1
|
Ahmmed R, Hossen MB, Ajadee A, Mahmud S, Ali MA, Mollah MMH, Reza MS, Islam MA, Mollah MNH. Bioinformatics analysis to disclose shared molecular mechanisms between type-2 diabetes and clear-cell renal-cell carcinoma, and therapeutic indications. Sci Rep 2024; 14:19133. [PMID: 39160196 PMCID: PMC11333728 DOI: 10.1038/s41598-024-69302-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 08/21/2024] Open
Abstract
Type 2 diabetes (T2D) and Clear-cell renal cell carcinoma (ccRCC) are both complicated diseases which incidence rates gradually increasing. Population based studies show that severity of ccRCC might be associated with T2D. However, so far, no researcher yet investigated about the molecular mechanisms of their association. This study explored T2D and ccRCC causing shared key genes (sKGs) from multiple transcriptomics profiles to investigate their common pathogenetic processes and associated drug molecules. We identified 259 shared differentially expressed genes (sDEGs) that can separate both T2D and ccRCC patients from control samples. Local correlation analysis based on the expressions of sDEGs indicated significant association between T2D and ccRCC. Then ten sDEGs (CDC42, SCARB1, GOT2, CXCL8, FN1, IL1B, JUN, TLR2, TLR4, and VIM) were selected as the sKGs through the protein-protein interaction (PPI) network analysis. These sKGs were found significantly associated with different CpG sites of DNA methylation that might be the cause of ccRCC. The sKGs-set enrichment analysis with Gene Ontology (GO) terms and KEGG pathways revealed some crucial shared molecular functions, biological process, cellular components and KEGG pathways that might be associated with development of both T2D and ccRCC. The regulatory network analysis of sKGs identified six post-transcriptional regulators (hsa-mir-93-5p, hsa-mir-203a-3p, hsa-mir-204-5p, hsa-mir-335-5p, hsa-mir-26b-5p, and hsa-mir-1-3p) and five transcriptional regulators (YY1, FOXL1, FOXC1, NR2F1 and GATA2) of sKGs. Finally, sKGs-guided top-ranked three repurposable drug molecules (Digoxin, Imatinib, and Dovitinib) were recommended as the common treatment for both T2D and ccRCC by molecular docking and ADME/T analysis. Therefore, the results of this study may be useful for diagnosis and therapies of ccRCC patients who are also suffering from T2D.
Collapse
Affiliation(s)
- Reaz Ahmmed
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Bayazid Hossen
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Agricultural and Applied Statistics, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Alvira Ajadee
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Sabkat Mahmud
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Ahad Ali
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Chemistry, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Manir Hossain Mollah
- Department of Physical Sciences, Independent University, Bangladesh (IUB), Dhaka, Bangladesh
| | - Md Selim Reza
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Division of Biomedical Informatics and Genomics, School of Medicine, Tulane University, 1440 Canal St., RM 1621C, New Orleans, LA, 70112, USA
| | - Mohammad Amirul Islam
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Nurul Haque Mollah
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| |
Collapse
|
2
|
Zaki HM, Ali KM, Abd Allah MYY, Abouelnaga AM, Abdraboh ME, Hussein O. Metronomic cyclophosphamide and metformin inhibited tumor growth and repopulated tumor-infiltrating lymphocytes in an experimental carcinoma model. BMC Res Notes 2024; 17:4. [PMID: 38167322 PMCID: PMC10759693 DOI: 10.1186/s13104-023-06651-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Metformin is a widely used antidiabetic biguanide. Retrospective data demonstrated the association of metformin use with survival benefit in multiple tumor types. Interest in repurposing metformin to treat cancer has not been translated into encouraging clinical benefit. In animal models, metformin activated cytotoxic T cells and exerted an immune-mediated anticancer effect. The current research was conducted to investigate the possible therapeutic benefit of metformin in combination with metronomic cyclophosphamide in an experimental cancer model. Ehrlich ascites carcinoma was injected into the subcutaneous tissue to induce solid tumors in syngeneic mice. Exponential solid tumor growth ensued and was effectively arrested with the administration of a cytotoxic dose of parenteral cyclophosphamide. Alternatively, oral metformin and continuous, low-dose cyclophosphamide significantly inhibited tumor growth relative to untreated mice. The drug combination was well tolerated. Histopathological examination of the tumor showed an increased number of tumor-infiltrating lymphocytes and enhanced expression of granzyme B by this drug combination. The current data suggests a potential role of metformin and metronomic chemotherapy that warrants further investigation.
Collapse
Affiliation(s)
- Heba Mohamed Zaki
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Khadiga Mohamed Ali
- Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | | | | | | | - Osama Hussein
- Surgical Oncology Department, Faculty of Medicine, Mansoura University Oncology Center, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
3
|
Takei K, Kijima T, Okubo N, Kurashina R, Kokubun H, Uematsu T, Betsunoh H, Yashi M, Kamai T. Association between Immune Checkpoint Inhibitor Treatment Outcomes and Body Composition Factors in Metastatic Renal Cell Carcinoma Patients. Cancers (Basel) 2023; 15:5591. [PMID: 38067295 PMCID: PMC10705346 DOI: 10.3390/cancers15235591] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 10/21/2024] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of metastatic renal cell carcinoma (mRCC); however, validating body composition-related biomarkers for their efficacy remains incomplete. We evaluated the association between body composition-related markers and the prognosis of patients with mRCC who received ICI-based first-line therapies. PATIENTS AND METHODS We retrospectively investigated 60 patients with mRCC who underwent ICI-based therapy as their first-line treatment between 2019 and 2023. Body composition variables, including skeletal muscle, subcutaneous fat, and visceral fat indices, were calculated using baseline computed tomography scans. Sarcopenia was defined according to sex-specific cut-off values of the skeletal mass index. The associations between body composition indices and objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) were evaluated. RESULTS Patients with sarcopenia had lower ORR and DCR than those without sarcopenia (33.3% vs. 61.1%, p = 0.0436 and 52.4% vs. 94.4%, p = 0.0024, respectively). Patients with sarcopenia had a significantly shorter median PFS (14 months vs. not reached, p = 0.0020) and OS (21 months vs. not reached, p = 0.0023) than patients without sarcopenia did. Sarcopenia was a significant predictor of PFS (hazard ratio [HR], 4.31; 95% confidence interval [CI], 1.65-14.8; p = 0.0018) and OS (HR, 5.44; 95% CI, 1.83-23.4; p = 0.0013) along with poor IMDC risk. No association was found between the subcutaneous, visceral, and total fat indices and the therapeutic effect of ICI-based therapy. CONCLUSIONS Sarcopenia was associated with a lower response and shorter survival rates in patients with mRCC who received first-line ICI-based therapy.
