometrial cancer, bone cancer, colorectal cancer, and melanoma), obesity, liver diseases, cardiovascular disease, and renal diseases. Metformin exerts different effects through different signaling pathways. However, the underlying mechanisms of these different benefits remain to be elucidated. The aim of this review is to provide a brief summary of the benefits of metformin and to discuss the possible underlying mechanisms.
Collapse
Affiliation(s)
- Ziquan Lv
- Department of Molecular Epidemiology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yajie Guo
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- *Correspondence: Yajie Guo
| |
Collapse
|
8
|
Su T, Wang YP, Wang XN, Li CY, Zhu PL, Huang YM, Yang ZY, Chen SB, Yu ZL. The JAK2/STAT3 pathway is involved in the anti-melanoma effects of brevilin A. Life Sci 2019; 241:117169. [PMID: 31843524 DOI: 10.1016/j.lfs.2019.117169] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/22/2019] [Accepted: 12/09/2019] [Indexed: 01/14/2023]
Abstract
AIMS Melanoma is lethal. Constitutively active signal transducer and activator of transcription 3 (STAT3) has been proposed as a pathogenic factor and a therapeutic target of melanoma. Brevilin A, a sesquiterpene lactone isolated from Centipeda minima (L.) A. Br. et Aschers., has been shown to exert antineoplastic effects and inhibit the STAT3 pathway in nasopharyngeal, lung, prostate and breast cancer cells. This study aimed to determine whether brevilin A has anti-melanoma effects, and whether STAT3 signaling is involved in the effects. MAIN METHODS A mouse A375 xenograft model, as well as A375 and A2058 cell models were employed to assess the in vivo and in vitro anti-melanoma effects of brevilin A. A375 cells stably expressing STAT3C, a constitutively active STAT3 mutant, were used to determine the role of STAT3 signaling in brevilin A's anti-melanoma effects. KEY FINDINGS Intraperitoneal injection of brevilin A dose-dependently inhibited melanoma growth in mice and suppressed STAT3 phosphorylation in the tumors. In cultured cells, brevilin A reduced cell viability, induced apoptosis, suppressed migration and invasion, decreased protein levels of phospho-JAK2 (Y1007/1008) and phospho-STAT3 (Tyr705), and restrained STAT3 nuclear localization. STAT3 over-activation diminished brevilin A's effects on cell viability and migration. Collectively, brevilin A exerts anti-melanoma effects and these effects are at least in part attributed to the inhibition of the JAK2/STAT3 pathway. SIGNIFICANCE Our findings provide a pharmacological basis for developing brevilin A as a new phytotherapeutic agent against melanoma.
Collapse
Affiliation(s)
- Tao Su
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Ya-Ping Wang
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Xin-Ning Wang
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Chun-Yu Li
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Pei-Li Zhu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Yu-Mei Huang
- Guangzhou Caizhilin Pharmaceutical Co., Ltd., Guangzhou, Guangdong, China
| | - Zhi-Ye Yang
- Guangdong Institute For Drug Control, Guangzhou, Guangdong, China
| | - Si-Bao Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhi-Ling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.
| |
Collapse
|
9
|
Rouaud F, Boucher JL, Slama-Schwok A, Rocchi S. Mechanism of melanoma cells selective apoptosis induced by a photoactive NADPH analogue. Oncotarget 2018; 7:82804-82819. [PMID: 27756874 PMCID: PMC5347734 DOI: 10.18632/oncotarget.12651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/02/2016] [Indexed: 02/07/2023] Open
Abstract
Melanoma is one of the most lethal cancers when it reaches a metastatic stage. Despite the spectacular achievements of targeted therapies (BRAF inhibitors) or immuno-therapies (anti-CTLA4 or anti-PD1), most patients with melanoma will need additional treatments. Here we used a photoactive NADPH analogue called NS1 to induce cell death by inhibition of NADPH oxidases NOX in melanoma cells, including melanoma cells isolated from patients. In contrast, healthy melanocytes growth was unaffected by NS1 treatment. NS1 established an early Endoplasmic Reticulum stress by the early release of calcium mediated by (a) calcium-dependent redox-sensitive ion channel(s). These events initiated autophagy and apoptosis in all tested melanoma cells independently of their mutational status. The autophagy promoted by NS1 was incomplete. The autophagic flux was blocked at late stage events, consistent with the accumulation of p62, and a close localization of LC3 with NS1 associated with NS1 inhibition of NOX1 in autophagosomes. This hypothesis of a specific incomplete autophagy and apoptosis driven by NS1 was comforted by the use of siRNAs and pharmacological inhibitors blocking different processes. This study highlights the potential therapeutic interest of NS1 inducing cell death by triggering a selective ER stress and incomplete autophagy in melanoma cells harbouring wt and BRAF mutation.
