101
|
Chai X, Chu H, Yang X, Meng Y, Shi P, Gou S. Metformin Increases Sensitivity of Pancreatic Cancer Cells to Gemcitabine by Reducing CD133+ Cell Populations and Suppressing ERK/P70S6K Signaling. Sci Rep 2015; 5:14404. [PMID: 26391180 PMCID: PMC4585731 DOI: 10.1038/srep14404] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/26/2015] [Indexed: 12/14/2022] Open
Abstract
The prognosis of pancreatic cancer remains dismal, with little advance in chemotherapy because of its high frequency of chemoresistance. Metformin is widely used to treat type II diabetes, and was shown recently to inhibit pancreatic cancer stem cell proliferation. In the present study, we investigated the role of metformin in chemoresistance of pancreatic cancer cells to gemcitabine, and its possible cellular and molecular mechanisms. Metformin increases sensitivity of pancreatic cancer cells to gemcitabine. The mechanism involves, at least in part, the inhibition of CD133+ cells proliferation and suppression of P70S6K signaling activation via inhibition of ERK phosphorylation. Studies of primary tumor samples revealed a relationship between P70S6K signaling activation and the malignancy of pancreatic cancer. Analysis of clinical data revealed a trend of the benefit of metformin for pancreatic cancer patients with diabetes. The results suggested that metformin has a potential clinical use in overcoming chemoresistance of pancreatic cancer.
Collapse
Affiliation(s)
- Xinqun Chai
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Hongpeng Chu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Xuan Yang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Yuanpu Meng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Pengfei Shi
- Department of Breast and Thyroid Surgery, Central Hospital of Wuhan
| | - Shanmiao Gou
- Pancreatic Disease Institute, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
102
|
Chang HH, Young SH, Sinnett-Smith J, Chou CEN, Moro A, Hertzer KM, Hines OJ, Rozengurt E, Eibl G. Prostaglandin E2 activates the mTORC1 pathway through an EP4/cAMP/PKA- and EP1/Ca2+-mediated mechanism in the human pancreatic carcinoma cell line PANC-1. Am J Physiol Cell Physiol 2015; 309:C639-49. [PMID: 26310818 DOI: 10.1152/ajpcell.00417.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/14/2015] [Indexed: 02/07/2023]
Abstract
Obesity, a known risk factor for pancreatic cancer, is associated with inflammation and insulin resistance. Proinflammatory prostaglandin E2 (PGE2) and elevated insulin-like growth factor type 1 (IGF-1), related to insulin resistance, are shown to play critical roles in pancreatic cancer progression. We aimed to explore a potential cross talk between PGE2 signaling and the IGF-1/Akt/mammalian target of rapamycin complex 1 (mTORC1) pathway in pancreatic cancer, which may be a key to unraveling the obesity-cancer link. In PANC-1 human pancreatic cancer cells, we showed that PGE2 stimulated mTORC1 activity independently of Akt, as evaluated by downstream signaling events. Subsequently, using pharmacological and genetic approaches, we demonstrated that PGE2-induced mTORC1 activation is mediated by the EP4/cAMP/PKA pathway, as well as an EP1/Ca(2+)-dependent pathway. The cooperative roles of the two pathways were supported by the maximal inhibition achieved with the combined pharmacological blockade, and the coexistence of highly expressed EP1 (mediating the Ca(2+) response) and EP2 or EP4 (mediating the cAMP/PKA pathway) in PANC-1 cells and in the prostate cancer line PC-3, which also robustly exhibited PGE2-induced mTORC1 activation, as identified from a screen in various cancer cell lines. Importantly, we showed a reinforcing interaction between PGE2 and IGF-1 on mTORC1 signaling, with an increase in IL-23 production as a cellular outcome. Our data reveal a previously unrecognized mechanism of PGE2-stimulated mTORC1 activation mediated by EP4/cAMP/PKA and EP1/Ca(2+) signaling, which may be of great importance in elucidating the promoting effects of obesity in pancreatic cancer. Ultimately, a precise understanding of these molecular links may provide novel targets for efficacious interventions devoid of adverse effects.
Collapse
Affiliation(s)
- Hui-Hua Chang
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Steven H Young
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - James Sinnett-Smith
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Caroline Ei Ne Chou
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Aune Moro
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Kathleen M Hertzer
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Oscar Joe Hines
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| |
Collapse
|
103
|
Wu GF, Zhang XL, Luo ZG, Yan JJ, Pan SH, Ying XR, Pan JG, Zhang GF. Metformin therapy and prostate cancer risk: a meta-analysis of observational studies. Int J Clin Exp Med 2015; 8:13089-13098. [PMID: 26550231 PMCID: PMC4612916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/21/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE Several observational studies have shown that metformin therapy may modify the risk of prostate cancer. We carried out a meta-analysis of relevant studies evaluating the effect of metformin therapy on prostate cancer risk. METHODS We searched pubmed database (January 1966-February 2014) for case-control and cohort studies that assessed metformin therapy and prostate cancer risk. Two authors independently assessed eligibility and extracted data. Summary RRs was calculated using fixed-effects model or random-effects model. Heterogeneity among studies was examined using Q and I(2) statistics. RESULTS We included six cohort studies and four case-control studies in the present meta-analysis, comprising 863,769 participants and 39,073 prostate cancer cases. The pooled RR of prostate cancer in relation to metformin therapy was 0.92 (95% CI: 0.84-1.02, P = 0.112). When we stratified the various studies by study type, we found that metformin therapy was associated with a significant reduced risk of prostate cancer among cohort studies (RR = 0.92, 95% CI [0.87, 0.96], P<0.001); however, no significant association was detected among case-control studies (RR = 0.95, 95% CI [0.78, 1.16], P = 0.632). There was also no indication of publication bias as suggested by Begg's test (P = 0.421) and Egger's test (P = 0.627). CONCLUSION Our findings indicate that metformin therapy is not significantly associated with lower prostate cancer risk.
Collapse
Affiliation(s)
- Gang-Feng Wu
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| | - Xiao-Long Zhang
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| | - Zhen-Gang Luo
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| | - Jia-Jun Yan
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| | - Shou-Hua Pan
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| | - Xiang-Rong Ying
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| | - Jian-Gang Pan
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| | - Guan-Fu Zhang
- Department of Urology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University Shaoxing, Zhejiang, China
| |
Collapse
|
104
|
Liu KL, Yu RJ, Feng GS, Wu J. Expression of insulin-like growth factor 1 in colorectal cancer: Relationship with angiogenesis. Shijie Huaren Xiaohua Zazhi 2015; 23:3384-3389. [DOI: 10.11569/wcjd.v23.i21.3384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the relationship between the expression of insulin-like growth factor 1 (IGF-1) and clinicopathological parameters, as well as tumor angiogenesis in colorectal cancer.
METHODS: The expression of IGF-1 was detected using immunohistochemical method in 56 colorectal carcinoma and 20 normal colon tissues. Microvessel density (MVD) was counted by evaluating the expression of endothelial marker CD34.
RESULTS: The positive rates of IGF-1 in colorectal carcinoma and normal mucosa were 85.71% and 35%, respectively. The expression of IGF-1 correlated with lymph node metastasis significantly (P < 0.05). MVD values were 8.76±2.67 and 35.55 ± 7.78 in normal colon tissue and colorectal cancer, respectively. MVD correlated significantly with differentiation degree, invasion depth, Duke's stage and lymph node metastasis (P < 0.05 for all).
CONCLUSION: IGF-1 is highly expressed in colorectal adenocarcinoma and may be involved in the progression of colorectal cancer through enhancing tumor angiogenesis.
Collapse
|
105
|
Cohen R, Neuzillet C, Tijeras-Raballand A, Faivre S, de Gramont A, Raymond E. Targeting cancer cell metabolism in pancreatic adenocarcinoma. Oncotarget 2015; 6:16832-47. [PMID: 26164081 PMCID: PMC4627277 DOI: 10.18632/oncotarget.4160] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 05/29/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second leading cause of cancer death by 2030. Current therapeutic options are limited, warranting an urgent need to explore innovative treatment strategies. Due to specific microenvironment constraints including an extensive desmoplastic stroma reaction, PDAC faces major metabolic challenges, principally hypoxia and nutrient deprivation. Their connection with oncogenic alterations such as KRAS mutations has brought metabolic reprogramming to the forefront of PDAC therapeutic research. The Warburg effect, glutamine addiction, and autophagy stand as the most important adaptive metabolic mechanisms of cancer cells themselves, however metabolic reprogramming is also an important feature of the tumor microenvironment, having a major impact on epigenetic reprogramming and tumor cell interactions with its complex stroma. We present a comprehensive overview of the main metabolic adaptations contributing to PDAC development and progression. A review of current and future therapies targeting this range of metabolic pathways is provided.
Collapse
Affiliation(s)
- Romain Cohen
- INSERM U728, Beaujon University Hospital (AP-HP – PRES Paris 7 Diderot), Clichy La Garenne, France
| | - Cindy Neuzillet
- INSERM U728, Beaujon University Hospital (AP-HP – PRES Paris 7 Diderot), Clichy La Garenne, France
- Department of Medical Oncology, Henri Mondor University Hospital, Créteil, France
| | | | - Sandrine Faivre
- Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Armand de Gramont
- New Drug Evaluation Laboratory, Centre of Experimental Therapeutics and Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Eric Raymond
- Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
106
|
Salvatore T, Marfella R, Rizzo MR, Sasso FC. Pancreatic cancer and diabetes: A two-way relationship in the perspective of diabetologist. Int J Surg 2015; 21 Suppl 1:S72-7. [PMID: 26123386 DOI: 10.1016/j.ijsu.2015.06.063] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 03/25/2015] [Accepted: 04/10/2015] [Indexed: 12/31/2022]
Abstract
Diabetes is a risk factor for pancreatic cancer as roughly half of all patients with pancreatic cancer are found to have diabetes at time of diagnosis. Moreover, an around 2-fold risk of pancreatic malignancy in diabetic patients has even be recently resulted from two meta-analysis. Actually, there is a bidirectional association between the two entities that implies a complex and reverse causality. In fact, while the risk for pancreatic cancer is modestly but significantly increased in patients with long-standing diabetes, recent-onset diabetes appears to be very frequently associated with pancreatic malignancy. Therefore, diabetes could serve as an excellent clue for early detection of pancreatic cancer. Moreover, recent epidemiological findings support the hypothesis that chronic exposure to hyperglycemia, higher insulin concentrations, and insulin resistance may be responsible for the enhanced risk of developing pancreatic cancer. Epidemiological data suggest that the type of anti-diabetic therapy may affect the risk of developing pancreatic cancer. In particular, metformin has been shown to reduce the risk of pancreatic cancer, as well as several other malignancies. On the other hand, some hypoglycemic agents could determine an increase of pancreatic cancer risk. These last findings were not confirmed. Finally, pancreatic cancer necessitates of a multidisciplinary management, primarily including surgeons and oncologists. In this context, the diabetologist plays an important role, given that his actions may influence the prevention and early diagnosis of pancreatic cancer, the perioperative complications associated to glycemic derangement, as well as the proper treatment of postpancreactomy diabetes.
