1
|
Sahu P, Camarillo IG, Dettin M, Zamuner A, Teresa Conconi M, Barozzi M, Giri P, Sundararajan R, Sieni E. Electroporation enhances cell death in 3D scaffold-based MDA-MB-231 cells treated with metformin. Bioelectrochemistry 2024; 159:108734. [PMID: 38762949 DOI: 10.1016/j.bioelechem.2024.108734] [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: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype of breast cancer lacks estrogen, progesterone, and HER2 receptors and hence, is therapeutically challenging. Towards this, we studied an alternate therapy by repurposing metformin (FDA-approved type-2 diabetic drug with anticancer properties) in a 3D-scaffold culture, with electrical pulses. 3D cell culture was used to simulate the tumor microenvironment more closely and MDA-MB-231, human TNBC cells, treated with both 5 mM metformin (Met) and 8 electrical pulses at 2500 V/cm, 10 µs (EP1) and 800 V/cm, 100 µs (EP2) at 1 Hz were studied in 3D and 2D. They were characterized using cell viability, reactive oxygen species (ROS), glucose uptake, and lactate production assays at 24 h. Cell viability, as low as 20 % was obtained with EP1 + 5 mM Met. They exhibited 1.65-fold lower cell viability than 2D with EP1 + 5 mM Met. ROS levels indicated a 2-fold increase in oxidative stress for EP1 + 5 mM Met, while the glucose uptake was limited to only 9 %. No significant change in the lactate production indicated glycolytic arrest and a non-conducive environment for MDA-MB-231 growth. Our results indicate that 3D cell culture, with a more realistic tumor environment that enhances cell death using metformin and electrical pulses could be a promising approach for TNBC therapeutic intervention studies.
Collapse
Affiliation(s)
- Praveen Sahu
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Ignacio G Camarillo
- Deptartment of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
| | - Monica Dettin
- Department of Industrial Engineering, University of Padova, Padova 35122, Italy
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padova, Padova 35122, Italy; Department of Civil, Environmental, and Architectural Engineering, University of Padova, Italy
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35131, Italy
| | - Marco Barozzi
- Department of Theoretical and Applied Sciences, University of Insubria, Varese 21100, Italy
| | - Pragatheiswar Giri
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Elisabetta Sieni
- Department of Theoretical and Applied Sciences, University of Insubria, Varese 21100, Italy.
| |
Collapse
|
2
|
Dhas Y, Biswas N, M R D, Jones LD, Ashili S. Repurposing metabolic regulators: antidiabetic drugs as anticancer agents. MOLECULAR BIOMEDICINE 2024; 5:40. [PMID: 39333445 PMCID: PMC11436690 DOI: 10.1186/s43556-024-00204-z] [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: 05/10/2024] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Drug repurposing in cancer taps into the capabilities of existing drugs, initially designed for other ailments, as potential cancer treatments. It offers several advantages over traditional drug discovery, including reduced costs, reduced development timelines, and a lower risk of adverse effects. However, not all drug classes align seamlessly with a patient's condition or long-term usage. Hence, repurposing of chronically used drugs presents a more attractive option. On the other hand, metabolic reprogramming being an important hallmark of cancer paves the metabolic regulators as possible cancer therapeutics. This review emphasizes the importance and offers current insights into the repurposing of antidiabetic drugs, including metformin, sulfonylureas, sodium-glucose cotransporter 2 (SGLT2) inhibitors, dipeptidyl peptidase 4 (DPP-4) inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RAs), thiazolidinediones (TZD), and α-glucosidase inhibitors, against various types of cancers. Antidiabetic drugs, regulating metabolic pathways have gained considerable attention in cancer research. The literature reveals a complex relationship between antidiabetic drugs and cancer risk. Among the antidiabetic drugs, metformin may possess anti-cancer properties, potentially reducing cancer cell proliferation, inducing apoptosis, and enhancing cancer cell sensitivity to chemotherapy. However, other antidiabetic drugs have revealed heterogeneous responses. Sulfonylureas and TZDs have not demonstrated consistent anti-cancer activity, while SGLT2 inhibitors and DPP-4 inhibitors have shown some potential benefits. GLP-1RAs have raised concerns due to possible associations with an increased risk of certain cancers. This review highlights that further research is warranted to elucidate the mechanisms underlying the potential anti-cancer effects of these drugs and to establish their efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Yogita Dhas
- Rhenix Lifesciences, Hyderabad, 500038, Telangana, India
| | - Nupur Biswas
- Rhenix Lifesciences, Hyderabad, 500038, Telangana, India.