Collapse
Affiliation(s)
- Florian Rouaud
- INSERM U1065 Team 1, Université de Nice Sophia Antipolis et Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | | | | | - Stéphane Rocchi
- INSERM U1065 Team 1, Université de Nice Sophia Antipolis et Centre Méditerranéen de Médecine Moléculaire, Nice, France
| |
Collapse
|
10
|
Abstract
Metformin is the most common biguanide used in the treatment of diabetes, with 120 million treated patients worldwide. Metformin decreases hyperglycemia without inducing hypoglycemia in diabetic patients and is very well tolerated. The principal effects of metformin are to decrease hepatic gluconeogenesis and increase glucose absorption by skeletal muscles. These effects are primarily due to metformin's action on mitochondria, which requires the activation of metabolic checkpoint AMP-activated protein kinase (AMPK). AMPK is implicated in several pathways, and following metformin activation, it decreases protein synthesis and cell proliferation. Many studies have examined the role of metformin in the regulation of cancer cells, particularly its effects on cancer cell proliferation and cell death. Encouraging results have been obtained in different types of cancers, including prostate, breast, lung, and skin cancers (melanoma). Furthermore, many retrospective epidemiological studies in diabetes patients have shown that metformin treatment decreased the risk of cancers compared with other antidiabetic treatments. In this review, we will discuss the effects of metformin on melanoma cells. Together, our novel data demonstrate the importance of developing metformin and new biguanide-derived compounds as potential treatments against a number of different cancers, particularly melanoma.
Collapse
Affiliation(s)
- Emilie Jaune
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Université de Nice Sophia Antipolis, UFR de Médecine, Nice, France
| | - Stéphane Rocchi
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Université de Nice Sophia Antipolis, UFR de Médecine, Nice, France
- *Correspondence: Stéphane Rocchi
| |
Collapse
|
11
|
Kudryavtseva AV, Fedorova MS, Zhavoronkov A, Moskalev AA, Zasedatelev AS, Dmitriev AA, Sadritdinova AF, Karpova IY, Nyushko KM, Kalinin DV, Volchenko NN, Melnikova NV, Klimina KM, Sidorov DV, Popov AY, Nasedkina TV, Kaprin AD, Alekseev BY, Krasnov GS, Snezhkina AV. Effect of lentivirus-mediated shRNA inactivation of HK1, HK2, and HK3 genes in colorectal cancer and melanoma cells. BMC Genet 2016; 17:156. [PMID: 28105937 PMCID: PMC5249010 DOI: 10.1186/s12863-016-0459-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The switch from oxidative phosphorylation to glycolysis in proliferating cancer cells, even under aerobic conditions, has been shown first in 1926 by Otto Warburg. Today this phenomenon is known as the “Warburg effect” and recognized as a hallmark of cancer. The metabolic shift to glycolysis is associated with the alterations in signaling pathways involved in energy metabolism, including glucose uptake and fermentation, and regulation of mitochondrial functions. Hexokinases (HKs), which catalyze the first step of glycolysis, have been identified to play a role in tumorigenesis of human colorectal cancer (CRC) and melanoma. However, the mechanism of action of HKs in the promotion of tumor growth remains unclear. Results The purpose of the present study was to investigate the effect of silencing of hexokinase genes (HK1, HK2, and HK3) in colorectal cancer (HT-29, SW 480, HCT-15, RKO, and HCT 116) and melanoma (MDA-MB-435S and SK-MEL-28) cell lines using short hairpin RNA (shRNA) lentiviral vectors. shRNA lentiviral plasmid vectors pLSLP-HK1, pLSLP-HK2, and pLSLP-HK3 were constructed and then transfected separately or co-transfected into the cells. HK2 inactivation was associated with increased expression of HK1 in colorectal cancer cell lines pointing to the compensation effect. Simultaneous attenuation of HK1 and HK2 levels led to decreased cell viability. Co-transfection with shRNA vectors against HK1, HK2, and HK3 mRNAs resulted in a rapid cell death via apoptosis. Conclusions We have demonstrated that simultaneous inactivation of HK1 and HK2 was sufficient to decrease proliferation and viability of melanoma and colorectal cancer cells. Our results suggest that HK1 and HK2 could be the key therapeutic targets for reducing aerobic glycolysis in examined cancers.