Collapse
Affiliation(s)
| | - Toshiki Kijima
- Department of Urology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga 321-0293, Tochigi, Japan; (K.T.); (N.O.); (R.K.); (H.K.); (T.U.); (H.B.); (M.Y.); (T.K.)
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Campi R, Rebez G, Klatte T, Roussel E, Ouizad I, Ingels A, Pavan N, Kara O, Erdem S, Bertolo R, Capitanio U, Mir MC. Effect of smoking, hypertension and lifestyle factors on kidney cancer - perspectives for prevention and screening programmes. Nat Rev Urol 2023; 20:669-681. [PMID: 37328546 DOI: 10.1038/s41585-023-00781-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 06/18/2023]
Abstract
Renal cell carcinoma (RCC) incidence has doubled over the past few decades. However, death rates have remained stable as the number of incidental renal mass diagnoses peaked. RCC has been recognized as a European health care issue, but to date, no screening programmes have been introduced. Well-known modifiable risk factors for RCC are smoking, obesity and hypertension. A direct association between cigarette consumption and increased RCC incidence and RCC-related death has been reported, but the underlying mechanistic pathways for this association are still unclear. Obesity is associated with an increased risk of RCC, but interestingly, improved survival outcomes have been reported in obese patients, a phenomenon known as the obesity paradox. Data on the association between other modifiable risk factors such as diet, dyslipidaemia and physical activity with RCC incidence are conflicting, and potential mechanisms underlying these associations remain to be elucidated.
Collapse
Affiliation(s)
- Riccardo Campi
- Department of Urology, University of Florence, Careggi Hospital, Florence, Italy
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
| | - Giacomo Rebez
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Tobias Klatte
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Royal Bournemouth Hospital, Bournemouth, UK
| | - Eduard Roussel
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, KU Leuven, Leuven, Belgium
| | - Idir Ouizad
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Bichat-Claude Bernard Hospital, Paris, France
| | - Alexander Ingels
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Henri Mondor Hospital, Créteil, France
| | - Nicola Pavan
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Onder Kara
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Faculty of Medicine, Kocaeli University, İzmit, Turkey
| | - Selcuk Erdem
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Istanbul University, Istanbul, Turkey
| | - Riccardo Bertolo
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Urology Unit, Department of Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Umberto Capitanio
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, San Raffaele Scientific Institute, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Maria Carmen Mir
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands.
- Department of Urology, Hospital Universitario La Ribera, Valencia, Spain.
| |
Collapse
|
5
|
Lu MZ, Li DY, Wang XF. Effect of metformin use on the risk and prognosis of ovarian cancer: an updated systematic review and meta-analysis. Panminerva Med 2023; 65:351-361. [PMID: 31290300 DOI: 10.23736/s0031-0808.19.03640-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Emerging evidence suggests that metformin has a potential antitumor effect both in vitro and in vivo. Increasing epidemiological studies indicate that diabetic patients receiving metformin therapy have lower incidences of cancer and have better survival rates. However, there are limited and inconsistent studies available about the effect of metformin therapy on ovarian cancer (OC). Thus, we conducted this meta-analysis to study the effect of metformin therapy on OC. Meanwhile, we systematically reviewed relevant studies to provide a framework for future research. EVIDENCE ACQUISITION We conducted a systematic literature search on PubMed, Web of Science, Springerlink, CNKI, VIP, SinoMed, and Wanfang up to the period of October 2018. A random-effects meta-analysis model was used to derive pooled effect estimates. EVIDENCE SYNTHESIS A total of 13 studies were retrieved of which 5 studies explained the prevention and 8 studies explained the treatment for OC. Our pooled results showed that metformin has a potential preventive effect on OC in diabetic women (pooled odds ratio [OR] 0.62, 95% confidence interval [95% CI] 0.34, 1.11; P<0.001). In addition, metformin can also significantly prolong progression-free survival (PFS) (pooled hazard ratio [HR] 0.49, 95% CI 0.34, 0.70; P=0.002), and overall survival (OS) (HR 0.71, 95%CI 0.61, 0.82; P<0.001) in patients with OC, regardless of whether they had diabetes. CONCLUSIONS The use of metformin can potentially reduce the risk of OC among diabetics, and it also can significantly improve PFS and OS in patients with OC. A further large clinical investigation would be needed to adopt our finding in practice, however, our systematic review provides an insight for future study designs.
Collapse
Affiliation(s)
- Min-Zhen Lu
- Second Clinical Medical College of Southern Medical University, Guangzhou, China -
| | - De-Yu Li
- Department of Oncology, Fujian Provincial Hospital, Fujian, China
| | - Xue-Feng Wang
- Department of Obstetrics and Gynecology, Third Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Metformin Suppresses Thioacetamide-Induced Chronic Kidney Disease in Association with the Upregulation of AMPK and Downregulation of Oxidative Stress and Inflammation as Well as Dyslipidemia and Hypertension. Molecules 2023; 28:molecules28062756. [PMID: 36985728 PMCID: PMC10056045 DOI: 10.3390/molecules28062756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Toxic chemicals such as carbon tetrachloride and thioacetamide (TAA) are reported to induce hepato-nephrotoxicity. The potential protective outcome of the antidiabetic and pleiotropic drug metformin against TAA-induced chronic kidney disease in association with the modulation of AMP-activated protein kinase (AMPK), oxidative stress, inflammation, dyslipidemia, and systemic hypertension has not been investigated before. Therefore, 200 mg/kg TAA was injected (via the intraperitoneal route) in a model group of rats twice a week starting at week 3 for 8 weeks. The control rats were injected with the vehicle for the same period. The metformin-treated group received 200 mg/kg metformin daily for 10 weeks, beginning week 1, and received TAA injections with dosage and timing similar to those of the model group. All rats were culled at week 10. It was observed that TAA induced substantial renal injury, as demonstrated by significant kidney tissue damage and fibrosis, as well as augmented blood and kidney tissue levels of urea, creatinine, inflammation, oxidative stress, dyslipidemia, tissue inhibitor of metalloproteinases-1 (TIMP-1), and hypertension. TAA nephrotoxicity substantially inhibited the renal expression of phosphorylated AMPK. All these markers were significantly protected by metformin administration. In addition, a link between kidney fibrosis and these parameters was observed. Thus, metformin provides profound protection against TAA-induced kidney damage and fibrosis associated with the augmentation of the tissue protective enzyme AMPK and inhibition of oxidative stress, inflammation, the profibrogenic gene TIMP-1, dyslipidemia, and hypertension for a period of 10 weeks in rats.