Collapse
Affiliation(s)
- Teresa Salvatore
- Depart. of Internal and Experimental Medicine "Magrassi - Lanzara", Second University of Naples, Italy.
| | - Raffaele Marfella
- Depart. of Medicine, Surgery, Neurology, Metabolism and Geriatrics, Second University of Naples, Italy.
| | - Maria Rosaria Rizzo
- Depart. of Medicine, Surgery, Neurology, Metabolism and Geriatrics, Second University of Naples, Italy.
| | - Ferdinando Carlo Sasso
- Depart. of Internal and Experimental Medicine "Magrassi - Lanzara", Second University of Naples, Italy.
| |
Collapse
|
107
|
Matyszewski A, Czarnecka A, Kawecki M, Korzeń P, Safir IJ, Kukwa W, Szczylik C. Impaired glucose metabolism treatment and carcinogenesis. Oncol Lett 2015; 10:589-594. [PMID: 26622538 DOI: 10.3892/ol.2015.3324] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/29/2015] [Indexed: 12/29/2022] Open
Abstract
Carbohydrate metabolism disorders increase the risk of carcinogenesis. Diabetes mellitus alters numerous physiological processes that may encourage cancer growth. However, treating impaired glucose homeostasis may actually promote neoplasia; maintaining proper glucose plasma concentrations reduces metabolic stresses, however, certain medications may themselves result in oncogenic effects. A number of previous studies have demonstrated that metformin reduces the cancer risk. However, the use of sulfonylurea derivatives correlates with an increased risk of developing a malignancy. Another form of treatment, insulin therapy, involves using various forms of insulin that differ in pharmacodynamics, pharmacokinetics and efficacy. Previous studies have indicated that certain insulin variants also affect the cancer risk. The results from analyses that address the safety of long-lasting insulin types raise the most concern regarding the increased risk of malignancy. Rapid development of novel diabetic medications and their widespread use carries the risk of potentially increased rates of cancer, unnoticeable in limited, randomized, controlled trials. In the present review, the results of clinical and epidemiological studies are evaluated to assess the safety of anti-hyperglycemic medications and their effect on cancer risk and outcomes.
Collapse
Affiliation(s)
- Artur Matyszewski
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland ; Department of Internal Medicine, Cardiology and Hypertensionr, L. Rydygier Province Hospital, Suwałki, Poland ; Department of Nephrology and Dialysis Center, L. Rydygier Province Hospital, Suwałki, Poland
| | - Anna Czarnecka
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Maciej Kawecki
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Piotr Korzeń
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Ilan J Safir
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wojciech Kukwa
- Department of Otorhinolaryngology, Medical University of Warsaw, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
108
|
Zhang JW, Sun Q. Metformin May Improve the Prognosis of Patients with Pancreatic Cancer. Asian Pac J Cancer Prev 2015; 16:3937-40. [DOI: 10.7314/apjcp.2015.16.9.3937] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
109
|
Smith JP, Whitcomb DC, Matters GL, Brand RE, Liao J, Huang YJ, Frazier ML. Distribution of cholecystokinin-B receptor genotype between patients with pancreatic cancer and controls and its impact on survival. Pancreas 2015; 44:236-42. [PMID: 25469546 PMCID: PMC4326549 DOI: 10.1097/mpa.0000000000000263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Cholecystokinin (CCK) and gastrin stimulate growth of pancreatic cancer through the CCK-B receptor (CCK-BR). A splice variant of the CCK-BR that results from a single nucleotide polymorphism (SNP) has been identified. Because the splice variant receptor has an extended third intracellular loop, an area involved in cell signaling and growth, we hypothesized that this genetic variant could contribute to the poor prognosis and short survival of this malignancy. METHODS DNA from 931 patients with pancreatic cancer was evaluated for the SNP (C > A; rs1800843) in the CCK-BR gene. For statistical analysis, the Fisher exact test was used to compare the genotype and allele frequency between the cancer cohort and normal controls and the dependence of genotype on factors, such as stage of disease and age, was analyzed using Cox proportional hazards models. RESULTS Compared to the normal cohort, the frequency of the A-allele in pancreatic cancer subjects was increased (P = 0.01123; odds ratio, 2.283). Even after adjustment for stage of disease, survival of subjects with the minor allele was significantly shorter than those with the wild-genotype (hazard ratio, 1.83; P = 3.11 × 10(-11)). CONCLUSIONS The CCK-BR SNP predicts survival and should be studied as a candidate genetic biomarker for those at risk of pancreatic cancer.
Collapse
Affiliation(s)
- Jill P. Smith
- Dept. of Medicine, Georgetown University, Washington, DC, USA
- Dept. of Medicine, Pennsylvania State University, College of Medicine, Hershey, PA
| | - David C. Whitcomb
- Department of Medicine, University of Pittsburgh & UPMC, Pittsburgh, PA
| | - Gail L. Matters
- Dept. of Biochemistry & Molecular Biology, Pennsylvania State University, College of Medicine, Hershey, PA
| | - Randall E. Brand
- Department of Medicine, University of Pittsburgh & UPMC, Pittsburgh, PA
| | - Jiangang Liao
- Public Health Sciences, Pennsylvania State University, College of Medicine, Hershey, PA
| | - Yu-Jing Huang
- Department of Epidemiology, University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| | - Marsha L. Frazier
- Department of Epidemiology, University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
110
|
Soares HP, Ming M, Mellon M, Young SH, Han L, Sinnet-Smith J, Rozengurt E. Dual PI3K/mTOR Inhibitors Induce Rapid Overactivation of the MEK/ERK Pathway in Human Pancreatic Cancer Cells through Suppression of mTORC2. Mol Cancer Ther 2015; 14:1014-23. [PMID: 25673820 DOI: 10.1158/1535-7163.mct-14-0669] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 01/30/2015] [Indexed: 12/17/2022]
Abstract
The PI3K/AKT/mTOR pathway, which is aberrantly stimulated in many cancer cells, has emerged as a target for therapy. However, mTORC1/S6K also mediates negative feedback loops that attenuate upstream signaling. Suppression of these feedback loops opposes the growth-suppressive effects of mTOR inhibitors and leads to drug resistance. Here, we demonstrate that treatment of PANC-1 or MiaPaCa-2 pancreatic ductal adenocarcinoma (PDAC) cells with the dual PI3K/mTOR kinase inhibitor (PI3K/TOR-KI) BEZ235 blocked mTORC1/S6K activation (scored by S6 phosphorylation at Ser(240/244)), mTORC1/4E-BP1 (assayed by 4E-BP1 phosphorylation at Thr(37/46)), and mTORC2-mediated AKT phosphorylation at Ser(473), in a concentration-dependent manner. Strikingly, BEZ235 markedly enhanced the MEK/ERK pathway in a dose-dependent manner. Maximal ERK overactivation coincided with complete inhibition of phosphorylation of AKT and 4E-BP1. ERK overactivation was induced by other PI3K/TOR-KIs, including PKI-587 and GDC-0980. The MEK inhibitors U126 or PD0325901 prevented ERK overactivation induced by PI3K/TOR-KIs. The combination of BEZ235 and PD0325901 caused a more pronounced inhibition of cell growth than that produced by each inhibitor individually. Mechanistic studies assessing PI3K activity in single PDAC cells indicate that PI3K/TOR-KIs act through a PI3K-independent pathway. Doses of PI3K/TOR-KIs that enhanced MEK/ERK activation coincided with those that inhibited mTORC2-mediated AKT phosphorylation on Ser(473), suggesting a role of mTORC2. Knockdown of RICTOR via transfection of siRNA markedly attenuated the enhancing effect of BEZ235 on ERK phosphorylation. We propose that dual PI3K/mTOR inhibitors suppress a novel negative feedback loop mediated by mTORC2, thereby leading to enhanced MEK/ERK pathway activity in pancreatic cancer cells.
Collapse
Affiliation(s)
- Heloisa P Soares
- Division of Hematology-Oncology, David Geffen School of Medicine, Los Angeles, California
| | - Ming Ming
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Michelle Mellon
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Steven H Young
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California. CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, California
| | - Liang Han
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - James Sinnet-Smith
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California. CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, California
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California. CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, California. Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California.
| |
Collapse
|
111
|
Zhang Y, Li MX, Wang H, Zeng Z, Li XM. Metformin Down-regulates Endometrial Carcinoma Cell Secretion of IGF-1 and Expression of IGF-1R. Asian Pac J Cancer Prev 2015; 16:221-5. [DOI: 10.7314/apjcp.2015.16.1.221] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
112
|
Sun WY, Yun HY, Song YJ, Kim H, Lee OJ, Nam SJ, Koo JS. Insulin-like growth factor 1 receptor expression in breast cancer tissue and mammographic density. Mol Clin Oncol 2015; 3:572-580. [PMID: 26137269 DOI: 10.3892/mco.2015.497] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 01/16/2015] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to evaluate the association between insulin-like growth factor 1 receptor (IGF-1R) expression in breast cancer tissue and mammographic density and the clinical significance of IGF-1R overexpression. A total of 167 patients with primary invasive breast cancer were analyzed. Mammographic breast density and IGF-1R overexpression were correlated with clinicopathological parameters and analyzed by overall survival (OS) and disease-free survival (DFS). Increased breast tissue density was significantly associated with age, body mass index, menopausal status, histological grade and IGF-1R overexpression in the univariate analysis and with age (P=0.001), histological grade (P=0.045) and IGF-1R overexpression (P=0.021) in the multivariate analysis. IGF-1R overexpression was significantly associated with dense breast tissue in patients aged >40 years (P=0.002). IGF-1R overexpression in breast cancer in premenopausal women was associated with human epidermal growth factor receptor 2 (HER-2) positivity (P=0.016) and worse DFS (P=0.0414). There was no significant difference in OS and DFS between dense and non-dense breast tissue. IGF-1R expression in breast cancer tissue was significantly associated with mammographic breast tissue density in patients aged >40 years. It appears that IGF-1R expression in breast cancer tissue plays an important role in breast cancer in patients with dense breast tissue. In premenopausal women, IGF-1R overexpression in breast cancer tissue was significantly associated with HER-2 positivity and poor DFS.