Collapse
Affiliation(s)
- Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia. .,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alex Zhavoronkov
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University Eastern Campus, Baltimore, Maryland, USA
| | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Alexander S Zasedatelev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Asiya F Sadritdinova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Irina Y Karpova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Kirill M Nyushko
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Nadezhda N Volchenko
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Kseniya M Klimina
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry V Sidorov
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Tatiana V Nasedkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|
12
|
Targeting metabolic flexibility by simultaneously inhibiting respiratory complex I and lactate generation retards melanoma progression. Oncotarget 2016; 6:37281-99. [PMID: 26484566 PMCID: PMC4741930 DOI: 10.18632/oncotarget.6134] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/23/2015] [Indexed: 12/20/2022] Open
Abstract
Melanoma is a largely incurable skin malignancy owing to the underlying molecular and metabolic heterogeneity confounded by the development of resistance. Cancer cells have metabolic flexibility in choosing either oxidative phosphorylation (OXPHOS) or glycolysis for ATP generation depending upon the nutrient availability in tumor microenvironment. In this study, we investigated the involvement of respiratory complex I and lactate dehydrogenase (LDH) in melanoma progression. We show that inhibition of complex I by metformin promotes melanoma growth in mice via elevating lactate and VEGF levels. In contrast, it leads to the growth arrest in vitro because of enhanced extracellular acidification as a result of increased glycolysis. Inhibition of LDH or lactate generation causes decrease in glycolysis with concomitant growth arrest both in vitro and in vivo. Blocking lactate generation in metformin-treated melanoma cells results in diminished cell proliferation and tumor progression in mice. Interestingly, inhibition of either LDH or complex I alone does not induce apoptosis, whereas inhibiting both together causes depletion in cellular ATP pool resulting in metabolic catastrophe induced apoptosis. Overall, our study suggests that LDH and complex I play distinct roles in regulating glycolysis and cell proliferation. Inhibition of these two augments synthetic lethality in melanoma.
Collapse
|
13
|
Cerezo M, Lehraiki A, Millet A, Rouaud F, Plaisant M, Jaune E, Botton T, Ronco C, Abbe P, Amdouni H, Passeron T, Hofman V, Mograbi B, Dabert-Gay AS, Debayle D, Alcor D, Rabhi N, Annicotte JS, Héliot L, Gonzalez-Pisfil M, Robert C, Moréra S, Vigouroux A, Gual P, Ali MMU, Bertolotto C, Hofman P, Ballotti R, Benhida R, Rocchi S. Compounds Triggering ER Stress Exert Anti-Melanoma Effects and Overcome BRAF Inhibitor Resistance. Cancer Cell 2016; 29:805-819. [PMID: 27238082 DOI: 10.1016/j.ccell.2016.04.013] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 02/15/2016] [Accepted: 04/27/2016] [Indexed: 11/16/2022]
Abstract
We have discovered and developed a series of molecules (thiazole benzenesulfonamides). HA15, the lead compound of this series, displayed anti-cancerous activity on all melanoma cells tested, including cells isolated from patients and cells that developed resistance to BRAF inhibitors. Our molecule displayed activity against other liquid and solid tumors. HA15 also exhibited strong efficacy in xenograft mouse models with melanoma cells either sensitive or resistant to BRAF inhibitors. Transcriptomic, proteomic, and biochemical studies identified the chaperone BiP/GRP78/HSPA5 as the specific target of HA15 and demonstrated that the interaction increases ER stress, leading to melanoma cell death by concomitant induction of autophagic and apoptotic mechanisms.
Collapse
Affiliation(s)
- Michaël Cerezo
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Abdelali Lehraiki
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Antoine Millet
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice cedex 2, France
| | - Florian Rouaud
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Magali Plaisant
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Emilie Jaune
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Thomas Botton
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Cyril Ronco
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice cedex 2, France
| | - Patricia Abbe
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Hella Amdouni
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice cedex 2, France
| | - Thierry Passeron
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; Service de Dermatologie, Hôpital Archet II, CHU, 06204 Nice, France
| | - Veronique Hofman
- UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; Institute of Research on Cancer and Ageing of Nice (IRCAN), INSERM U1081, CNRS UMR7284, Nice 06107, France; Laboratoire de pathologie clinique et expérimentale et Hospital-related biobank (BB-0033-00025), Hôpital Pasteur, 06002 Nice, France
| | - Baharia Mograbi
- UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; Institute of Research on Cancer and Ageing of Nice (IRCAN), INSERM U1081, CNRS UMR7284, Nice 06107, France
| | - Anne-Sophie Dabert-Gay
- UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; CNRS UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Sophia Antipolis, France