Collapse
|
7
|
Tan SK, Hougen HY, Merchan JR, Gonzalgo ML, Welford SM. Fatty acid metabolism reprogramming in ccRCC: mechanisms and potential targets. Nat Rev Urol 2023; 20:48-60. [PMID: 36192502 PMCID: PMC10826284 DOI: 10.1038/s41585-022-00654-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2022] [Indexed: 01/11/2023]
Abstract
Lipid droplet formation is a defining histological feature in clear-cell renal cell carcinoma (ccRCC) but the underlying mechanisms and importance of this biological behaviour have remained enigmatic. De novo fatty acid (FA) synthesis, uptake and suppression of FA oxidation have all been shown to contribute to lipid storage, which is a necessary tumour adaptation rather than a bystander effect. Clinical studies and mechanistic investigations into the roles of different enzymes in FA metabolism pathways have revealed new metabolic vulnerabilities that hold promise for clinical effect. Several metabolic alterations are associated with worse clinical outcomes in patients with ccRCC, as lipogenic genes drive tumorigenesis. Enzymes involved in the intrinsic FA metabolism pathway include FA synthase, acetyl-CoA carboxylase, ATP citrate lyase, stearoyl-CoA desaturase 1, cluster of differentiation 36, carnitine palmitoyltransferase 1A and the perilipin family, and each might be potential therapeutic targets in ccRCC owing to the link between lipid deposition and ccRCC risk. Adipokines and lipid species are potential biomarkers for diagnosis and treatment monitoring in patients with ccRCC. FA metabolism could potentially be targeted for therapeutic intervention in ccRCC as small-molecule inhibitors targeting the pathway have shown promising results in preclinical models.
Collapse
Affiliation(s)
- Sze Kiat Tan
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Helen Y Hougen
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jaime R Merchan
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Mark L Gonzalgo
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Scott M Welford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
| |
Collapse
|
8
|
Blonde L, Umpierrez GE, Reddy SS, McGill JB, Berga SL, Bush M, Chandrasekaran S, DeFronzo RA, Einhorn D, Galindo RJ, Gardner TW, Garg R, Garvey WT, Hirsch IB, Hurley DL, Izuora K, Kosiborod M, Olson D, Patel SB, Pop-Busui R, Sadhu AR, Samson SL, Stec C, Tamborlane WV, Tuttle KR, Twining C, Vella A, Vellanki P, Weber SL. American Association of Clinical Endocrinology Clinical Practice Guideline: Developing a Diabetes Mellitus Comprehensive Care Plan-2022 Update. Endocr Pract 2022; 28:923-1049. [PMID: 35963508 PMCID: PMC10200071 DOI: 10.1016/j.eprac.2022.08.002] [Citation(s) in RCA: 187] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The objective of this clinical practice guideline is to provide updated and new evidence-based recommendations for the comprehensive care of persons with diabetes mellitus to clinicians, diabetes-care teams, other health care professionals and stakeholders, and individuals with diabetes and their caregivers. METHODS The American Association of Clinical Endocrinology selected a task force of medical experts and staff who updated and assessed clinical questions and recommendations from the prior 2015 version of this guideline and conducted literature searches for relevant scientific papers published from January 1, 2015, through May 15, 2022. Selected studies from results of literature searches composed the evidence base to update 2015 recommendations as well as to develop new recommendations based on review of clinical evidence, current practice, expertise, and consensus, according to established American Association of Clinical Endocrinology protocol for guideline development. RESULTS This guideline includes 170 updated and new evidence-based clinical practice recommendations for the comprehensive care of persons with diabetes. Recommendations are divided into four sections: (1) screening, diagnosis, glycemic targets, and glycemic monitoring; (2) comorbidities and complications, including obesity and management with lifestyle, nutrition, and bariatric surgery, hypertension, dyslipidemia, retinopathy, neuropathy, diabetic kidney disease, and cardiovascular disease; (3) management of prediabetes, type 2 diabetes with antihyperglycemic pharmacotherapy and glycemic targets, type 1 diabetes with insulin therapy, hypoglycemia, hospitalized persons, and women with diabetes in pregnancy; (4) education and new topics regarding diabetes and infertility, nutritional supplements, secondary diabetes, social determinants of health, and virtual care, as well as updated recommendations on cancer risk, nonpharmacologic components of pediatric care plans, depression, education and team approach, occupational risk, role of sleep medicine, and vaccinations in persons with diabetes. CONCLUSIONS This updated clinical practice guideline provides evidence-based recommendations to assist with person-centered, team-based clinical decision-making to improve the care of persons with diabetes mellitus.