Collapse
Affiliation(s)
- Woo-Young Sun
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Daejeon 301-723, Republic of Korea
| | - Hyo-Young Yun
- Department of Surgery, College of Medicine and Medical Research Institute, Cheongju, Chungcheong 361-763, Republic of Korea
| | - Young-Jin Song
- Department of Surgery, College of Medicine and Medical Research Institute, Cheongju, Chungcheong 361-763, Republic of Korea
| | - Heon Kim
- Department of Preventive Medicine, College of Medicine and Medical Research Institute, Cheongju, Chungcheong 361-763, Republic of Korea
| | - Ok-Jun Lee
- Department of Pathology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju, Chungcheong 361-763, Republic of Korea
| | - Seok-Jin Nam
- Department of Surgery, Samsung Medical Center, Seoul 135-710, Republic of Korea
| | - Ja-Seung Koo
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| |
Collapse
|
113
|
The Role of Nutraceuticals in Pancreatic Cancer Prevention and Therapy: Targeting Cellular Signaling, MicroRNAs, and Epigenome. Pancreas 2015; 44:1-10. [PMID: 25493373 PMCID: PMC4264839 DOI: 10.1097/mpa.0000000000000257] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer is one of the most aggressive malignancies in US adults. Experimental studies have found that antioxidant nutrients could reduce oxidative DNA damage, suggesting that these antioxidants may protect against pancreatic carcinogenesis. Several epidemiologic studies showed that dietary intake of antioxidants was inversely associated with the risk for pancreatic cancer, demonstrating the inhibitory effects of antioxidants on pancreatic carcinogenesis. Moreover, nutraceuticals, the anticancer agents from diet or natural plants, have been found to inhibit the development and progression of pancreatic cancer through the regulation of cellular signaling pathways. Importantly, nutraceuticals also up-regulate the expression of tumor-suppressive microRNAs (miRNAs) and down-regulate the expression of oncogenic miRNAs, leading to the inhibition of pancreatic cancer cell growth and pancreatic cancer stem cell self-renewal through modulation of cellular signaling network. Furthermore, nutraceuticals also regulate epigenetically deregulated DNAs and miRNAs, leading to the normalization of altered cellular signaling in pancreatic cancer cells. Therefore, nutraceuticals could have much broader use in the prevention and/or treatment of pancreatic cancer in combination with conventional chemotherapeutics. However, more in vitro mechanistic experiments, in vivo animal studies, and clinical trials are needed to realize the true value of nutraceuticals in the prevention and/or treatment of pancreatic cancer.
Collapse
|
114
|
De Marco P, Cirillo F, Vivacqua A, Malaguarnera R, Belfiore A, Maggiolini M. Novel Aspects Concerning the Functional Cross-Talk between the Insulin/IGF-I System and Estrogen Signaling in Cancer Cells. Front Endocrinol (Lausanne) 2015; 6:30. [PMID: 25798130 PMCID: PMC4351617 DOI: 10.3389/fendo.2015.00030] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022] Open
Abstract
The insulin/IGF system plays an important role in cancer progression. Accordingly, elevated levels of circulating insulin have been associated with an increased cancer risk as well as with aggressive and metastatic cancer phenotypes. Numerous studies have documented that estrogens cooperate with the insulin/IGF system in multiple pathophysiological conditions. The biological responses to estrogens are mainly mediated by the estrogen receptors (ER)α and ERβ, which act as transcription factors; however, several studies have recently demonstrated that a member of the G protein-coupled receptors, named GPR30/G-protein estrogen receptor (GPER), is also involved in the estrogen signaling in normal and malignant cells as well as in cancer-associated fibroblasts (CAFs). In this regard, novel mechanisms linking the action of estrogens through GPER with the insulin/IGF system have been recently demonstrated. This review recapitulates the relevant aspects of this functional cross-talk between the insulin/IGF and the estrogenic GPER transduction pathways, which occurs in various cell types and may account for cancer progression.
Collapse
Affiliation(s)
- Paola De Marco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Health, University Magna Graecia of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonino Belfiore, Università degli Studi Magna Graecia di Catanzaro, Viale Europa, Loc. Germaneto, Catanzaro 88100, Italy e-mail: ; Marcello Maggiolini, Università della Calabria, via P. Bucci, Rende 87036, Italy e-mail:
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- *Correspondence: Antonino Belfiore, Università degli Studi Magna Graecia di Catanzaro, Viale Europa, Loc. Germaneto, Catanzaro 88100, Italy e-mail: ; Marcello Maggiolini, Università della Calabria, via P. Bucci, Rende 87036, Italy e-mail:
| |
Collapse
|
115
|
Ming M, Sinnett-Smith J, Wang J, Soares HP, Young SH, Eibl G, Rozengurt E. Dose-Dependent AMPK-Dependent and Independent Mechanisms of Berberine and Metformin Inhibition of mTORC1, ERK, DNA Synthesis and Proliferation in Pancreatic Cancer Cells. PLoS One 2014; 9:e114573. [PMID: 25493642 PMCID: PMC4262417 DOI: 10.1371/journal.pone.0114573] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 11/11/2014] [Indexed: 12/13/2022] Open
Abstract
Natural products represent a rich reservoir of potential small chemical molecules exhibiting anti-proliferative and chemopreventive properties. Here, we show that treatment of pancreatic ductal adenocarcinoma (PDAC) cells (PANC-1, MiaPaCa-2) with the isoquinoline alkaloid berberine (0.3-6 µM) inhibited DNA synthesis and proliferation of these cells and delay the progression of their cell cycle in G1. Berberine treatment also reduced (by 70%) the growth of MiaPaCa-2 cell growth when implanted into the flanks of nu/nu mice. Mechanistic studies revealed that berberine decreased mitochondrial membrane potential and intracellular ATP levels and induced potent AMPK activation, as shown by phosphorylation of AMPK α subunit at Thr-172 and acetyl-CoA carboxylase (ACC) at Ser79. Furthermore, berberine dose-dependently inhibited mTORC1 (phosphorylation of S6K at Thr389 and S6 at Ser240/244) and ERK activation in PDAC cells stimulated by insulin and neurotensin or fetal bovine serum. Knockdown of α1 and α2 catalytic subunit expression of AMPK reversed the inhibitory effect produced by treatment with low concentrations of berberine on mTORC1, ERK and DNA synthesis in PDAC cells. However, at higher concentrations, berberine inhibited mitogenic signaling (mTORC1 and ERK) and DNA synthesis through an AMPK-independent mechanism. Similar results were obtained with metformin used at doses that induced either modest or pronounced reductions in intracellular ATP levels, which were virtually identical to the decreases in ATP levels obtained in response to berberine. We propose that berberine and metformin inhibit mitogenic signaling in PDAC cells through dose-dependent AMPK-dependent and independent pathways.
Collapse
Affiliation(s)
- Ming Ming
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - James Sinnett-Smith
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jia Wang
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Heloisa P. Soares
- Division of Hematology-Oncology, Department of Medicine David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Steven H. Young
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- CURE: Digestive Diseases Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
116
|
Krishan S, Richardson DR, Sahni S. Adenosine monophosphate-activated kinase and its key role in catabolism: structure, regulation, biological activity, and pharmacological activation. Mol Pharmacol 2014; 87:363-77. [PMID: 25422142 DOI: 10.1124/mol.114.095810] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor, which once activated, plays a role in several processes within the cell to restore energy homeostasis. The protein enhances catabolic pathways, such as β-oxidation and autophagy, to generate ATP, and inhibits anabolic processes that require energy, including fatty acid, cholesterol, and protein synthesis. Due to its key role in the regulation of critical cellular pathways, deregulation of AMPK is associated with the pathology of many diseases, including cancer, Wolff-Parkinson-White syndrome, neurodegenerative disorders, diabetes, and metabolic syndrome. In fact, AMPK is a target of some pharmacological agents implemented in the treatment of diabetes (metformin and thiazolidinediones) as well as other naturally derived products, such as berberine, which is used in traditional medicine. Due to its critical role in the cell and the pathology of several disorders, research into developing AMPK as a therapeutic target is becoming a burgeoning and exciting field of pharmacological research. A profound understanding of the regulation and activity of AMPK would enhance its development as a promising therapeutic target.
Collapse
Affiliation(s)
- Sukriti Krishan
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Sumit Sahni
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
117
|
Gong J, Robbins LA, Lugea A, Waldron RT, Jeon CY, Pandol SJ. Diabetes, pancreatic cancer, and metformin therapy. Front Physiol 2014; 5:426. [PMID: 25426078 PMCID: PMC4224068 DOI: 10.3389/fphys.2014.00426] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 10/14/2014] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer carries a poor prognosis as most patients present with advanced disease and preferred chemotherapy regimens offer only modest effects on survival. Risk factors include smoking, obesity, heavy alcohol, and chronic pancreatitis. Pancreatic cancer has a complex relationship with diabetes, as diabetes can be both a risk factor for pancreatic cancer and a result of pancreatic cancer. Insulin, insulin-like growth factor-1 (IGF-1), and certain hormones play an important role in promoting neoplasia in diabetics. Metformin appears to reduce risk for pancreatic cancer and improve survival in diabetics with pancreatic cancer primarily by decreasing insulin/IGF signaling, disrupting mitochondrial respiration, and inhibiting the mammalian target of rapamycin (mTOR) pathway. Other potential anti-tumorigenic effects of metformin include the ability to downregulate specificity protein transcription factors and associated genes, alter microRNAs, decrease cancer stem cell proliferation, and reduce DNA damage and inflammation. Here, we review the most recent knowledge on risk factors and treatment of pancreatic cancer and the relationship between diabetes, pancreatic cancer, and metformin as a potential therapy.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Medicine, Veterans Affairs Los Angeles, CA, USA
| | - Lori A Robbins
- Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Medicine, Veterans Affairs Los Angeles, CA, USA
| | - Aurelia Lugea
- Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Medicine, Veterans Affairs Los Angeles, CA, USA
| | - Richard T Waldron
- Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Medicine, Veterans Affairs Los Angeles, CA, USA
| | - Christie Y Jeon
- Cancer Prevention and Genetics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center Los Angeles, CA, USA ; Department of Medicine, Veterans Affairs Los Angeles, CA, USA ; Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles Los Angeles, CA, USA
| |
Collapse
|
118
|
Rozengurt E, Soares HP, Sinnet-Smith J. Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol Cancer Ther 2014; 13:2477-88. [PMID: 25323681 PMCID: PMC4222988 DOI: 10.1158/1535-7163.mct-14-0330] [Citation(s) in RCA: 219] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The development of drug resistance by cancer cells is recognized as a major cause for drug failure and disease progression. The PI3K/AKT/mTOR pathway is aberrantly stimulated in many cancer cells and thus it has emerged as a target for therapy. However, mTORC1 and S6K also mediate potent negative feedback loops that attenuate signaling via insulin/insulin growth factor receptor and other tyrosine kinase receptors. Suppression of these feedback loops causes overactivation of upstream pathways, including PI3K, AKT, and ERK that potentially oppose the antiproliferative effects of mTOR inhibitors and lead to drug resistance. A corollary of this concept is that release of negative feedback loops and consequent compensatory overactivation of promitogenic pathways in response to signal inhibitors can circumvent the mitogenic block imposed by targeting only one pathway. Consequently, the elucidation of the negative feedback loops that regulate the outputs of signaling networks has emerged as an area of fundamental importance for the rational design of effective anticancer combinations of inhibitors. Here, we review pathways that undergo compensatory overactivation in response to inhibitors that suppress feedback inhibition of upstream signaling and underscore the importance of unintended pathway activation in the development of drug resistance to clinically relevant inhibitors of mTOR, AKT, PI3K, or PI3K/mTOR.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California. CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, California. Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California.