| | - Delphine Debayle
- UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; CNRS UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Sophia Antipolis, France
| | - Damien Alcor
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France
| | - Nabil Rabhi
- University Lille, CNRS, Institut Pasteur de Lille, UMR 8199 - EGID, 59000 Lille, France
| | | | - Laurent Héliot
- Equipe Biophotonique Cellulaire Fonctionnelle, Laboratoire de Physique des Lasers, Atomes et Molécules (PhLAM) GDR 2588, 59658 Villeneuve d'Ascq, France
| | - Mariano Gonzalez-Pisfil
- Equipe Biophotonique Cellulaire Fonctionnelle, Laboratoire de Physique des Lasers, Atomes et Molécules (PhLAM) GDR 2588, 59658 Villeneuve d'Ascq, France
| | - Caroline Robert
- Department of Dermatology, Cancer Campus, Gustave Roussy Institute, 114, rue Edouard-Vaillant, 94805 Villejuif, France
| | - Solange Moréra
- Institute for Integrative Biology of the Cell (I2BC), CNRS CEA University Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Armelle Vigouroux
- Institute for Integrative Biology of the Cell (I2BC), CNRS CEA University Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Philippe Gual
- INSERM, U1065, Team 8, Centre Méditerranéen de Médecine Moléculaire (C3M), 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France
| | - Maruf M U Ali
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Corine Bertolotto
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; Service de Dermatologie, Hôpital Archet II, CHU, 06204 Nice, France
| | - Paul Hofman
- UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; Institute of Research on Cancer and Ageing of Nice (IRCAN), INSERM U1081, CNRS UMR7284, Nice 06107, France; Laboratoire de pathologie clinique et expérimentale et Hospital-related biobank (BB-0033-00025), Hôpital Pasteur, 06002 Nice, France
| | - Robert Ballotti
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; Service de Dermatologie, Hôpital Archet II, CHU, 06204 Nice, France
| | - Rachid Benhida
- Institut de Chimie de Nice UMR UNS-CNRS 7272, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice cedex 2, France.
| | - Stéphane Rocchi
- INSERM, U1065, Equipe Biologie et Pathologie des cellules mélanocytaire: de la pigmentation cutanée au mélanome, Centre Méditerranéen de Médecine Moléculaire (C3M), Bâtiment ARCHIMED, 151 route de Saint Antoine de Ginestière, 06204 Nice cedex 3, France; UFR de Médecine, Université de Nice Sophia Antipolis, 06000 Nice, France; Service de Dermatologie, Hôpital Archet II, CHU, 06204 Nice, France.
| |
Collapse
|
14
|
Oswald W, Mayer C, Tarmann L, Langmann G, Wackernagel W. PROCHORN – Prospektives Register von Patientinnen mit Aderhautnävi. SPEKTRUM DER AUGENHEILKUNDE 2013. [DOI: 10.1007/s00717-013-0192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
Cerezo M, Tichet M, Abbe P, Ohanna M, Lehraiki A, Rouaud F, Allegra M, Giacchero D, Bahadoran P, Bertolotto C, Tartare-Deckert S, Ballotti R, Rocchi S. Metformin blocks melanoma invasion and metastasis development in AMPK/p53-dependent manner. Mol Cancer Ther 2013; 12:1605-15. [PMID: 23741061 DOI: 10.1158/1535-7163.mct-12-1226-t] [Citation(s) in RCA: 168] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Metformin was reported to inhibit the proliferation of many cancer cells, including melanoma cells. In this report, we investigated the effect of metformin on melanoma invasion and metastasis development. Using different in vitro approaches, we found that metformin inhibits cell invasion without affecting cell migration and independently of antiproliferation action. This inhibition is correlated with modulation of expression of proteins involved in epithelial-mesenchymal transition such as Slug, Snail, SPARC, fibronectin, and N-cadherin and with inhibition of MMP-2 and MMP-9 activation. Furthermore, our data indicate that this process is dependent on activation of AMPK and tumor suppressor protein p53. Finally, we showed that metformin inhibits melanoma metastasis development in mice using extravasation and metastasis models. The presented data reinforce the fact that metformin might be a good candidate for clinical trial in melanoma treatment.
Collapse
Affiliation(s)
- Michaël Cerezo
- Equipe Biologie et Pathologie des cellulesmelanocytaire: de la pigmentation cutanee au melanome, Centre Mediterraneen de Medecine Moleculaire (C3M), INSERM, U1065
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Superficial spreading and nodular melanoma are distinct biological entities: a challenge to the linear progression model. Melanoma Res 2012; 22:1-8. [PMID: 22108608 DOI: 10.1097/cmr.0b013e32834e6aa0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The classification of melanoma subtypes into prognostically relevant and therapeutically insightful categories has been a challenge since the first description of melanoma in the 1800s. One limitation has been the assumption that the two most common histological subtypes of melanoma, superficial spreading and nodular, evolve according to a linear model of progression, as malignant melanocytes spread radially and then invade vertically. However, recent clinical, pathological, and molecular data indicate that these two histological subtypes might evolve as distinct entities. Here, we review the published data that support distinct molecular characterization of superficial spreading and nodular melanoma, the clinical significance of this distinction including prognostic relevance and the therapeutic implications.