Collapse
Affiliation(s)
| | | | - S Sethu Reddy
- Central Michigan University, Mount Pleasant, Michigan
| | | | | | | | | | | | - Daniel Einhorn
- Scripps Whittier Diabetes Institute, La Jolla, California
| | | | | | - Rajesh Garg
- Lundquist Institute/Harbor-UCLA Medical Center, Torrance, California
| | | | | | | | | | | | - Darin Olson
- Colorado Mountain Medical, LLC, Avon, Colorado
| | | | | | - Archana R Sadhu
- Houston Methodist; Weill Cornell Medicine; Texas A&M College of Medicine; Houston, Texas
| | | | - Carla Stec
- American Association of Clinical Endocrinology, Jacksonville, Florida
| | | | - Katherine R Tuttle
- University of Washington and Providence Health Care, Seattle and Spokane, Washington
| | | | | | | | - Sandra L Weber
- University of South Carolina School of Medicine-Greenville, Prisma Health System, Greenville, South Carolina
| |
Collapse
|
9
|
The Impact of Metformin Use with Survival Outcomes in Urologic Cancers: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5311828. [PMID: 34660791 PMCID: PMC8519697 DOI: 10.1155/2021/5311828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/06/2021] [Accepted: 09/18/2021] [Indexed: 01/11/2023]
Abstract
Background Conflicting results exist between the potential protective effects of metformin and the prognosis of urologic cancers. This meta-analysis summarized the effects of metformin exposure on the recurrence, progression, cancer-specific survival (CSS), and overall survival (OS) of the three main urologic cancers (kidney cancer, bladder cancer, and prostate cancer). Methods We systematically searched PubMed, Embase, Web of Science, Wanfang, and China National Knowledge Infrastructure databases (January 2010 to December 2019), which identified studies regarding metformin users and nonusers with urologic cancers and extracted patient data. A random effect model or fixed effect model was used to analyze hazard ratios (HRs) and 95% confidence intervals (CIs). Results Among the 1883 confirmed studies, 27 eligible studies were identified, including 123,212 participants. In prostate cancer, patients using metformin have significant benefits for recurrence (HR = 0.74; 95% CI: 0.61-0.90; P = 0.007; I2 = 56%), CSS (HR = 0.74; 95% CI: 0.61-0.91; P = 0.002; I2 = 79%), and OS (HR = 0.76; 95% CI: 0.65-0.90; P < 0.001; I2 = 86%). Moreover, further subgroup analysis showed that the beneficial effects of metformin may be more significant for patients receiving radical radiotherapy. For kidney cancer, metformin was beneficial for progression (HR = 0.80; 95% CI: 0.65-0.98; P = 0.14; I2 = 46%). Analysis revealed that the effect of metformin on the overall survival of kidney cancer patients may be related to nationality (American: HR = 0.76; 95% CI: 0.59-0.98; P = 0.88; I2 = 0%). For bladder cancer, no obvious benefits of metformin use were identified. However, subgroup analysis indicated that metformin may improve the recurrence of bladder cancer, but this improvement was only found in patients with a median follow-up time of more than 4 years (HR = 0.43; 95% CI: 0.28-0.67; P = 0.61; I2 = 0%).
Collapse
|
10
|
Fiala O, Ostašov P, Rozsypalová A, Hora M, Šorejs O, Šustr J, Bendová B, Trávníček I, Filipovský J, Fínek J, Büchler T. Metformin Use and the Outcome of Metastatic Renal Cell Carcinoma Treated with Sunitinib or Pazopanib. Cancer Manag Res 2021; 13:4077-4086. [PMID: 34054309 PMCID: PMC8153069 DOI: 10.2147/cmar.s305321] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/10/2021] [Indexed: 12/25/2022] Open
Abstract
Background The anticancer properties of metformin have been suggested in numerous experimental studies and several retrospective clinical studies show that its use is associated with improved outcome of patients with cancer. However, limited data are available for patients with metastatic renal cell carcinoma (mRCC) treated with targeted therapy. The aim of this retrospective study was to assess the impact of the metformin use on survival of mRCC patients treated with sunitinib or pazopanib. Methods Clinical data from 343 patients with mRCC treated with sunitinib or pazopanib in the first line were analyzed. Progression-free survival (PFS) and overall survival (OS) were compared according to the use of metformin. Results The median PFS and OS for patients using metformin was 31.1 (95% CI 20.6–35.1) and 51.6 (95% CI 44.7-NR) months compared to 9.3 (95% CI 8.0–12.0) and 22.4 (95% CI 19.4–26.8) months for patients not using metformin (p<0.0001 and p=0.0002, respectively). Cox multivariate analysis shows that the use of metformin remains a significant factor for PFS (HR=0.55 [95% CI 0.343–0.883], p=0.013) and also for OS (HR=0.45 [95% CI 0.256–0.794], p=0.006). Conclusion The present study results suggest that the use of metformin was associated with favorable outcome of mRCC patients treated with sunitinib or pazopanib.
Collapse
Affiliation(s)
- Ondřej Fiala
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic.,Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech, Czech Republic
| | - Pavel Ostašov
- Laboratory of Tumor Biology and Immunotherapy, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech, Czech Republic
| | - Aneta Rozsypalová
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Milan Hora
- Department of Urology, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Šorejs
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic.,Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech, Czech Republic
| | - Jan Šustr
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Barbora Bendová
- Department of Urology, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ivan Trávníček
- Department of Urology, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jan Filipovský
- 2nd Department of Internal Medicine, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jindřich Fínek
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Tomáš Büchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| |
Collapse
|
11
|
Abstract
Renal cell carcinoma is associated with chronic kidney disease as well as with common risk factors including hypertension and diabetes mellitus. Localized renal cell carcinoma is treated surgically and in these cases has a favorable prognosis. In particular, in those individuals with small renal masses (≤4 cm), preservation of kidney function should be prioritized. Postoperative chronic kidney disease or end-stage renal disease prevention should include baseline kidney function and risk factor assessment, nontumor renal biopsy, as well as counseling on treatment options to discuss maximizing kidney function preservation. Postnephrectomy prognosis can be determined with repeat laboratory and clinical assessment. Ultimately, early involvement of the nephrologist in a multidisciplinary team including the urology team will enable the reduction of postsurgical kidney disease related morbidity and potentially mortality.
Collapse
Affiliation(s)
- Susie L Hu
- Division of Kidney Disease and Hypertension, Department of Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI.
| |
Collapse
|
12
|
Curran CS, Kopp JB. PD-1 immunobiology in glomerulonephritis and renal cell carcinoma. BMC Nephrol 2021; 22:80. [PMID: 33676416 PMCID: PMC7936245 DOI: 10.1186/s12882-021-02257-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/31/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Programmed cell death protein (PD)-1 receptors and ligands on immune cells and kidney parenchymal cells help maintain immunological homeostasis in the kidney. Dysregulated PD-1:PD-L1 binding interactions occur during the pathogenesis of glomerulopathies and renal cell carcinoma (RCC). The regulation of these molecules in the kidney is important to PD-1/PD-L1 immunotherapies that treat RCC and may induce glomerulopathies as an adverse event. METHODS The expression and function of PD-1 molecules on immune and kidney parenchymal cells were reviewed in the healthy kidney, PD-1 immunotherapy-induced nephrotoxicity, glomerulopathies and RCC. RESULTS PD-1 and/or its ligands are expressed on kidney macrophages, dendritic cells, lymphocytes, and renal proximal tubule epithelial cells. Vitamin D3, glutathione and AMP-activated protein kinase (AMPK) regulate hypoxic cell signals involved in the expression and function of PD-1 molecules. These pathways are altered in kidney disease and are linked to the production of vascular endothelial growth factor, erythropoietin, adiponectin, interleukin (IL)-18, IL-23, and chemokines that bind CXCR3, CXCR4, and/or CXCR7. These factors are differentially produced in glomerulonephritis and RCC and may be important biomarkers in patients that receive PD-1 therapies and/or develop glomerulonephritis as an adverse event CONCLUSION: By comparing the functions of the PD-1 axis in glomerulopathies and RCC, we identified similar chemokines involved in the recruitment of immune cells and distinct mediators in T cell differentiation. The expression and function of PD-1 and PD-1 ligands in diseased tissue and particularly on double-negative T cells and parenchymal kidney cells needs continued exploration. The possible regulation of the PD-1 axis by vitamin D3, glutathione and/or AMPK cell signals may be important to kidney disease and the PD-1 immunotherapeutic response.