- CureScience, 5820 Oberlin Dr, Suite 202, San Diego, CA, 92121, USA.
| | | | - Lawrence D Jones
- CureScience, 5820 Oberlin Dr, Suite 202, San Diego, CA, 92121, USA
| | | |
Collapse
|
3
|
Asiri A, Al Qarni A, Bakillah A. The Interlinking Metabolic Association between Type 2 Diabetes Mellitus and Cancer: Molecular Mechanisms and Therapeutic Insights. Diagnostics (Basel) 2024; 14:2132. [PMID: 39410536 PMCID: PMC11475808 DOI: 10.3390/diagnostics14192132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 10/20/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) and cancer share common risk factors including obesity, inflammation, hyperglycemia, and hyperinsulinemia. High insulin levels activate the PI3K/Akt/mTOR signaling pathway promoting cancer cell growth, survival, proliferation, metastasis, and anti-apoptosis. The inhibition of the PI3K/Akt/mTOR signaling pathway for cancer remains a promising therapy; however, drug resistance poses a major problem in clinical settings resulting in limited efficacy of agents; thus, combination treatments with therapeutic inhibitors may solve the resistance to such agents. Understanding the metabolic link between diabetes and cancer can assist in improving the therapeutic strategies used for the management of cancer patients with diabetes and vice versa. This review provides an overview of shared molecular mechanisms between diabetes and cancer as well as discusses established and emerging therapeutic anti-cancer agents targeting the PI3K/Akt/mTOR pathway in cancer management.
Collapse
Affiliation(s)
- Abutaleb Asiri
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 36428, Saudi Arabia; (A.A.); (A.A.Q.)
- Division of Medical Research Core-A, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Ali Al Qarni
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 36428, Saudi Arabia; (A.A.); (A.A.Q.)
- Division of Medical Research Core-A, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Ahmed Bakillah
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 36428, Saudi Arabia; (A.A.); (A.A.Q.)
- Division of Medical Research Core-A, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| |
Collapse
|
4
|
Maimaitijiang A, He D, Li D, Li W, Su Z, Fan Z, Li J. Progress in Research of Nanotherapeutics for Overcoming Multidrug Resistance in Cancer. Int J Mol Sci 2024; 25:9973. [PMID: 39337463 PMCID: PMC11432649 DOI: 10.3390/ijms25189973] [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: 07/28/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Chemotherapy has been widely applied in oncotherapy. However, the development of multidrug resistance (MDR) has diminished the effectiveness of anticancer drugs against tumor cells. Such resistance often results in tumor recurrence, metastasis, and patient death. Fortunately, nanoparticle-based drug delivery systems provide a promising strategy by codelivery of multiple drugs and MDR reversal agents and the skillful, flexible, smart modification of drug targets. Such systems have demonstrated the ability to bypass the ABC transporter biological efflux mechanisms due to drug resistance. Hence, how to deliver drugs and exert potential antitumor effects have been successfully explored, applied, and developed. Furthermore, to overcome multidrug resistance, nanoparticle-based systems have been developed due to their good therapeutic effect, low side effects, and high tumor metastasis inhibition. In view of this, we systematically discuss the molecular mechanisms and therapeutic strategies of MDR from nanotherapeutics. Finally, we summarize intriguing ideas and future trends for further research in overcoming MDR.