Collapse
|
17
|
Abstract
Metformin is the most widely used antidiabetic drug because of its proven efficacy and limited secondary effects. Interestingly, recent studies have reported that metformin can block the growth of different tumor types. Here, we show that metformin exerts antiproliferative effects on melanoma cells, whereas normal human melanocytes are resistant to these metformin-induced effects. To better understand the basis of this antiproliferative effect of metformin in melanoma, we characterized the sequence of events underlying metformin action. We showed that 24 h metformin treatment induced a cell cycle arrest in G0/G1 phases, while after 72 h, melanoma cells underwent autophagy as demonstrated by electron microscopy, immunochemistry, and by quantification of the autolysosome-associated LC3 and Beclin1 proteins. In addition, 96 h post metformin treatment we observed robust apoptosis of melanoma cells. Interestingly, inhibition of autophagy by knocking down LC3 or ATG5 decreased the extent of apoptosis, and suppressed the antiproliferative effect of metformin on melanoma cells, suggesting that apoptosis is a consequence of autophagy. The relevance of these observations were confirmed in vivo, as we showed that metformin treatment impaired the melanoma tumor growth in mice, and induced autophagy and apoptosis markers. Taken together, our data suggest that metformin has an important impact on melanoma growth, and may therefore be beneficial in patients with melanoma.
Collapse
|
18
|
Moretti RM, Mai S, Montagnani Marelli M, Bani MR, Ghilardi C, Giavazzi R, Taylor DM, Martini PGV, Limonta P. Dual targeting of tumor and endothelial cells by gonadotropin-releasing hormone agonists to reduce melanoma angiogenesis. Endocrinology 2010; 151:4643-53. [PMID: 20685877 DOI: 10.1210/en.2010-0163] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We showed previously that GnRH receptors are expressed in melanoma cells; their activation reduces cell growth and metastatic behavior. Here, we investigated whether GnRH agonists might affect the expression of genes involved in melanoma progression. By genome-wide transcriptomic and real-time PCR analysis, we first observed that GnRH agonists decrease the expression of the pro-angiogenic factor vascular endothelial growth factor (VEGF) (all isoforms) in BLM melanoma cells. Then, we demonstrated that GnRH agonists specifically decrease the expression of the VEGF165 isoform as well as its secretion from BLM cells. These data suggested that activation of GnRH receptors might reduce the pro-angiogenic behavior of melanoma cells. To verify this hypothesis, we treated BLM cells with a GnRH agonist; the conditioned medium from these cells was tested to assess its capability to stimulate human umbilical vein endothelial cell (HUVEC) motility. The migration of HUVECs towards the conditioned medium of GnRH agonist-treated BLM cells was significantly lower than the migration of HUVECs toward the conditioned medium of untreated cells. Thus, GnRH agonists reduce the pro-angiogenic behavior of melanoma cells through a decreased production of bioactive VEGF. We then found that GnRH receptors are also expressed on HUVECs and that GnRH agonists reduce their ability to proliferate and to form capillary-like tubes when stimulated by VEGF. These findings suggest that GnRH agonists exert an anti-angiogenic activity indirectly by decreasing VEGF secretion from tumor cells and directly by counteracting the pro-angiogenic activity of the growth factor. These data might lead to the development of novel targeted approaches for melanoma.
Collapse
Affiliation(s)
- Roberta M Moretti
- Department of Endocrinology, Physiopathology, and Applied Biology, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Botton T, Puissant A, Cheli Y, Tomic T, Giuliano S, Fajas L, Deckert M, Ortonne JP, Bertolotto C, Tartare-Deckert S, Ballotti R, Rocchi S. Ciglitazone negatively regulates CXCL1 signaling through MITF to suppress melanoma growth. Cell Death Differ 2010; 18:109-21. [PMID: 20596077 DOI: 10.1038/cdd.2010.75] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
We have previously demonstrated that the thiazolidinedione ciglitazone inhibited, independently of PPARγ activation, melanoma cell growth. Further investigations now show that ciglitazone effects are mediated through the regulation of secreted factors. Q-PCR screening of several genes involved in melanoma biology reveals that ciglitazone inhibits expression of the CXCL1 chemokine gene. CXCL1 is overexpressed in melanoma and contributes to tumorigenicity. We show that ciglitazone induces a diminution of CXCL1 level in different human melanoma cell lines. This effect is mediated by the downregulation of microphthalmia-associated transcription factor, MITF, the master gene in melanocyte differentiation and involved in melanoma development. Further, recombinant CXCL1 protein is sufficient to abrogate thiazolidinedione effects such as apoptosis induction, whereas extinction of the CXCL1 pathway mimics phenotypic changes observed in response to ciglitazone. Finally, inhibition of human melanoma tumor development in nude mice treated with ciglitazone is associated with a strong decrease in MITF and CXCL1 levels. Our results show that anti-melanoma effects of thiazolidinediones involve an inhibition of the MITF/CXCL1 axis and highlight the key role of this specific pathway in melanoma malignancy.