Collapse
Affiliation(s)
- Colleen S Curran
- Critical Care Medicine Department, Clinical Center, NIH, BG 10 RM 2C135, 10 Center Drive, Bethesda, MD, 20814, USA.
| | | |
Collapse
|
13
|
Sciannimanico S, Grimaldi F, Vescini F, De Pergola G, Iacoviello M, Licchelli B, Guastamacchia E, Giagulli VA, Triggiani V. Metformin: Up to Date. Endocr Metab Immune Disord Drug Targets 2020; 20:172-181. [PMID: 31670618 DOI: 10.2174/1871530319666190507125847] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Metformin is an oral hypoglycemic agent extensively used as first-line therapy for type 2 diabetes. It improves hyperglycemia by suppressing hepatic glucose production and increasing glucose uptake in muscles. Metformin improves insulin sensitivity and shows a beneficial effect on weight control. Besides its metabolic positive effects, Metformin has direct effects on inflammation and can have immunomodulatory and antineoplastic properties. AIM The aim of this narrative review was to summarize the up-to-date evidence from the current literature about the metabolic and non-metabolic effects of Metformin. METHODS We reviewed the current literature dealing with different effects and properties of Metformin and current recommendations about the use of this drug. We identified keywords and MeSH terms in Pubmed and the terms Metformin and type 2 diabetes, type 1 diabetes, pregnancy, heart failure, PCOS, etc, were searched, selecting only significant original articles and review in English, in particular of the last five years. CONCLUSION Even if many new effective hypoglycemic agents have been launched in the market in the last few years, Metformin would always keep a place in the treatment of type 2 diabetes and its comorbidities because of its multiple positive effects and low cost.
Collapse
Affiliation(s)
| | - Franco Grimaldi
- Endocrinology and Metabolism Unit, University Hospital of Udine, Udine, Italy
| | - Fabio Vescini
- Endocrinology and Metabolism Unit, University Hospital of Udine, Udine, Italy
| | - Giovanni De Pergola
- Clinical Nutrition Unit, Medical Oncology, Department of Biomedical Science and Human Oncology, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Massimo Iacoviello
- University Cardiology Unit, Cardiothoracic Department, Policlinic University Hospital, Bari, Italy
| | - Brunella Licchelli
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Vito A Giagulli
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Italy
| |
Collapse
|
14
|
Is metformin a geroprotector? A peek into the current clinical and experimental data. Mech Ageing Dev 2020; 191:111350. [DOI: 10.1016/j.mad.2020.111350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/25/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
|
15
|
Pusceddu S, Vernieri C, Prinzi N, Torchio M, Coppa J, Antista M, Niger M, Milione M, Giacomelli L, Corti F, Prisciandaro M, Monteleone M, Colombo E, Di Bartolomeo M, de Braud F. The potential role of metformin in the treatment of patients with pancreatic neuroendocrine tumors: a review of preclinical to clinical evidence. Therap Adv Gastroenterol 2020; 13:1756284820927271. [PMID: 32821286 PMCID: PMC7406937 DOI: 10.1177/1756284820927271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/15/2020] [Indexed: 02/04/2023] Open
Abstract
The incidence of pancreatic neuroendocrine tumors (panNETs) has increased worldwide in the last two decades. Given the indolent nature of these tumors, several patients are diagnosed with metastatic disease, which partially impairs the long-term efficacy of currently available treatments and reduces survival rates. The search for new therapeutic strategies for cancer patients has pushed towards the retrospective analysis of studies involving patients who concomitantly received other drugs together with standard anticancer agents. In this light, several retrospective analyses have shown that metformin use is associated with improved prognosis in patients with different tumor types treated with standard antitumor agents. Metformin, the cornerstone oral agent for the treatment of type 2 diabetes, plays a role in modulating glucose cell metabolism. Its potential ability to interfere with tumors may derive from the tight relationship between metabolic reprogramming in cancer cells and tumor progression. Indications for metformin use as an anticancer drug result from pre-clinical and clinical observations. In particular, metformin use in diabetic patients with advanced panNETs has been associated with better progression-free survival in patients treated with somatostatin analogues with or without metformin.
Collapse
Affiliation(s)
| | - Claudio Vernieri
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Natalie Prinzi
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Martina Torchio
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Jorgelina Coppa
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Maria Antista
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Monica Niger
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Massimo Milione
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Luca Giacomelli
- Polistudium SRL (Milan, Italy) and Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Francesca Corti
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Michele Prisciandaro
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Michela Monteleone
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Elena Colombo
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Maria Di Bartolomeo
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Filppo de Braud
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy University of Milan, Milan, Italy
| |
Collapse
|
16
|
Abstract
Renal cancer is a serious and common type of cancer affecting old ages. The growth of such type of cancer can be stopped by detecting it before it reaches advanced or end-stage. Hence, renal cancer must be identified and diagnosed in the initial stages. In this research paper, an intelligent medical diagnostic system to diagnose renal cancer is developed by using fuzzy and neuro-fuzzy techniques. Essentially, for a fuzzy inference system, two layers are used. The first layer gives the output about whether the patient is having renal cancer or not. Similarly, the second layer detects the current stage of suffering patients. While in the development of a medical diagnostic system by using a neuro-fuzzy technique, the Gaussian membership functions are used for all the input variables considered for the diagnosis. In this paper, the comparison between the performance of developed systems has been done by taking some suitable parameters. The results obtained from this comparison study show that the intelligent medical system developed by using a neuro-fuzzy model gives the more precise and accurate results than existing systems.