| | - Heloisa P Soares
- Division of Hematology-Oncology and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - James Sinnet-Smith
- Division of Digestive Diseases and Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California. CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
119
|
Mancini P, Angeloni A, Risi E, Orsi E, Mezi S. Standard of care and promising new agents for triple negative metastatic breast cancer. Cancers (Basel) 2014; 6:2187-223. [PMID: 25347122 PMCID: PMC4276962 DOI: 10.3390/cancers6042187] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/05/2014] [Accepted: 09/26/2014] [Indexed: 12/11/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a cluster of heterogeneous diseases, all of them sharing the lack of expression of estrogen and progesterone receptors and HER2 protein. They are characterized by different biological, molecular and clinical features, including a poor prognosis despite the increased sensitivity to the current cytotoxic therapies. Several studies have identified important molecular features which enable further subdivision of this type of tumor. We are drawing from genomics, transcription and translation analysis at different levels, to improve our knowledge of the molecular alterations along the pathways which are activated during carcinogenesis and tumor progression. How this information should be used for the rational selection of therapy is an ongoing challenge and the subject of numerous research studies in progress. Currently, the vascular endothelial growth factor (VEGF), poly (ADP-ribose) polymerase (PARP), HSP90 and Aurora inhibitors are most used as targeting agents in metastatic setting clinical trials. In this paper we will review the current knowledge about the genetic subtypes of TNBC and their different responses to conventional therapeutic strategies, as well as to some new promising molecular target agents, aimed to achieve more tailored therapies.
Collapse
Affiliation(s)
- Patrizia Mancini
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Antonio Angeloni
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Emanuela Risi
- Department of Radiology, Oncology and Human Pathology, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Errico Orsi
- Department of Surgical Science, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| | - Silvia Mezi
- Department of Radiology, Oncology and Human Pathology, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy.
| |
Collapse
|
120
|
Nair V, Sreevalsan S, Basha R, Abdelrahim M, Abudayyeh A, Rodrigues Hoffman A, Safe S. Mechanism of metformin-dependent inhibition of mammalian target of rapamycin (mTOR) and Ras activity in pancreatic cancer: role of specificity protein (Sp) transcription factors. J Biol Chem 2014; 289:27692-701. [PMID: 25143389 PMCID: PMC4183806 DOI: 10.1074/jbc.m114.592576] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/07/2014] [Indexed: 12/21/2022] Open
Abstract
The antidiabetic drug metformin exhibits both chemopreventive and chemotherapeutic activity for multiple cancers including pancreatic cancer; however, the underlying mechanism of action of metformin is unclear. A recent study showed that metformin down-regulated specificity protein (Sp) transcription factors (TFs) Sp1, Sp3, and Sp4 in pancreatic cancer cells and tumors, and this was accompanied by down-regulation of several pro-oncogenic Sp-regulated genes. Treatment with metformin or down-regulation of Sp TFs by RNAi also inhibits two major pro-oncogenic pathways in pancreatic cancer cells, namely mammalian target of rapamycin (mTOR) signaling and epidermal growth factor (EGFR)-dependent activation of Ras. Metformin and Sp knockdown by RNAi decreased expression of the insulin-like growth factor-1 receptor (IGF-1R), resulting in inhibition of mTOR signaling. Ras activity was also decreased by metformin and Sp knockdown of EGFR, another Sp-regulated gene. Thus, the antineoplastic activities of metformin in pancreatic cancer are due, in part, to down-regulation of Sp TFs and Sp-regulated IGF-1R and EGFR, which in turn results in inhibition of mTOR and Ras signaling, respectively.
Collapse
Affiliation(s)
| | | | - Riyaz Basha
- the Cancer Research Institute, M.D. Anderson Cancer Center-Orlando, Orlando, Florida 32806, the Baylor College of Medicine, Houston, Texas 77030
| | - Maen Abdelrahim
- the Cancer Research Institute, M.D. Anderson Cancer Center-Orlando, Orlando, Florida 32806, the Baylor College of Medicine, Houston, Texas 77030
| | - Ala Abudayyeh
- the Department of Emergency Medicine, M.D. Anderson Cancer Center, Houston, Texas 77030, and
| | | | - Stephen Safe
- From the Departments of Veterinary Physiology and Pharmacology and the Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, Texas 77030
| |
Collapse
|
121
|
Rozengurt E. Mechanistic target of rapamycin (mTOR): a point of convergence in the action of insulin/IGF-1 and G protein-coupled receptor agonists in pancreatic cancer cells. Front Physiol 2014; 5:357. [PMID: 25295009 PMCID: PMC4171984 DOI: 10.3389/fphys.2014.00357] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/02/2014] [Indexed: 12/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common form of pancreatic cancer, is one of the most lethal human diseases. PDAC is now the fourth leading cause of cancer mortality in both men and women and deaths due to PDAC are projected to increase dramatically. Novel targets and agents for chemoprevention are urgently needed and will most likely arise from a more detailed understanding of the signaling mechanisms that stimulate the promotion and progression of sub-malignant cells into pancreatic cancer cells and from the identification of modifiable risk factors for PDAC. Many epidemiological studies have linked obesity and long-standing type 2 diabetes mellitus (T2DM) with increased risk and worse clinical outcomes for developing PDAC. These diet-related metabolic disorders are multifaceted but characterized by peripheral insulin resistance, compensatory overproduction of insulin and increased bioavailability of insulin-like growth factor-1 (IGF-1). Mounting evidence indicates that the insulin/IGF-1 receptor system plays a critical role in PDAC development and multiple studies support the notion that crosstalk between the insulin receptor and heptahelical G protein-coupled receptor (GPCR) signaling systems is an important element in the biological responses elicited by these signaling systems, including cell proliferation. This article highlights the central role of the mechanistic target of rapamycin (mTOR) in mediating crosstalk between insulin/IGF-1 and GPCR signaling in pancreatic cancer cells and proposes strategies, including the use of metformin, to target this signaling system in PDAC cells.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine, Molecular Biology Institute, University of California at Los Angeles Los Angeles, CA, USA
| |
Collapse
|
122
|
Jose P, Sundar K, Anjali CH, Ravindran A. Metformin-Loaded BSA Nanoparticles in Cancer Therapy: A New Perspective for an Old Antidiabetic Drug. Cell Biochem Biophys 2014; 71:627-36. [DOI: 10.1007/s12013-014-0242-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
123
|
Chiu JW, Wong H, Leung R, Pang R, Cheung TT, Fan ST, Poon R, Yau T. Advanced pancreatic cancer: flourishing novel approaches in the era of biological therapy. Oncologist 2014; 19:937-50. [PMID: 25117068 PMCID: PMC4153449 DOI: 10.1634/theoncologist.2012-0131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/06/2014] [Indexed: 12/13/2022] Open
Abstract
The progress in the development of systemic treatment for advanced pancreatic cancer (APC) has been slow. The mainstream treatment remains using chemotherapy including gemcitabine, FOLFIRINOX, and nab-paclitaxel. Erlotinib is the only approved biological therapy with marginal benefit. Studies of agents targeting epidermal growth factor receptor, angiogenesis, and RAS signaling have not been satisfying, and the usefulness of targeted therapy in APC is uncertain. Understanding in molecular processes and tumor biology has opened the door for new treatment strategies such as targeting insulin-like growth factor 1 receptor, transforming growth factor β, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin pathway, and Notch pathway. New directions also include the upcoming immunotherapy and many novel agents that act on the microenvironment. The practice of personalized medicine using predictive biomarkers and pharmacogenomics signatures may also enhance the effectiveness of existing treatment. Future treatment approaches may involve comprehensive genomic assessment of tumor and integrated combinations of multiple agents to overcome treatment resistance.
Collapse
Affiliation(s)
- Joanne W Chiu
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Hilda Wong
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Roland Leung
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Roberta Pang
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Tan-To Cheung
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Sheung-Tat Fan
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Ronnie Poon
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| | - Thomas Yau
- University Departments of Medicine and Surgery, Queen Mary Hospital, Hong Kong, People's Republic of China; Centre for Cancer Research, University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
124
|
In Vitro and In Vivo Biological Evaluation of O-Carboxymethyl Chitosan Encapsulated Metformin Nanoparticles for Pancreatic Cancer Therapy. Pharm Res 2014; 31:3361-70. [DOI: 10.1007/s11095-014-1425-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/12/2014] [Indexed: 02/08/2023]
|
125
|
Al Haddad AHI, Adrian TE. Challenges and future directions in therapeutics for pancreatic ductal adenocarcinoma. Expert Opin Investig Drugs 2014; 23:1499-515. [PMID: 25078674 DOI: 10.1517/13543784.2014.933206] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death in the USA. The 5-year survival of < 5% has not changed in decades. In contrast to other major cancers, the incidence of PDAC is increasing. AREAS COVERED The aims of this paper are first to analyze why PDAC is so difficult to treat and, second, to suggest future directions for PDAC therapeutics. The authors provide an article that is based on a comprehensive search through MEDLINE and the clinicalTrials.gov website. EXPERT OPINION Progress has been made recently. Notably, FOLFIRINOX or nab-paclitaxel plus gemcitabine provide survival benefit over gemcitabine alone, which was previously the mainstay of therapy for PDAC. Most of the current trials are testing combinations of repurposed drugs rather than addressing key targets in the PDAC pathogenesis. It is clear that to really make an impact on this disease, it will be necessary to address three different problems with targeted therapeutics. First, it is important to eradicate PDAC stem cells that result in recurrence. Second, it is important to reduce the peritumoral stroma that provides the tumors with growth support and provides a barrier to access of therapeutic agents. Finally, it is important to address the marked cachexia and metabolic derangement that contribute to morbidity and mortality and further complicate therapeutic intervention.