Collapse
Affiliation(s)
- T Botton
- INSERM, U895, équipe 1 Nice, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Woodall CE, Martin RCG, Stromberg AJ, Ginter B, Burton A, Ross MI, Edwards MJ, Mcmasters KM, Scoggins CR. Do Melanoma Patients from Southern Climates have a Worse Outcome than those from Northern Climates? Am Surg 2009. [DOI: 10.1177/000313480907500809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sun exposure is known to cause melanoma; what is not known is whether patients from the Southern United States have a different profile of clinicopathologic factors and outcomes than those from the Northern United States. Data from a prospective, randomized trial on surgery for cutaneous melanoma were analyzed. All patients underwent wide excision and sentinel lymph node biopsy. Patients were categorized into two groups: Northern or Southern according to their state of residence. Clinicopathologic factors and outcomes were compared between groups. A total of 2025 patients were included in the analysis; 1214 (60%) were from Southern states. The median follow-up was 52 months. Despite significant differences in clinicopathologic features between groups on both univariate and multivariate analysis, two important factors, namely primary tumor thickness and ulceration were not different, nor was the rate of lymph node metastasis. Additionally, there were no differences in disease-free survival or overall survival between the two groups. Significant differences exist between primary melanomas based on geographic regions; however there are no differences in survival. Cumulative versus episodic sun exposure may play some role in these differences.
Collapse
Affiliation(s)
- Charles E. Woodall
- Department of Surgery, Division of Surgical Oncology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Robert C. G. Martin
- Department of Surgery, Division of Surgical Oncology, University of Louisville School of Medicine, Louisville, Kentucky
| | | | - Brooke Ginter
- Department of Surgery, Division of Surgical Oncology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Alison Burton
- Department of Surgery, Division of Surgical Oncology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Merrick I. Ross
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J. Edwards
- Department of Surgery, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Kelly M. Mcmasters
- Department of Surgery, Division of Surgical Oncology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Charles R. Scoggins
- Department of Surgery, Division of Surgical Oncology, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
21
|
Nihal M, Ahsan H, Siddiqui IA, Mukhtar H, Ahmad N, Wood GS. (-)-Epigallocatechin-3-gallate (EGCG) sensitizes melanoma cells to interferon induced growth inhibition in a mouse model of human melanoma. Cell Cycle 2009; 8:2057-63. [PMID: 19502799 DOI: 10.4161/cc.8.13.8862] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Melanoma incidence has increased over the last few decades and metastatic melanoma is one of the hardest malignancies to treat. Thus, novel approaches are needed for an effective management of melanoma. Interferon-alpha2b (IFN), an immunomodulatory cytokine commonly used in melanoma treatment, has shown marginal efficacy and often results in discontinuation of therapy due to toxicity. We earlier demonstrated that epigallocatechin-3-gallate (EGCG), the major polyphenolic constituent of green tea, caused cell cycle arrest and apoptosis of human melanoma cells via modulation in cki-cyclin-cdk machinery and Bcl-2 family proteins. This study was undertaken to determine if EGCG could enhance the anti-proliferative effects of IFN. In this study, we demonstrated that EGCG and/or IFN treatments to melanoma cells resulted in a marked (1) decrease in cell proliferation and colony formation ability, and (2) induction of apoptosis. Interestingly, the combination was found to be more effective than either of the agents alone. Further, the anti-proliferative effects of EGCG and/or IFN were accompanied with an increase in Fas protein levels and a decrease in nuclear factor NFkappaB/p65 in the nucleus as well as NFkappaB promoter activity. EGCG and/or IFN also resulted in an increase in Fas-L mediated apoptosis. Further, EGCG and/or IFN treatments resulted in a decrease in melanoma tumor growth and protein levels of proliferation marker PCNA, in athymic nude mice implanted with melanoma tumors. The combination of the two modalities demonstrated a better response than either of them alone. Our data suggest that EGCG could impart therapeutic advantage if used in conjunction with IFN.