Collapse
|
17
|
Pan Q, Lu X, Zhao C, Liao S, Chen X, Guo F, Yang C, Liu HF. Metformin: the updated protective property in kidney disease. Aging (Albany NY) 2020; 12:8742-8759. [PMID: 32364526 PMCID: PMC7244070 DOI: 10.18632/aging.103095] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
Abstract
Metformin is a frontline hypoglycemic agent, which is mainly prescribed to manage type 2 diabetes mellitus with obesity. Emerging evidence suggests that metformin also exerts protective effects against various kidney diseases. Some postulate that kidney disease is actually a metabolic disease, accompanied by nonresolving pathophysiologic pathways controlling oxidative stress, endoplasmic reticulum stress, inflammation, lipotoxicity, fibrosis, and senescence, as well as insufficient host defense mechanisms such as AMP-activated protein kinase (AMPK) signaling and autophagy. Metformin may interfere with these pathways by orchestrating AMPK signaling and AMPK-independent pathways to protect the kidneys from injury. Furthermore, the United States Food and Drug Administration declared metformin is safe for patients with mild or moderate kidney impairment in 2016, assuaging some conservative attitudes about metformin management in patients with renal insufficiency and broadening the scope of research on the renal protective effects of metformin. This review focuses on the molecular mechanisms by which metformin imparts renal protection and its potential in the treatment of various kidney diseases.
Collapse
Affiliation(s)
- Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Xing Lu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Chunfei Zhao
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Shuzhen Liao
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Xiaoqun Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Chen Yang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| |
Collapse
|
18
|
Barbieri F, Verduci I, Carlini V, Zona G, Pagano A, Mazzanti M, Florio T. Repurposed Biguanide Drugs in Glioblastoma Exert Antiproliferative Effects via the Inhibition of Intracellular Chloride Channel 1 Activity. Front Oncol 2019; 9:135. [PMID: 30918838 PMCID: PMC6424887 DOI: 10.3389/fonc.2019.00135] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
The lack of in-depth knowledge about the molecular determinants of glioblastoma (GBM) occurrence and progression, combined with few effective and BBB crossing-targeted compounds represents a major challenge for the discovery of novel and efficacious drugs for GBM. Among relevant molecular factors controlling the aggressive behavior of GBM, chloride intracellular channel 1 (CLIC1) represents an emerging prognostic and predictive biomarker, as well as a promising therapeutic target. CLIC1 is a metamorphic protein, co-existing as both soluble cytoplasmic and membrane-associated conformers, with the latter acting as chloride selective ion channel. CLIC1 is involved in several physiological cell functions and its abnormal expression triggers tumor development, favoring tumor cell proliferation, invasion, and metastasis. CLIC1 overexpression is associated with aggressive features of various human solid tumors, including GBM, in which its expression level is correlated with poor prognosis. Moreover, increasing evidence shows that modification of microglia ion channel activity, and CLIC1 in particular, contributes to the development of different neuropathological states and brain tumors. Intriguingly, CLIC1 is constitutively active within cancer stem cells (CSCs), while it seems less relevant for the survival of non-CSC GBM subpopulations and for normal cells. CSCs represent GBM development and progression driving force, being endowed with stem cell-like properties (self-renewal and differentiation), ability to survive therapies, to expand and differentiate, causing tumor recurrence. Downregulation of CLIC1 results in drastic inhibition of GBM CSC proliferation in vitro and in vivo, making the control of the activity this of channel a possible innovative pharmacological target. Recently, drugs belonging to the biguanide class (including metformin) were reported to selectively inhibit CLIC1 activity in CSCs, impairing their viability and invasiveness, but sparing normal stem cells, thus representing potential novel antitumor drugs with a safe toxicological profile. On these premises, we review the most recent insights into the biological role of CLIC1 as a potential selective pharmacological target in GBM. Moreover, we examine old and new drugs able to functionally target CLIC1 activity, discussing the challenges and potential development of CLIC1-targeted therapies.
Collapse
Affiliation(s)
- Federica Barbieri
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica, Università di Genoa, Genoa, Italy
| | - Ivan Verduci
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Valentina Carlini
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Gianluigi Zona
- Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università di Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Aldo Pagano
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Dipartimento di Medicina Sperimentale, Università di Genoa, Genoa, Italy
| | - Michele Mazzanti
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica, Università di Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
19
|
|
20
|
Yu H, Zhong X, Gao P, Shi J, Wu Z, Guo Z, Wang Z, Song Y. The Potential Effect of Metformin on Cancer: An Umbrella Review. Front Endocrinol (Lausanne) 2019; 10:617. [PMID: 31620081 PMCID: PMC6760464 DOI: 10.3389/fendo.2019.00617] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/27/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Metformin has been reported to possess anti-cancer properties in addition to glucose-lowering activity and numerous systematic reviews and meta-analyses have studied the association between metformin use and cancer incidence or survival outcomes. We performed an umbrella review to assess the robustness of these associations to facilitate proper interpretation of these results to inform clinical and policy decisions. Methods: We searched PubMed and Embase systematic reviews and meta-analyses investigating the effect of metformin use on cancer incidence or survival outcomes published from inception to September 2, 2018. We estimated the summary effect size, the 95% CI, and the 95% prediction interval, heterogeneity, evidence of small-study effects, and evidence of excess significance bias. Results: We included 21 systematic reviews and meta-analyses covering 11 major anatomical sites and 33 associations. There was strong evidence for the association between metformin use and decreased pancreatic cancer incidence. The association between metformin use and improved colorectal cancer overall survival (OS) was supported by highly suggestive evidence. Seven associations (all cancer incidence, all cancer OS, breast cancer OS, colorectal cancer incidence, liver cancer incidence, lung cancer OS, and pancreatic cancer OS) presented only suggestive evidence. The remaining 24 associations were supported by weak or not-suggestive evidence. Conclusions: Associations between metformin use and pancreatic cancer incidence or colorectal cancer OS are supported by strong or highly suggestive evidence, respectively. However, these results should be interpreted with caution due to the poor methodological quality of the systematic reviews and meta-analyses.