Collapse
Affiliation(s)
- Ayitila Maimaitijiang
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dongze He
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dingyang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Wenfang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhengding Su
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhongxiong Fan
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
5
|
Sirtori CR, Castiglione S, Pavanello C. METFORMIN: FROM DIABETES TO CANCER TO PROLONGATION OF LIFE. Pharmacol Res 2024; 208:107367. [PMID: 39191336 DOI: 10.1016/j.phrs.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The metformin molecule dates back to over a century, but its clinical use started in the '50s. Since then, its use in diabetics has grown constantly, with over 150 million users today. The therapeutic profile also expanded, with improved understanding of novel mechanisms. Metformin has a major activity on insulin resistance, by acting on the insulin receptors and mitochondria, most likely by activation of the adenosine monophosphate-activated kinase. These and associated mechanisms lead to significant lipid lowering and body weight loss. An anti-cancer action has come up in recent years, with mechanisms partly dependent on the mitochondrial activity and also on phosphatidylinositol 3-kinase resistance occurring in some malignant tumors. The potential of metformin to raise life-length is the object of large ongoing studies and of several basic and clinical investigations. The present review article will attempt to investigate the basic mechanisms behind these diverse activities and the potential clinical benefits. Metformin may act on transcriptional activity by histone modification, DNA methylation and miRNAs. An activity on age-associated inflammation (inflammaging) may occur via activation of the nuclear factor erythroid 2 related factor and changes in gut microbiota. A senolytic activity, leading to reduction of cells with the senescent associated secretory phenotype, may be crucial in lifespan prolongation as well as in ancillary properties in age-associated diseases, such as Parkinson's disease. Telomere prolongation may be related to the activity on mitochondrial respiratory factor 1 and on peroxisome gamma proliferator coactivator 1-alpha. Very recent observations on the potential to act on the most severe neurological disorders, such as amyotrophic lateral sclerosis and frontotemporal dementia, have raised considerable hope.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Sofia Castiglione
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Pavanello
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
6
|
Sekino N, Kano M, Murakami K, Toyozumi T, Hayano K, Ohira G, Matsubara H. Current findings on the antitumor effects of metformin on esophageal squamous cell carcinoma (Review). Mol Clin Oncol 2024; 21:58. [PMID: 39006474 PMCID: PMC11240871 DOI: 10.3892/mco.2024.2756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 05/21/2024] [Indexed: 07/16/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is an intractable type of cancer that requires novel therapeutic modalities, since the therapeutic outcomes are often inadequate, even in response to multidisciplinary treatment. The antitumor effect of metformin, an antidiabetic drug, has been reported in esophageal cancer; however, its effects are diverse. Since various multidisciplinary therapies are used in ESCC, the antitumor effect of metformin is expected to be synergistic in some treatment strategies. The present review summarizes the antitumor effects of metformin and discusses its use in combination with existing therapies. The present study reviewed relevant studies where the molecular targets of metformin (AMPK and inflammatory system signals) were described, followed by the classification and organization of its antitumor effects, and subsequently summarized the current research on its antitumor effects, especially in ESCC. A number of studies have reported that metformin prevents the development of ESCC and exerts its antitumor effects through various pathways. In addition, metformin has been shown to inhibit tumor growth, induce apoptosis, inhibit cancer cell invasion, migration and angiogenesis into the tumor, and decrease tumor malignancy, such as metastasis. Furthermore, it may modulate host tumor immunity in a tumor-suppressive manner and is expected to improve prognosis following treatment for ESCC. Notably, metformin may be beneficial in combination with chemotherapy, such as cisplatin, and radiation. By contrast, it has been shown to potentially induce resistance to 5-fluorouracil. Finally, the effects of metformin in combination with other therapies are discussed in the present study, and perspectives on the potential benefits of metformin for future ESCC treatment are presented. In conclusion, the present review may be useful in improving the understanding of the wide range of antitumor effects of metformin. Although some concerning points remain, using metformin in ESCC treatment could be promising. Notably, more knowledge needs to be accumulated regarding the effects of metformin on ESCC.