Collapse
Affiliation(s)
- Minakshi Nihal
- Department of Dermatology, University of Wisconsin Medical School and William S. Middleton VA Medical Center, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Bristow IR, Bowling J. Dermoscopy as a technique for the early identification of foot melanoma. J Foot Ankle Res 2009; 2:14. [PMID: 19435498 PMCID: PMC2694773 DOI: 10.1186/1757-1146-2-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Accepted: 05/12/2009] [Indexed: 01/03/2023] Open
Abstract
Malignant melanoma is the most common primary malignant tumour arising on the foot. Where improvements in the prognosis have been observed for patients with melanoma elsewhere on the skin, pedal lesions are still frequently delayed in presentation through neglect or misdiagnosis. Detection of foot melanoma relies on the health care practitioner's skills and observations in recognising early changes. Recent publications have documented the use a dermoscopy as a tool to improve recognition of such suspicious lesions. This paper reviews current literature with a special emphasis of its potential applications on plantar and nail unit melanoma. Data from these studies suggest that the technique is a useful and significant adjunct to clinical examination, which ultimately may lead to earlier recognition of this aggressive tumour.
Collapse
Affiliation(s)
- Ivan R Bristow
- School of Health Sciences, University of Southampton, UK.
| | | |
Collapse
|
24
|
Botton T, Puissant A, Bahadoran P, Annicotte JS, Fajas L, Ortonne JP, Gozzerino G, Zamoum T, Tartare-Deckert S, Bertolotto C, Ballotti R, Rocchi S. In vitro and in vivo anti-melanoma effects of ciglitazone. J Invest Dermatol 2009; 129:1208-18. [PMID: 19177142 DOI: 10.1038/jid.2008.346] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of PPARgamma by synthetic ligands, thiazolidinediones, inhibits the proliferation of cancer cells. In this report, focusing our attention on ciglitazone, we show that ciglitazone inhibits melanoma growth by inducing apoptosis and cell-cycle arrest, whereas normal melanocytes are resistant to ciglitazone. In melanoma cells, ciglitazone-induced apoptosis is associated with caspase activations and a loss of mitochondrial membrane potential. Induction of cell-cycle arrest by ciglitazone is associated with changes in expression of key cell-cycle regulators such as p21, cyclin D1, and pRB hypophosphorylation. Cell-cycle arrest occurs at low ciglitazone concentrations and through a PPARgamma-dependent pathway, whereas the induction of apoptosis is caused by higher ciglitazone concentrations and independently of PPARgamma. These results allow an effective molecular dissociation between proapoptotic effects and growth inhibition evoked by ciglitazone in melanoma cells. Finally, we show that in vivo treatment of nude mice by ciglitazone dramatically inhibits human melanoma xenograft development. The data presented suggest that ciglitazone might be a better candidate for clinical trials in melanoma treatment than the thiazolidinediones currently used in the treatment of type 2 diabetes, such as rosiglitazone, which is devoid of a proapoptotic PPARgamma-independent function.
Collapse
Affiliation(s)
- Thomas Botton
- INSERM, U895, Biologie et Pathologie des Cellules Mélanocytaires: de la Pigmentation Cutanée au Mélanome, Nice, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Hensen P, Müller ML, Stadler R, Luger TA. Dermatologic subspecialties in German inpatient dermatology:a national survey. J Dtsch Dermatol Ges 2008; 6:735-40. [PMID: 18371050 DOI: 10.1111/j.1610-0387.2008.06677.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND In German-speaking countries, the specialty of dermatology is characterized by a large amount of subspecialties. Data providing information whether and to what extent these subspecialties are represented in German dermatology departments and clinics is not available. METHODS A national questionnaire survey was performed to determine the extent of involvement of dermatological subspecialties in dermatologic hospitals.Methods of descriptive statistics were applied. RESULTS Overall, 90 dermatologic departments participated in this survey with a response rate of 78.3 %.The subspecialties allergology, dermatooncology,and dermatologic surgery are represented in over 90 % of departments; the sub-specialties andrology, aesthetic dermatology, dermatopathology, microbiology,phlebology,photobiology,proctology, and wound healing are represented in over 50 % of clinics. Furthermore, the subspecialties allergology, dermato-oncology, photobiology, and dermatopathology have strong research or academic representation. CONCLUSIONS The present survey demonstrates that dermatologic subspecialties are widely represented in dermatologic departments and clinics not only by individuals with specialty training but also by those in higher academic posts. This demonstrated high expertise should be maintained in German dermatology and could be promoted by academic programs.
Collapse
Affiliation(s)
- Peter Hensen
- Department of Dermatology, University of Münster, Germany.