Collapse
|
21
|
Wen-Xiu X, Xiao-Wei Z, Hai-Ying D, Ying-Hui T, Si-Si K, Xiao-Fang Z, Huang P. Impact of metformin use on survival outcomes in non-small cell lung cancer treated with platinum. Medicine (Baltimore) 2018; 97:e13652. [PMID: 30572481 PMCID: PMC6320173 DOI: 10.1097/md.0000000000013652] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Preclinical evidence suggests that metformin, a widely used antidiabetic drug, may have a sensitizing effect on platinum. The purpose of this study was to evaluate the survival outcomes for non-small cell lung cancer (NSCLC) patients with type 2 diabetes mellitus (T2DM) using metformin during platinum-based chemotherapy.The clinicopathological parameters and survival data of 75 NSCLC patients with T2DM from January 2008 to December 2011 were collected and analyzed retrospectively. Patients were divided into 2 groups: metformin exposure group (n = 27) and non-metformin group (patients using other hypoglycemic agents or no drug for controlling n = 48). Univariate and multivariate analyses were performed to assess the association of metformin usage with overall survival (OS).Mean follow-up time was 58.7 months. The mean survival time was 36.74 months in the metformin group and 40.21 months in the non-metformin group. There was no significant difference in survival time between the 2 groups (P = .661). After adjusting gender, age, smoking status, tumor stage, tumor histology, and differentiation, multivariate analysis showed that metformin was not associated with the OS in NSCLC patients treated with concurrent platinum-based chemotherapy (hazard ratio: 1.071, 95% confidence interval: 0.577-1.986, P = .828).Our results indicated that metformin exposure had no significant effect on OS in NSCLC patients treated with platinum-based chemotherapy. Further studies are warranted to evaluate whether metformin could affect the survival of NSCLC patients treated with platinum-based chemotherapy.
Collapse
Affiliation(s)
- Xin Wen-Xiu
- Laboratory of Clinical Pharmacy
- Key Laboratory of Head and Neck Translational Research of Zhejiang Province Zhejiang Cancer Hospital, Hangzhou, P.R. China
| | | | | | | | | | | | - Ping Huang
- Laboratory of Clinical Pharmacy
- Key Laboratory of Head and Neck Translational Research of Zhejiang Province Zhejiang Cancer Hospital, Hangzhou, P.R. China
| |
Collapse
|
22
|
Abstract
Metformin is a lipophilic biguanide which inhibits hepatic gluconeogenesis and improves peripheral utilization of glucose. It is the first line pharmacotherapy for glucose control in patients with Type 2 diabetes due to its safety, efficacy and tolerability. Metformin exhibits pleotropic effects, which may have beneficial effects on a variety of tissues independent of glucose control. A potential anti-tumourigenic effect of metformin may be mediated by its role in activating AMP-kinase, which in turn inhibits mammalian target of rapamycin (mTOR). Non-AMPK dependent protective pathways may include reduction of insulin, insulin-like growth factor-1, leptin, inflammatory pathways and potentiation of adiponectin, all of which may have a role in tumourigenesis. A role in inhibiting cancer stem cells is also postulated. A number of large scale observational and cohort studies suggest metformin is associated with a reduced risk of a number of cancers, although the data is not conclusive. Recent randomised studies reporting use of metformin in treatment of cancer have revealed mixed results, and the results of much larger randomised trials of metformin as an adjuvant therapy in breast and colorectal cancers are awaited.
Collapse
Affiliation(s)
- Ritwika Mallik
- (a)International Training Fellow in Endocrinology and Diabetes, Department of Diabetes and Metabolism, Barts Health NHS Trust, London, UK
| | - Tahseen A Chowdhury
- Department of Diabetes and Metabolism, Barts and the London School of Medicine and Dentistry, London, UK.
| |
Collapse
|
23
|
Wei M, Mao S, Lu G, Li L, Lan X, Huang Z, Chen Y, Zhao M, Zhao Y, Xia Q. Valproic acid sensitizes metformin-resistant human renal cell carcinoma cells by upregulating H3 acetylation and EMT reversal. BMC Cancer 2018; 18:434. [PMID: 29665787 PMCID: PMC5902941 DOI: 10.1186/s12885-018-4344-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 04/08/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Metformin (Met) is a widely available diabetic drug and shows suppressed effects on renal cell carcinoma (RCC) metabolism and proliferation. Laboratory studies in RCC suggested that metformin has remarkable antitumor activities and seems to be a potential antitumor drug. But the facts that metformin may be not effective in reducing the risk of RCC in cancer clinical trials made it difficult to determine the benefits of metformin in RCC prevention and treatment. The mechanisms underlying the different conclusions between laboratory experiments and clinical analysis remains unclear. The goal of the present study was to determine whether long-term metformin use can induce resistance in RCC, whether metformin resistance could be used to explain the disaccord in laboratory and clinical studies, and whether the drug valproic acid (VPA), which inhibits histone deacetylase, exhibits synergistic cytotoxicity with metformin and can counteract the resistance of metformin in RCC. METHODS We performed CCK8, transwell, wound healing assay, flow cytometry and western blotting to detect the regulations of proliferation, migration, cell cycle and apoptosis in 786-O, ACHN and metformin resistance 786-O (786-M-R) cells treated with VPA, metformin or a combination of two drugs. We used TGF-β, SC79, LY294002, Rapamycin, protein kinase B (AKT) inhibitor to treat the 786-O or 786-M-R cells and detected the regulations in TGF-β /pSMAD3 and AMPK/AKT pathways. RESULTS 786-M-R was refractory to metformin-induced antitumor effects on proliferation, migration, cell cycle and cell apoptosis. AMPK/AKT pathways and TGF-β/SMAD3 pathways showed low sensibilities in 786-M-R. The histone H3 acetylation diminished in the 786-M-R cells. However, the addition of VPA dramatically upregulated histone H3 acetylation, increased the sensibility of AKT and inhibited pSMAD3/SMAD4, letting the combination of VPA and metformin remarkably reappear the anti-tumour effects of metformin in 786-M-R cells. CONCLUSIONS VPA not only exhibits synergistic cytotoxicity with metformin but also counteracts resistance to metformin in renal cell carcinoma cell. The re-sensitization to metformin induced by VPA in metformin-resistant cells may help treat renal cell carcinoma patients.