Collapse
Affiliation(s)
- Amal H I Al Haddad
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University , PO Box 17666, Al Ain , UAE
| | | |
Collapse
|
126
|
Duan L, Ying G, Danzer B, Perez RE, Shariat-Madar Z, Levenson VV, Maki CG. The prolyl peptidases PRCP/PREP regulate IRS-1 stability critical for rapamycin-induced feedback activation of PI3K and AKT. J Biol Chem 2014; 289:21694-705. [PMID: 24936056 DOI: 10.1074/jbc.m114.550038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/AKT)/mammalian target of rapamycin (mTOR) pathway conveys signals from receptor tyrosine kinases (RTKs) to regulate cell metabolism, proliferation, survival, and motility. Previously we found that prolylcarboxypeptidase (PRCP) regulate proliferation and survival in breast cancer cells. In this study, we found that PRCP and the related family member prolylendopeptidase (PREP) are essential for proliferation and survival of pancreatic cancer cells. Depletion/inhibition of PRCP and PREP-induced serine phosphorylation and degradation of IRS-1, leading to inactivation of the cellular PI3K and AKT. Notably, depletion/inhibition of PRCP/PREP destabilized IRS-1 in the cells treated with rapamycin, blocking the feedback activation PI3K/AKT. Consequently, inhibition of PRCP/PREP enhanced rapamycin-induced cytotoxicity. Thus, we have identified PRCP and PREP as a stabilizer of IRS-1 which is critical for PI3K/AKT/mTOR signaling in pancreatic cancer cells.
Collapse
Affiliation(s)
- Lei Duan
- From the Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois 60612,
| | - Guoguang Ying
- the Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Brian Danzer
- From the Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois 60612
| | - Ricardo E Perez
- From the Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois 60612
| | - Zia Shariat-Madar
- the Department of Pharmacology, University of Mississippi, University, Mississippi 38677, and
| | | | - Carl G Maki
- From the Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, Illinois 60612,
| |
Collapse
|
127
|
Fang Y, French J, Zhao H, Benkovic S. G-protein-coupled receptor regulation of de novo purine biosynthesis: a novel druggable mechanism. Biotechnol Genet Eng Rev 2014; 29:31-48. [PMID: 24568251 DOI: 10.1080/02648725.2013.801237] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Spatial organization of metabolic enzymes may represent a general cellular mechanism to regulate metabolic flux. One recent example of this type of cellular phenomenon is the purinosome, a newly discovered multi-enzyme metabolic assembly that includes all of the enzymes within the de novo purine biosynthetic pathway. Our understanding of the components and regulation of purinosomes has significantly grown in recent years. This paper reviews the purine de novo biosynthesis pathway and its regulation, and presents the evidence supporting the purinosome assembly and disassembly processes under the control of G-protein-coupled receptor (GPCR) signaling. This paper also discusses the implications of purinosome and GPCR regulation in drug discovery.
Collapse
Affiliation(s)
- Ye Fang
- a Biochemical Technologies, Science and Technology Division , Corning Incorporated , Corning , New York , USA
| | | | | | | |
Collapse
|
128
|
Li C, Kong D. Cancer risks from diabetes therapies: evaluating the evidence. Pharmacol Ther 2014; 144:71-81. [PMID: 24844968 DOI: 10.1016/j.pharmthera.2014.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/05/2014] [Indexed: 12/25/2022]
Abstract
Epidemiological studies have identified positive associations between diabetes, obesity and cancer. Insulin, metformin and thiazolidinediones (TDZs) are among the major diabetes therapies that improve glycaemic control by acting via molecular targets including the insulin receptor and insulin-like growth factor pathways, adenosine monophosphate-activated kinase and peroxisome proliferator-activated receptor γ. It is well-established that clinical application of insulin and TDZs is associated with weight gain, but the potential of these therapies to promote tumourigenesis is less well-studied. In addition, although anti-tumour properties of metformin have been proposed, recently published data do not support a protective effect of metformin against cancer in diabetic patients. Given that diabetes and cancer each account for 8% and 13% of global deaths and there is a substantial financial burden incurred by both disorders, developing diabetes therapies that are safe, efficacious and cost-effective has never been more desirable. This timely review examines recent progress in delineating the molecular mechanisms responsible for the anti-diabetic actions of insulin, metformin and TZDs and considers evidence implicating these therapies in cell transformation and tumourigenesis. In addition, since the endocannabinoid signalling system (ECS) is now considered a therapeutic target and biomarker candidate for hyperglycaemia, obesity and cell growth, a brief section covering recent scientific advances regarding the ECS, particularly its functions in regulating glucose metabolism and cell survival, is also included in this review.
Collapse
Affiliation(s)
- Chen Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Science, China.
| | - Deling Kong
- Institute of Biomedical Engineering, Chinese Academy of Medical Science, China
| |
Collapse
|
129
|
Diabetes and pancreatic cancer survival: a prospective cohort-based study. Br J Cancer 2014; 111:181-5. [PMID: 24786605 PMCID: PMC4090724 DOI: 10.1038/bjc.2014.224] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 04/01/2014] [Accepted: 04/04/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Diabetes is a risk factor for pancreatic cancer but its association with survival from pancreatic cancer is poorly understood. Our objective was to investigate the association of diabetes with survival among pancreatic cancer patients in a prospective cohort-based study where diabetes history was ascertained before pancreatic cancer diagnosis. METHODS We evaluated survival by baseline (1993-2001) self-reported diabetes history (n=62) among 504 participants that developed exocrine pancreatic cancer within the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. Hazard ratios (HRs) and 95% confidence intervals (CIs) for mortality were estimated using Cox proportional hazards model, adjusted for age, sex, body mass index, race, smoking, and tumour stage (local, locally advanced, and metastatic). RESULTS The multivariable-adjusted HR for mortality comparing participants with diabetes to those without was 1.52 (95% CI=1.14-2.04, P-value <0.01). After excluding those diagnosed with pancreatic cancer within 3 years of study enrolment, HR for mortality among those with diabetes was 1.45 (95% CI=1.06-2.00, P-value=0.02). CONCLUSIONS Using prospectively collected data, our findings indicate that diabetes is associated with worse survival among patients with pancreatic cancer.
Collapse
|
130
|
McCarty MF. AMPK activation--protean potential for boosting healthspan. AGE (DORDRECHT, NETHERLANDS) 2014; 36:641-663. [PMID: 24248330 PMCID: PMC4039279 DOI: 10.1007/s11357-013-9595-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/22/2013] [Indexed: 06/01/2023]
Abstract
AMP-activated kinase (AMPK) is activated when the cellular (AMP+ADP)/ATP ratio rises; it therefore serves as a detector of cellular "fuel deficiency." AMPK activation is suspected to mediate some of the health-protective effects of long-term calorie restriction. Several drugs and nutraceuticals which slightly and safely impede the efficiency of mitochondrial ATP generation-most notably metformin and berberine-can be employed as clinical AMPK activators and, hence, may have potential as calorie restriction mimetics for extending healthspan. Indeed, current evidence indicates that AMPK activators may reduce risk for atherosclerosis, heart attack, and stroke; help to prevent ventricular hypertrophy and manage congestive failure; ameliorate metabolic syndrome, reduce risk for type 2 diabetes, and aid glycemic control in diabetics; reduce risk for weight gain; decrease risk for a number of common cancers while improving prognosis in cancer therapy; decrease risk for dementia and possibly other neurodegenerative disorders; help to preserve the proper structure of bone and cartilage; and possibly aid in the prevention and control of autoimmunity. While metformin and berberine appear to have the greatest utility as clinical AMPK activators-as reflected by their efficacy in diabetes management-regular ingestion of vinegar, as well as moderate alcohol consumption, may also achieve a modest degree of health-protective AMPK activation. The activation of AMPK achievable with any of these measures may be potentiated by clinical doses of the drug salicylate, which can bind to AMPK and activate it allosterically.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity, 7831 Rush Rose Dr., Apt. 316, Carlsbad, CA, 92009, USA,
| |
Collapse
|
131
|
Schutte AE, Volpe M, Tocci G, Conti E. Revisiting the relationship between blood pressure and insulin-like growth factor-1. Hypertension 2014; 63:1070-7. [PMID: 24566078 DOI: 10.1161/hypertensionaha.113.03057] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Conflicting evidence exists on the relationship between blood pressure (BP) and insulin-like growth factor-1 (IGF-1). We reviewed available articles and pooled extrapolated regression coefficients for the association between BP and total IGF-1 as reported in the literature and included additional data from 912 individuals from the general population. We identified 20 studies including 11 704 subjects. We also measured total IGF-1, insulin-like binding protein-3, and BP in 912 black and white men and women from South Africa (aged 20-70 years). When plotting positive and negative weighed regression coefficients (29 data points) against IGF-1, we found a significant positive relationship (r=0.31; P<0.001; n=11 704) intercepting the 0 point at 191 ng/mL IGF-1, suggesting an inverse BP/IGF-1 relationship in low IGF-1 conditions, and a positive relationship in overtly high IGF-1 conditions. In conclusion, our findings suggest that the relationship between BP and IGF-1 is dependent on, or related to, IGF-1 concentrations, as an expression of direct or reverse causality. Low IGF-1 bioavailability (associated with aging and vascular deterioration), resistance to IGF-1, and the complex interplay between IGF-1 and other vasoactive hormones could mask the vasoprotective functions of IGF-1 in cross-sectional studies or could modify their functions in prospective studies.