| | | | | | | |
Collapse
|
26
|
Exposure to sunlamps, tanning beds, and melanoma risk. Cancer Causes Control 2008; 19:659-69. [PMID: 18273687 DOI: 10.1007/s10552-008-9129-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 01/29/2008] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To estimate the separate effects of sunlamp and tanning bed device use on melanoma risk. METHODS Population-based case-control study of 423 cases of melanoma and 678 controls in the state of New Hampshire. Exposure data, including sunlamp and tanning bed use, were collected by telephone interview. Associations were evaluated using logistic regression analyses. RESULTS About 17% of participants ever used a sunlamp, and most use (89%) occurred before 1980. The OR was 1.39 (95% CI 1.00-1.96) for ever using a sunlamp, 1.23 (95% CI 0.81-1.88) for those starting sunlamp use <20 years, and 1.71 (95% CI 1.00-2.92) for those starting >/=20 years. Data suggested increasing risk with number of sunlamp uses and with duration of use (tests of trend p = 0.02). The overall prevalence of tanning bed use was 22% and most use (83%) occurred after 1980. The OR was 1.14 (95% CI 0.80-1.61) for ever using a tanning bed; there was no evidence that risk increased with frequency or duration of use. The OR was 1.96 (95% CI 1.06-3.61) for having used both devices. CONCLUSION Results suggest a modest association between sunlamp use and melanoma risk, and increasing risk with greater frequency and duration of use. No association with tanning bed use was found, but sufficient lag time may not have elapsed to assess a potential effect.
Collapse
|
27
|
Hepatic Arterial Chemoembolization for Management of Metastatic Melanoma. AJR Am J Roentgenol 2008; 190:99-104. [DOI: 10.2214/ajr.07.2675] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Cotter MA, Thomas J, Cassidy P, Robinette K, Jenkins N, Florell SR, Leachman S, Samlowski WE, Grossman D. N-acetylcysteine protects melanocytes against oxidative stress/damage and delays onset of ultraviolet-induced melanoma in mice. Clin Cancer Res 2007; 13:5952-8. [PMID: 17908992 PMCID: PMC2409148 DOI: 10.1158/1078-0432.ccr-07-1187] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE UV radiation is the major environmental risk factor for melanoma and a potent inducer of oxidative stress, which is implicated in the pathogenesis of several malignancies. We evaluated whether the thiol antioxidant N-acetylcysteine (NAC) could protect melanocytes from UV-induced oxidative stress/damage in vitro and from UV-induced melanoma in vivo. EXPERIMENTAL DESIGN In vitro experiments used the mouse melanocyte line melan-a. For in vivo experiments, mice transgenic for hepatocyte growth factor and survivin, shown previously to develop melanoma following a single neonatal dose of UV irradiation, were given NAC (7 mg/mL; mother's drinking water) transplacentally and through nursing until 2 weeks after birth. RESULTS NAC (1-10 mmol/L) protected melan-a cells from several UV-induced oxidative sequelae, including production of intracellular peroxide, formation of the signature oxidative DNA lesion 8-oxoguanine, and depletion of free reduced thiols (primarily glutathione). Delivery of NAC reduced thiol depletion and blocked formation of 8-oxoguanine in mouse skin following neonatal UV treatment. Mean onset of UV-induced melanocytic tumors was significantly delayed in NAC-treated compared with control mice (21 versus 14 weeks; P = 0.0003). CONCLUSIONS Our data highlight the potential importance of oxidative stress in the pathogenesis of melanoma and suggest that NAC may be useful as a chemopreventive agent.
Collapse
Affiliation(s)
- Murray A Cotter
- Department of Dermatology, Huntsman Cancer Institute, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
DeChello LM, Sheehan TJ. The geographic distribution of melanoma incidence in Massachusetts, adjusted for covariates. Int J Health Geogr 2006; 5:31. [PMID: 16884528 PMCID: PMC1557666 DOI: 10.1186/1476-072x-5-31] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 08/02/2006] [Indexed: 11/10/2022] Open
Abstract
Background The aims of this study were to determine whether observed geographic variations in melanoma cancer incidence in both gender groups are simply random or are statistically significant, whether statistically significant excesses are temporary or persistent, and whether they can be explained by risk factors such as socioeconomic status (SES) or the percent of the population residing in an urban rather than a rural area. Between 1990 and 1999, 4774 female and 5688 male melanomas were diagnosed in Massachusetts residents. Cases were aggregated to census tracts and analyzed for deviations from random occurrence with respect to both spatial location and time. Results Thirteen geographic areas that deviated significantly from randomness were uncovered in the age-adjusted analyses of males: five with higher incidence rates than expected and eight lower than expected. In the age-adjusted analyses of females, six areas with higher incidence rates and eight areas with lower than expected incidence rates were found. After adjustment for SES and percent urban, several of these areas were no longer significantly different. Conclusion These analyses identify geographic areas with invasive melanoma incidence higher or lower than expected, the times of their excess, and whether or not their status is affected when the model is adjusted for risk factors. These surveillance findings can be a sound starting point for the shoe-leather epidemiologist.
Collapse
Affiliation(s)
- Laurie M DeChello
- Department of Community Medicine and Health Care, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030-6325, USA
| | - T Joseph Sheehan
- Department of Community Medicine and Health Care, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT 06030-6325, USA
| |
Collapse
|