Collapse
Affiliation(s)
- Muyun Wei
- Department of Center Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, 544 Jingsi Road, Jinan, 250001, Shandong Province, China
| | - Shaowei Mao
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Guoliang Lu
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Liang Li
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Xiaopeng Lan
- Department of Urology, Qingdao center Hospital, Qingdao, 266042, Shandong Province, China
| | - Zhongxian Huang
- Department of Urology, Jinan center Hospital, Jinan, 250001, Shandong Province, China
| | - Yougen Chen
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China
| | - Miaoqing Zhao
- Department of pathology, Shandong Provincial Hospital Affiliated to Shandong University, 324, Jingwu weiqi Road, Jinan, 250001, Shandong Province, China
| | - Yueran Zhao
- Department of Center Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, 544 Jingsi Road, Jinan, 250001, Shandong Province, China
| | - Qinghua Xia
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, 9677 Jingshidong Road, Jinan, 250001, Shandong Province, China.
| |
Collapse
|
24
|
Saini N, Yang X. Metformin as an anti-cancer agent: actions and mechanisms targeting cancer stem cells. Acta Biochim Biophys Sin (Shanghai) 2018; 50:133-143. [PMID: 29342230 DOI: 10.1093/abbs/gmx106] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022] Open
Abstract
Metformin, a first line medication for type II diabetes, initially entered the spotlight as a promising anti-cancer agent due to epidemiologic reports that found reduced cancer risk and improved clinical outcomes in diabetic patients taking metformin. To uncover the anti-cancer mechanisms of metformin, preclinical studies determined that metformin impairs cellular metabolism and suppresses oncogenic signaling pathways, including receptor tyrosine kinase, PI3K/Akt, and mTOR pathways. Recently, the anti-cancer potential of metformin has gained increasing interest due to its inhibitory effects on cancer stem cells (CSCs), which are associated with tumor metastasis, drug resistance, and relapse. Studies using various cancer models, including breast, pancreatic, prostate, and colon, have demonstrated the potency of metformin in attenuating CSCs through the targeting of specific pathways involved in cell differentiation, renewal, metastasis, and metabolism. In this review, we provide a comprehensive overview of the anti-cancer actions and mechanisms of metformin, including the regulation of CSCs and related pathways. We also discuss the potential anti-cancer applications of metformin as mono- or combination therapies.
Collapse
Affiliation(s)
- Nipun Saini
- Julius L. Chambers Biomedical/Biotechnology Research Institute, Department of Biological and Biomedical Sciences, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - Xiaohe Yang
- Julius L. Chambers Biomedical/Biotechnology Research Institute, Department of Biological and Biomedical Sciences, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC 28081, USA
| |
Collapse
|
25
|
Campbell JM, Bellman SM, Stephenson MD, Lisy K. Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: A systematic review and meta-analysis. Ageing Res Rev 2017; 40:31-44. [PMID: 28802803 DOI: 10.1016/j.arr.2017.08.003] [Citation(s) in RCA: 279] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 12/19/2022]
Abstract
This systematic review investigated whether the insulin sensitiser metformin has a geroprotective effect in humans. Pubmed and Embase were searched along with databases of unpublished studies. Eligible research investigated the effect of metformin on all-cause mortality or diseases of ageing relative to non-diabetic populations or diabetics receiving other therapies with adjustment for disease control achieved. Overall, 260 full-texts were reviewed and 53 met the inclusion criteria. Diabetics taking metformin had significantly lower all-cause mortality than non-diabetics (hazard ratio (HR)=0.93, 95%CI 0.88-0.99), as did diabetics taking metformin compared to diabetics receiving non-metformin therapies (HR=0.72, 95%CI 0.65-0.80), insulin (HR=0.68, 95%CI 0.63-0.75) or sulphonylurea (HR=0.80, 95%CI 0.66-0.97). Metformin users also had reduced cancer compared to non-diabetics (rate ratio=0.94, 95%CI 0.92-0.97) and cardiovascular disease (CVD) compared to diabetics receiving non-metformin therapies (HR=0.76, 95%CI 0.66-0.87) or insulin (HR=0.78, 95%CI 0.73-0.83). Differences in baseline characteristics were observed which had the potential to bias findings, although statistical adjustments were made. The apparent reductions in all-cause mortality and diseases of ageing associated with metformin use suggest that metformin could be extending life and healthspans by acting as a geroprotective agent.
Collapse
Affiliation(s)
- Jared M Campbell
- The Joanna Briggs Institute, The University of Adelaide, Adelaide, South Australia, Australia; Centre for Nanoscale BioPhotonics, Macquarie University, Sydney, New South Wales, Australia.
| | - Susan M Bellman
- The Joanna Briggs Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Matthew D Stephenson
- The Joanna Briggs Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Karolina Lisy
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Xie W, Li T, Yang J, Shang M, Xiao Y, Li Q, Yang J. Metformin use and survival outcomes in endometrial cancer: a systematic review and meta-analysis. Oncotarget 2017; 8:73079-73086. [PMID: 29069850 PMCID: PMC5641193 DOI: 10.18632/oncotarget.20388] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/04/2017] [Indexed: 01/01/2023] Open
Abstract
Previous studies have evaluated the effects of metformin use on survival outcomes in endometrial cancer, but their results are inconsistent. We conducted a systematic review and meta-analysis to provide a quantitative assessment of the drug's effects based on available evidence. We searched PubMed, Embase, and the Cochrane Central Register of Controlled Trials to identify relevant studies that evaluated the association between metformin use on survival outcomes in endometrial cancer. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated to evaluate the association of metformin use with overall survival and with progression-free survival using a fixed-effects model. A total of nine studies involving 2,016 patients with endometrial cancer were identified. In a meta-analysis of eight studies involving 1,594 individuals, metformin use was associated with significant improvements in overall survival (HR, 0.51; 95% CI, 0.41 to 0.64). Metformin users similarly showed improved progression-free survival in a meta-analysis of two studies involving 632 individuals (HR, 0.63; 95% CI, 0.46 to 0.87). In conclusion, endometrial cancer patients who use metformin show improved overall survival and progression-free survival. Further studies are required to confirm the full potential effects of metformin use on survival outcomes in endometrial cancer.
Collapse
Affiliation(s)
- Weimin Xie
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianjia Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Yang
- Department of Obstetrics and Gynecology, Maternal and Child Health Care Hospital of Hunan Province, Changsha, China
| | - Mengmeng Shang
- Department of Scientific Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Xiao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Li
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaxin Yang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|