Collapse
Affiliation(s)
- Aletta Elisabeth Schutte
- Hypertension in Africa Research Team, North-West University, Hoffman St, Private Bag X6001, Potchefstroom 2520, South Africa.
| | | | | | | |
Collapse
|
132
|
Leone A, Di Gennaro E, Bruzzese F, Avallone A, Budillon A. New perspective for an old antidiabetic drug: metformin as anticancer agent. Cancer Treat Res 2014; 159:355-376. [PMID: 24114491 DOI: 10.1007/978-3-642-38007-5_21] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Metformin, an inexpensive, well-tolerated oral agent that is commonly used in the first-line treatment for type 2 diabetes, has become the focus of intense research as a potential anticancer agent. This research reflects a convergence of epidemiologic, clinical, and preclinical evidence, suggesting that metformin may lower cancer risk in diabetics and improve outcomes of many common cancers. Notably, metformin mediates an approximately 30 % reduction in the lifetime risk of cancer in diabetic patients. There is growing recognition that metformin may act (1) directly on cancer cells, primarily by impacting mitochondrial respiration leading to the activation of the AMP-activated protein kinase (AMPK), which controls energy homeostasis in cells, but also through other mechanisms or (2) indirectly on the host metabolism, largely through AMPK-mediated reduction in hepatic gluconeogenesis, leading to reduced circulating insulin levels and decreased insulin/IGF-1 receptor-mediated activation of the PI3K pathway. Support for this comes from the observation that metformin inhibits cancer cell growth in vitro and delays the onset of tobacco carcinogen-induced lung cancer in mice and that metformin and its analog phenformin delay spontaneous tumor development cancer-prone transgenic mice. The potential for both direct antitumor effects and indirect host-mediated effects has sparked enormous interest, but has led to added challenges in translating preclinical findings to the clinical setting. Nonetheless, the accumulation of evidence has been sufficient to justify initiation of clinical trials of metformin as an anticancer agent in the clinical setting, including a large-scale adjuvant study in breast cancer, with additional studies planned.
Collapse
Affiliation(s)
- Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori Fondazioni Giovanni Pascale - IRCCS, 80131, Naples, Italy
| | | | | | | | | |
Collapse
|
133
|
Garofalo C, Capristo M, Manara MC, Mancarella C, Landuzzi L, Belfiore A, Lollini PL, Picci P, Scotlandi K. Metformin as an adjuvant drug against pediatric sarcomas: hypoxia limits therapeutic effects of the drug. PLoS One 2013; 8:e83832. [PMID: 24391834 PMCID: PMC3877110 DOI: 10.1371/journal.pone.0083832] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/08/2013] [Indexed: 01/27/2023] Open
Abstract
Metformin, a well-known insulin-sensitizer commonly used for type 2 diabetes therapy, has recently emerged as potentially very attractive drug also in oncology. It is cheap, it is relatively safe and many reports have indicated effects in cancer prevention and therapy. These desirable features are particularly interesting for pediatric sarcomas, a group of rare tumors that have been shown to be dependent on IGF and insulin system for pathogenesis and progression. Metformin exerts anti-mitogenic activity in several cancer histotypes through several molecular mechanisms. In this paper, we analyzed its effects against osteosarcoma, Ewing sarcoma and rhabdomyosarcoma, the three most common pediatric sarcomas. Despite in vitro metformin gave remarkable antiproliferative and chemosensitizing effects both in sensitive and chemoresistant cells, its efficacy was not confirmed against Ewing sarcoma xenografts neither as single agent nor in combination with vincristine. This discrepancy between in vitro and in vivo effects may be due to hypoxia, a common feature of solid tumors. We provide evidences that in hypoxia conditions metformin was not able to activate AMPK and inhibit mTOR signaling, which likely prevents the inhibitory effects of metformin on tumor growth. Thus, although metformin may be considered a useful complement of conventional chemotherapy in normoxia, its therapeutic value in highly hypoxic tumors may be more limited. The impact of hypoxia should be considered when novel therapies are planned for pediatric sarcomas.
Collapse
Affiliation(s)
- Cecilia Garofalo
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Mariantonietta Capristo
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Maria Cristina Manara
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Caterina Mancarella
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Lorena Landuzzi
- Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | | | - Pier-Luigi Lollini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Piero Picci
- Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Katia Scotlandi
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
- * E-mail:
| |
Collapse
|
134
|
Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. ACTA ACUST UNITED AC 2013; Chapter 14:Unit 14.25. [PMID: 23744710 DOI: 10.1002/0471141755.ph1425s61] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The identification of small subpopulations of cancer stem cells (CSCs) from blood mononuclear cells in human acute myeloid leukemia (AML) in 1997 was a landmark observation that recognized the potential role of CSCs in tumor aggressiveness. Two critical properties contribute to the functional role of CSCs in the establishment and recurrence of cancerous tumors: their capacity for self-renewal and their potential to differentiate into unlimited heterogeneous populations of cancer cells. These findings suggest that CSCs may represent novel therapeutic targets for the treatment and/or prevention of tumor progression, since they appear to be involved in cell migration, invasion, metastasis, and treatment resistance-all of which lead to poor clinical outcomes. The identification of CSC-specific markers, the isolation and characterization of CSCs from malignant tissues, and targeting strategies for the destruction of CSCs provide a novel opportunity for cancer research. This overview describes the potential implications of several common CSC markers in the identification of CSC subpopulations that are restricted to common malignant diseases, e.g., leukemia, and breast, prostate, pancreatic, and lung cancers. The role of microRNAs (miRNAs) in the regulation of CSC function is also discussed, as are several methods commonly used in CSC research. The potential role of the antidiabetic drug metformin- which has been shown to have effects on CSCs, and is known to function as an antitumor agent-is discussed as an example of this new class of chemotherapeutics.
Collapse
Affiliation(s)
- Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
135
|
Michel G, Matthes HWD, Hachet-Haas M, El Baghdadi K, de Mey J, Pepperkok R, Simpson JC, Galzi JL, Lecat S. Plasma membrane translocation of REDD1 governed by GPCRs contributes to mTORC1 activation. J Cell Sci 2013; 127:773-87. [PMID: 24338366 DOI: 10.1242/jcs.136432] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mTORC1 kinase promotes cell growth in response to growth factors by activation of receptor tyrosine kinase. It is regulated by the cellular energy level and the availability of nutrients. mTORC1 activity is also inhibited by cellular stresses through overexpression of REDD1 (regulated in development and DNA damage responses). We report the identification of REDD1 in a fluorescent live-imaging screen aimed at discovering new proteins implicated in G-protein-coupled receptor signaling, based on translocation criteria. Using a sensitive and quantitative plasma membrane localization assay based on bioluminescent resonance energy transfer, we further show that a panel of endogenously expressed GPCRs, through a Ca(2+)/calmodulin pathway, triggers plasma membrane translocation of REDD1 but not of its homolog REDD2. REDD1 and REDD2 share a conserved mTORC1-inhibitory motif characterized at the functional and structural level and differ most in their N-termini. We show that the N-terminus of REDD1 and its mTORC1-inhibitory motif participate in the GPCR-evoked dynamic interaction of REDD1 with the plasma membrane. We further identify REDD1 as a novel effector in GPCR signaling. We show that fast activation of mTORC1 by GPCRs correlates with fast and maximal translocation of REDD1 to the plasma membrane. Overexpression of functional REDD1 leads to a reduction of mTORC1 activation by GPCRs. By contrast, depletion of endogenous REDD1 protein unleashes mTORC1 activity. Thus, translocation to the plasma membrane appears to be an inactivation mechanism of REDD1 by GPCRs, which probably act by sequestering its functional mTORC1-inhibitory motif that is necessary for plasma membrane targeting.
Collapse
Affiliation(s)
- Grégory Michel
- GPCRs, Pain and Inflammation Team, UMR7242, CNRS-University of Strasbourg, LabEx Medalis, 67412 Illkirch, France
| | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Mohammed A, Janakiram NB, Brewer M, Ritchie RL, Marya A, Lightfoot S, Steele VE, Rao CV. Antidiabetic Drug Metformin Prevents Progression of Pancreatic Cancer by Targeting in Part Cancer Stem Cells and mTOR Signaling. Transl Oncol 2013; 6:649-59. [PMID: 24466367 PMCID: PMC3890699 DOI: 10.1593/tlo.13556] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 12/12/2022] Open
Abstract
Epidemiologic studies have shown that diabetes mellitus is associated positively with increased risk of pancreatic ductal adenocarcinoma (PDAC), and recent meta-analysis studies showed that metformin, reduces the risk of pancreatic cancer (PC). We tested the effects of metformin on pancreatic intraepithelial neoplasia (PanIN) and their progression to PDAC in p48Cre/+.LSL-KrasG12D/+ transgenic mice. Mice fed control diet showed 80% and 62% incidence of PDAC in males and females, respectively. Male mice showed 20% and 26%, and female mice showed 7% and 0% PDAC incidence with 1000- and 2000-ppm metformin treatments, respectively. Both doses of metformin decreased pancreatic tumor weights by 34% to 49% (P < 0.03-0.001). The drug treatment caused suppression of PanIN 3 (carcinoma in situ) lesions by 28% to 39% (P < .002) and significant inhibition of carcinoma spread in the pancreas. The pancreatic tissue and/or serum of mice fed metformin showed a significant inhibition of mammalian target of rapamycin (mTOR), extracellular signal-regulated kinases (ERK), phosphorylated extracellular signal-regulated kinases (pErk), and insulin-like growth factor 1 (IGF-1) with an increase in phosphorylated 5' adenosine monophosphate kinase (pAMPK), tuberous sclerosis complex 1 (TSC1, TSC2), C-protein and an autophagy related protein 2 (ATG2). The cancer stem cell (CSC) markers were significantly decreased (P < 0.04-0.0002) in the pancreatic tissue. These results suggest that biologic effects of metformin are mediated through decreased CSC markers cluster of differentiation 44 (CD44 and CD133), aldehyde dehydrogenase isoform 1 (ALDH1), and epithelial cell adhesion molecule (EPCAM) and modulation of the mTOR signaling pathway. Our preclinical data indicate that metformin has significant potential for use in clinical trials for PC chemoprevention.
Collapse
Affiliation(s)
- Altaf Mohammed
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Naveena B Janakiram
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Misty Brewer
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Rebekah L Ritchie
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Anuj Marya
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Stan Lightfoot
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Vernon E Steele
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology-Oncology Section, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
137
|
Heng BC, Aubel D, Fussenegger M. An overview of the diverse roles of G-protein coupled receptors (GPCRs) in the pathophysiology of various human diseases. Biotechnol Adv 2013; 31:1676-94. [DOI: 10.1016/j.biotechadv.2013.08.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 12/23/2022]
|
138
|
Ahn J, Lee JK. [New-onset diabetes as an early sign of pancreatic cancer]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2013; 62:263-6. [PMID: 24262590 DOI: 10.4166/kjg.2013.62.5.263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
While long-standing diabetes is a risk factor of pancreatic cancer, new-onset diabetes could be a consequence of underlying pancreatic malignancy. About 30% to 50% of pancreatic cancer patients have new-onset diabetes. Because diabetes develops in preclinical or early stages of pancreatic cancer, it could serve as an excellent clue for early detection of pancreatic cancer. Insulin resistance associated with hyperglycemia and hyperinsulinemia by diabetogenic factors secreted from cancer cells have been suggested to be a possible mechanism of pancreatic cancer-induced diabetes. It is difficult to differentiate pancreatic cancer-induced diabetes from the more common type 2 diabetes. Although several clinical features and potential biomarkers have been investigated, optimal strategies and modalities to screen pancreatic cancer among the new-onset diabetes have not yet been fully determined.
Collapse
Affiliation(s)
- Jemma Ahn
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | |
Collapse
|
139
|
Kappe C, Zhang Q, Holst JJ, Nyström T, Sjöholm Å. Evidence for paracrine/autocrine regulation of GLP-1-producing cells. Am J Physiol Cell Physiol 2013; 305:C1041-9. [DOI: 10.1152/ajpcell.00227.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Glucagon-like peptide-1 (GLP-1), secreted from gut L cells upon nutrient intake, forms the basis for novel drugs against type 2 diabetes (T2D). Secretion of GLP-1 has been suggested to be impaired in T2D and in conditions associated with hyperlipidemia and insulin resistance. Further, recent studies support lipotoxicity of GLP-1-producing cells in vitro. However, little is known about the regulation of L-cell viability/function, the effects of insulin signaling, or the potential effects of stable GLP-1 analogs and dipeptidyl peptidase-4 (DPP-4) inhibitors. We determined effects of insulin as well as possible autocrine action of GLP-1 on viability/apoptosis of GLP-1-secreting cells in the presence/absence of palmitate, while also assessing direct effects on function. The studies were performed using the GLP-1-secreting cell line GLUTag, and palmitate was used to simulate hyperlipidemia. Our results show that palmitate induced production of reactive oxygen species and caspase-3 activity and reduced cell viability are significantly attenuated by preincubation with insulin/exendin-4. The indicated lipoprotective effect of insulin/exendin-4 was not detectable in the presence of the GLP-1 receptor (GLP-1R) antagonist exendin (9–39) and attenuated in response to pharmacological inhibition of exchange protein activated by cAMP (Epac) signaling, while protein kinase A inhibition had no significant effect. Insulin/exendin-4 also significantly stimulate acute and long-term GLP-1 secretion in the presence of glucose, suggesting novel beneficial effects of insulin signaling and GLP-1R activation on glycemia through enhanced mass of GLP-1-producing cells and enhanced GLP-1 secretion. In addition, the effects of insulin indicate that not only is GLP-1 important for insulin secretion but altered insulin signaling may contribute to an altered GLP-1 secretion.
Collapse
Affiliation(s)
- Camilla Kappe
- Karolinska Institutet, Department of Clinical Science and Education, Unit for Diabetes Research, Södersjukhuset, Stockholm, Sweden
| | - Qimin Zhang
- Karolinska Institutet, Department of Clinical Science and Education, Unit for Diabetes Research, Södersjukhuset, Stockholm, Sweden
| | - Jens J. Holst
- Department of Biomedical Sciences, Panum Institute, Copenhagen, Denmark; and
| | - Thomas Nyström
- Karolinska Institutet, Department of Clinical Science and Education, Unit for Diabetes Research, Södersjukhuset, Stockholm, Sweden
| | - Åke Sjöholm
- Karolinska Institutet, Department of Clinical Science and Education, Unit for Diabetes Research, Södersjukhuset, Stockholm, Sweden
- Department of Biochemistry and Molecular Biology, University of South Alabama, College of Medicine, Mobile, Alabama
| |
Collapse
|
140
|
Lee DJ, Kim B, Lee JH, Park SJ, Hong SP, Cheon JH, Kim TI, Kim WH. [The effect of metformin on responses to chemotherapy and survival in stage IV colorectal cancer with diabetes]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2013; 60:355-61. [PMID: 23242018 DOI: 10.4166/kjg.2012.60.6.355] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND/AIMS Metformin is known to lower the risk of cancer and cancer mortality. However, the effect of metformin in stage IV colorectal cancer (CRC) patients with diabetes mellitus (DM) remains unknown. The aim of this study was to evaluate the effect of metformin on tumor response and survival in stage IV CRC patients with DM. METHODS We identified 106 patients who were diagnosed with both stage IV CRC and DM (81 patients who underwent palliative chemotherapy and 25 patients who underwent curative resection). Retrospective data of each patient's clinical characteristics, tumor response, and survival rate were compared between two groups of patients who either were or were not administered metformin. RESULTS For the palliative chemotherapy group, tumor response, change in target lesion size, progression free survival rate, and overall survival rate were not significantly different between the metformin group and the non-metformin group on univariate and multivariate analysis. For the curative resection patient group, metformin use was associated with increased disease free survival on univariate analysis (p=0.012) and multivariate analysis (hazard ratio, 0.024; 95% CI 0.001-0.435; p=0.010), but not with overall survival. CONCLUSIONS Metformin use in stage IV CRC patients with diabetes was shown to be associated with a lower risk of tumor recurrence after curative resection.
Collapse
Affiliation(s)
- Dong Jun Lee
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
141
|
Andersen DK, Andren-Sandberg Å, Duell EJ, Goggins M, Korc M, Petersen GM, Smith JP, Whitcomb DC. Pancreatitis-diabetes-pancreatic cancer: summary of an NIDDK-NCI workshop. Pancreas 2013; 42:1227-37. [PMID: 24152948 PMCID: PMC3878448 DOI: 10.1097/mpa.0b013e3182a9ad9d] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A workshop sponsored by the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and the National Cancer Institute (NCI) on "Pancreatitis-Diabetes-Pancreatic Cancer" focused on the risk factors of chronic pancreatitis (CP) and diabetes mellitus (DM) on the development of pancreatic ductal adenocarcinoma (PDAC). Sessions were held on (a) an overview of the problem of PDAC; (b) CP as a risk factor of PDAC; (c) DM as a risk factor of PDAC; (d) pancreatogenic, or type 3c, DM; (e) genomic associations of CP, DM, and PDAC; (f) surveillance of high-risk populations and early detection of PDAC; and (g) effects of DM treatment on PDAC. Recent data and current understandings of the mechanisms of CP- and DM-associated factors on PDAC development were discussed, and a detailed review of the possible risks of DM treatment on the development of PDAC was provided by representatives from academia, industry, and the Food and Drug Administration. The current status of possible biomarkers of PDAC and surveillance strategies for high-risk populations were discussed, and the gaps in knowledge and opportunities for further research were elucidated. A broad spectrum of expertise of the speakers and the discussants provided an unusually productive workshop, the highlights of which are summarized in the accompanying article.
Collapse
Affiliation(s)
- Dana K. Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | | | - Eric J. Duell
- Unit of Nutrition, Environment and Cancer, Catalan Institute of Oncology, Barcelona, Spain
| | - Michael Goggins
- Departments of Medicine and Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Murray Korc
- Departments of Medicine, Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Gloria M. Petersen
- Department of Epidemiology, Mayo Graduate School of Medicine, Rochester, MN
| | - Jill P. Smith
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - David C. Whitcomb
- Departments of Medicine, Cell Biology and Physiology, and Human Genetics, University of Pittsburgh School of Medicine
| |
Collapse
|
142
|
Shimoyama S. Diabetes mellitus carries a risk of gastric cancer: A meta-analysis. World J Gastroenterol 2013; 19:6902-6910. [PMID: 24187468 PMCID: PMC3812492 DOI: 10.3748/wjg.v19.i40.6902] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 05/27/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the association and quantify the relationship between diabetes mellitus (DM) and gastric cancer (GC) by an updated meta-analysis.
METHODS: The initial PubMed search identified 1233 publications. Studies not reporting GC or those not reporting actual number of GC were excluded. Twelve pertinent studies were retrieved from the PubMed database or from a manual search and considered for the meta-analysis. Pooled risk ratios and 95%CI were estimated by a random-effects model. Subgroup analysis was performed according to gender or geographical regions. Heterogeneity and publication bias were evaluated by I2 and funnel plot analysis, respectively.
RESULTS: DM was significantly associated with GC with a RR of 1.41 (P = 0.006) (95%CI: 1.10-1.81). Subgroup analyses revealed that both sexes showed a significant association with GC, with a greater magnitude of risk in females (RR = 1.90; 95%CI: 1.27-2.85; P = 0.002) than in males (RR = 1.24; 95%CI: 1.08-1.43; P = 0.002). In addition, the link between DM and GC was significant in East Asian DM patients (RR = 1.77; 95%CI: 1.38-2.26; P < 0.00001) but not in Western DM patients (RR = 1.23; 95%CI: 0.90-1.68; P = 0.2). There was no evidence of publication bias, but the results indicated significant heterogeneity.
CONCLUSION: This updated meta-analysis has provided evidence of positive DM-GC associations. The limited information on potentially important clinical confounding factors in each study deserves further investigation.
Collapse
|
143
|
Choi YK, Park KG. Metabolic roles of AMPK and metformin in cancer cells. Mol Cells 2013; 36:279-87. [PMID: 23794020 PMCID: PMC3887985 DOI: 10.1007/s10059-013-0169-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 06/06/2013] [Indexed: 12/20/2022] Open
Abstract
Metformin is one of the most widely used anti-diabetic agents in the world, and a growing body of evidence suggests that it may also be effective as an anti-cancer drug. Observational studies have shown that metformin reduces cancer incidence and cancer-related mortality in multiple types of cancer. These results have drawn attention to the mechanisms underlying metformin's anti-cancer effects, which may include triggering of the AMP-activated protein kinase (AMPK) pathway, resulting in vulnerability to an energy crisis (leading to cell death under conditions of nutrient deprivation) and a reduction in circulating insulin/IGF-1 levels. Clinical trials are currently underway to determine the benefits, appropriate dosage, and tolerability of metformin in the context of cancer therapy. This review highlights fundamental aspects of the molecular mechanisms underlying metformin's anti-cancer effects, describes the epidemiological evidence and ongoing clinical challenges, and proposes directions for future translational research.
Collapse
Affiliation(s)
- Yeon Kyung Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 700-721, Korea
| | - Keun-Gyu Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 700-721, Korea
| |
Collapse
|
144
|
Lappano R, De Marco P, De Francesco EM, Chimento A, Pezzi V, Maggiolini M. Cross-talk between GPER and growth factor signaling. J Steroid Biochem Mol Biol 2013; 137:50-6. [PMID: 23542661 DOI: 10.1016/j.jsbmb.2013.03.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/03/2013] [Accepted: 03/18/2013] [Indexed: 12/11/2022]
Abstract
G protein-coupled receptors (GPCRs) and growth factor receptors mediate multiple physio-pathological responses to a diverse array of extracellular stimuli. In this regard, it has been largely demonstrated that GPCRs and growth factor receptors generate a multifaceted signaling network, which triggers relevant biological effects in normal and cancer cells. For instance, some GPCRs transactivate the epidermal growth factor receptor (EGFR), which stimulates diverse transduction pathways leading to gene expression changes, cell migration, survival and proliferation. Moreover, it has been reported that a functional interaction between growth factor receptors and steroid hormones like estrogens is involved in the growth of many types of tumors as well as in the resistance to endocrine therapy. This review highlights recent findings on the cross-talk between a member of the GPCR family, the G protein-coupled estrogen receptor 1 (GPER, formerly known as GPR30) and two main growth factor receptors like EGFR and insulin-like growth factor-I receptor (IGF-IR). The biological implications of the functional interaction between these important mediators of cell responses particularly in cancer are discussed. This article is part of a Special Issue entitled 'CSR 2013'.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Dipartimento Farmaco-Biologico, Università della Calabria, via P. Bucci, 87036 Rende, Italy
| | | | | | | | | | | |
Collapse
|
145
|
Hajjar J, Habra MA, Naing A. Metformin: an old drug with new potential. Expert Opin Investig Drugs 2013; 22:1511-7. [PMID: 23978196 DOI: 10.1517/13543784.2013.833604] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Metformin is the most commonly prescribed antidiabetic oral agent. It has also been used off-label for polycystic ovarian syndrome, steatohepatitis, and HIV-associated metabolic abnormalities. However, this oldie is a newbie for the oncologist. Population studies have suggested that metformin decreased the incidence and mortality rates of cancer in diabetic patients. With better understanding of its mechanisms of antitumor activity, metformin may become a new drug for cancer in combination with chemotherapy or targeted therapy.
Collapse
Affiliation(s)
- Joud Hajjar
- Virginia Commonwealth University, Department of Internal Medicine, Division of Rheumatology, Allergy and Immunology , Richmond, VA , USA
| | | | | |
Collapse
|
146
|
Sluka P, Davis ID. Cell mates: paracrine and stromal targets for prostate cancer therapy. Nat Rev Urol 2013; 10:441-51. [PMID: 23857181 DOI: 10.1038/nrurol.2013.146] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
After many years of limited treatment options for patients with metastatic castration-resistant prostate cancer (mCRPC), multiple systemic therapies are now available, providing patients with significant improvements in survival, symptom control and bone health. Most of the recent advances in this area have been based on better understanding of mCRPC biology, particularly with respect to the key role of androgen receptor signalling. However, most therapies are targeted towards the malignant epithelial cell component of the cancer and it should not be forgotten that cancer cells exist in close and symbiotic relationships with other components of the tumour. Paracrine and stromal signals are often critical to the growth of the cancer and represent new potential therapeutic targets that are separate from the malignant epithelial cells. The stroma produces numerous growth factors, including vascular endothelial growth factor family members, platelet-derived growth factors and fibroblast growth factors, which are all critical for tumour growth. Targeting prostate-cancer-associated fibroblasts in order to destroy the physical and functional scaffold of a cancer is also a logical approach. The interaction between prostate cancer and the immune system remains an active topic of basic and clinical research, with cytokines, chemokines and growth factors being potential targets for therapy. The biology of epithelial-mesenchymal transition and of circulating tumour cells might also provide insight into new therapeutic targets.
Collapse
Affiliation(s)
- Pavel Sluka
- Monash University Eastern Health Clinical School, Level 2, 5 Arnold Street, Box Hill, Melbourne, VIC 3128, Australia
| | | |
Collapse
|
147
|
Margel D, Urbach D, Lipscombe LL, Bell CM, Kulkarni G, Austin PC, Fleshner N. Association between metformin use and risk of prostate cancer and its grade. J Natl Cancer Inst 2013; 105:1123-31. [PMID: 23853056 DOI: 10.1093/jnci/djt170] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Metformin is commonly prescribed to treat type 2 diabetes. Recent evidence suggests that it may possess antitumoral properties. The aim of this study was to test the association between metformin use and risk of prostate cancer and its grade among men with diabetes. METHODS Data were obtained from population-based health-care administrative databases in Ontario, Canada. This retrospective cohort study used a nested case-control approach to examine the relationship between metformin exposure and the risk of prostate cancer within a cohort of incident diabetic men aged 66 years or older. We conducted four case-control analyses, defining case subjects as 1) any prostate cancer, 2) high-grade, 3) low-grade, and 4) biopsy-diagnosed. In each analysis, case subjects were matched to five control subjects on age and cohort entry date. Metformin exposure was determined based on prescriptions before cancer diagnosis, and adjusted odds ratios (aOR) were estimated using conditional logistic regression. All statistical tests were two-sided. RESULTS Within our cohort of 119 315 men with diabetes, there were 5306 case subjects with prostate cancer and 26 530 matched control subjects. Within the cancer case subjects, 1104 had high- grade cancer, 1719 had low-grade cancer, and 3524 had biopsy-diagnosed cancer. There was no association between metformin use and risk of any prostate cancer (aOR = 1.03, 95% confidence interval [CI] = 0.96 to 1.1), high-grade cancer (aOR = 1.13, 95% CI = 0.96 to 1.32), low-grade cancer (aOR = 0.94, 95% CI = 0.82 to 1.06), or biopsy-diagnosed cancer (aOR = 0.98, 95% CI = 0.84 to 1.02). CONCLUSIONS This large study did not find an association between metformin use and risk of prostate cancer among older men with diabetes, regardless of cancer grade or method of diagnosis.
Collapse
Affiliation(s)
- David Margel
- Division of Urology, Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
148
|
Lee D, Vanden Broeck J, Lange AB. Identification and expression of the CCAP receptor in the Chagas' disease vector, Rhodnius prolixus, and its involvement in cardiac control. PLoS One 2013; 8:e68897. [PMID: 23874803 PMCID: PMC3706402 DOI: 10.1371/journal.pone.0068897] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/09/2013] [Indexed: 01/31/2023] Open
Abstract
Rhodnius prolixus is the vector of Chagas' disease, by virtue of transmitting the parasite Trypanosoma cruzi. There is no cure for Chagas' disease and therefore controlling R. prolixus is currently the only method of prevention. Understanding the physiology of the disease vector is an important step in developing control measures. Crustacean cardioactive peptide (CCAP) is an important neuropeptide in insects because it has multiple physiological roles such as controlling heart rate and modulating ecdysis behaviour. In this study, we have cloned the cDNA sequence of the CCAP receptor (RhoprCCAPR) from 5(th) instar R. prolixus and found it to be a G-protein coupled receptor (GPCR). The spatial expression pattern in 5(th) instars reveals that the RhoprCCAPR transcript levels are high in the central nervous system, hindgut and female reproductive systems, and lower in the salivary glands, male reproductive tissues and a pool of tissues including the dorsal vessel, trachea, and fat body. Interestingly, the RhoprCCAPR expression is increased prior to ecdysis and decreased post-ecdysis. A functional receptor expression assay confirms that the RhoprCCAPR is activated by CCAP (EC50 = 12 nM) but not by adipokinetic hormone, corazonin or an extended FMRFamide. The involvement of CCAP in controlling heartbeat frequency was studied in vivo and in vitro by utilizing RNA interference. In vivo, the basal heartbeat frequency is decreased by 31% in bugs treated with dsCCAPR. Knocking down the receptor in dsCCAPR-treated bugs also resulted in loss of function of applied CCAP in vitro. This is the first report of a GPCR knock-down in R. prolixus and the first report showing that a reduction in CCAPR transcript levels leads to a reduction in cardiac output in any insect.
Collapse
Affiliation(s)
- Dohee Lee
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| | | | | |
Collapse
|
149
|
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma is a devastating disease, with an overall 5-year survival rate of only 3% to 5%. As the current therapies offer very limited survival benefits, novel therapeutic strategies are urgently required to treat this disease. Here, we determined whether metformin administration inhibits the growth of PANC-1 and MiaPaCa-2 tumor xenografts in vivo. METHODS Different xenograft models, including orthotopic implantation, were used to determine whether intraperitoneal or oral administration of metformin inhibits the growth of pancreatic cancer in vivo. RESULTS We demonstrate that metformin given once daily intraperitoneally at various doses (50-250 mg/kg) to nude mice inhibited the growth of PANC-1 xenografts in a dose-dependent manner. A significant effect of metformin was obtained at 50 mg/kg and maximal effect at 200 mg/kg. Metformin administration also caused a significant reduction in the phosphorylation of ribosomal S6 protein and ERK in these xenografts. Metformin also inhibited the growth of pancreatic cancer xenografts when administered orally (2.5 mg/mL) either before or after tumor implantation. Importantly, oral administration of metformin also inhibited the growth of MiaPaCa-2 tumors xenografted orthotopically. CONCLUSIONS The studies presented here provide further evidence indicating that metformin offers a potential novel approach for pancreatic ductal adenocarcinoma prevention and therapy.
Collapse
|
150
|
Abstract
Up to 85% of patients with pancreatic cancer have diabetes or hyperglycaemia, which frequently manifests as early as 2-3 years before a diagnosis of pancreatic cancer. Conversely, patients with new-onset diabetes have a 5-8-fold increased risk of being diagnosed with pancreatic cancer within 1-3 years of developing diabetes. Emerging evidence now indicates that pancreatic cancer causes diabetes. As in type 2 diabetes, β-cell dysfunction and peripheral insulin resistance are seen in pancreatic cancer-induced diabetes. However, unlike in patients with type 2 diabetes, glucose control worsens in patients with pancreatic cancer in the face of ongoing, often profound, weight loss. Diabetes and weight loss, which precede cachexia onset by several months, are paraneoplastic phenomena induced by pancreatic cancer. Although the pathogenesis of these pancreatic cancer-induced metabolic alterations is only beginning to be understood, these are likely mechanisms to promote the survival and growth of pancreatic cancer in a hostile and highly desmoplastic microenvironment. Interestingly, these metabolic changes could enable early diagnosis of pancreatic cancer, if they can be distinguished from the ones that occur in patients with type 2 diabetes. One such possible biomarker is adrenomedullin, which is a potential mediator of β-cell dysfunction in pancreatic cancer-induced diabetes.
Collapse
|