Collapse
Affiliation(s)
- Nobufumi Sekino
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Masayuki Kano
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Kentaro Murakami
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Takeshi Toyozumi
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Koichi Hayano
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Gaku Ohira
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
7
|
Szemerédi N, Schelz Z, Horvath DA, Rácz B, Szatmári AG, Muddather HF, Bózsity N, Zupkó I, Spengler G. Impact of V9302, a Competitive Antagonist of Transmembrane Glutamine Flux on Reversal of Resistance in Breast Cancer Cell Lines. Pharmaceutics 2024; 16:877. [PMID: 39065573 PMCID: PMC11280048 DOI: 10.3390/pharmaceutics16070877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Chemotherapy is a known treatment modality that improves the long-term survival of breast cancer patients. However, due to the resistance to numerous anticancer drugs, alternative chemotherapeutic strategies are required. Regarding antimetabolic drugs, several compounds have proven anticancer properties, such as statins. The present study aimed to investigate the in vitro effects of V9302, a competitive antagonist of glutamine flux, on different subtypes of breast cancers (estrogen, progesterone, and HER2 receptor-positive or negative, and Pgp-negative and Pgp-overexpressing). The interactions of V9302 with standard chemotherapeutic drugs (doxorubicin and cisplatin) were also determined by MTT staining on breast cancer cell lines. Furthermore, the influence of V9302 on the cell cycle of MCF-7 and its Pgp-overexpressing counterpart KCR was monitored by flow cytometry. It was shown that V9302 exerted synergistic interactions with doxorubicin in all breast cancer cell lines. In cell cycle analysis, the KCR cell line was more sensitive to V9302. After 48 h, cell proliferation was completely blocked, and elevated G1, suppressed S, and decreased G2/M could be detected. Inhibition of glutamate transport can be assumed to block resistance related to Pgp.
Collapse
Affiliation(s)
- Nikoletta Szemerédi
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, 6725 Szeged, Hungary; (N.S.); (B.R.)
| | - Zsuzsanna Schelz
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös utca. 6, 6720 Szeged, Hungary; (Z.S.); (H.F.M.); (N.B.)
| | - Dária Antónia Horvath
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, 6725 Szeged, Hungary; (N.S.); (B.R.)
| | - Bálint Rácz
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, 6725 Szeged, Hungary; (N.S.); (B.R.)
| | - András G. Szatmári
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, 6725 Szeged, Hungary; (N.S.); (B.R.)
| | - Hiba F. Muddather
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös utca. 6, 6720 Szeged, Hungary; (Z.S.); (H.F.M.); (N.B.)
| | - Noémi Bózsity
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös utca. 6, 6720 Szeged, Hungary; (Z.S.); (H.F.M.); (N.B.)
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös utca. 6, 6720 Szeged, Hungary; (Z.S.); (H.F.M.); (N.B.)
| | - Gabriella Spengler
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, 6725 Szeged, Hungary; (N.S.); (B.R.)
| |
Collapse
|
8
|
Al-Odat OS, Nelson E, Budak-Alpdogan T, Jonnalagadda SC, Desai D, Pandey MK. Discovering Potential in Non-Cancer Medications: A Promising Breakthrough for Multiple Myeloma Patients. Cancers (Basel) 2024; 16:2381. [PMID: 39001443 PMCID: PMC11240591 DOI: 10.3390/cancers16132381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
MM is a common type of cancer that unfortunately leads to a significant number of deaths each year. The majority of the reported MM cases are detected in the advanced stages, posing significant challenges for treatment. Additionally, all MM patients eventually develop resistance or experience relapse; therefore, advances in treatment are needed. However, developing new anti-cancer drugs, especially for MM, requires significant financial investment and a lengthy development process. The study of drug repurposing involves exploring the potential of existing drugs for new therapeutic uses. This can significantly reduce both time and costs, which are typically a major concern for MM patients. The utilization of pre-existing non-cancer drugs for various myeloma treatments presents a highly efficient and cost-effective strategy, considering their prior preclinical and clinical development. The drugs have shown promising potential in targeting key pathways associated with MM progression and resistance. Thalidomide exemplifies the success that can be achieved through this strategy. This review delves into the current trends, the challenges faced by conventional therapies for MM, and the importance of repurposing drugs for MM. This review highlights a noncomprehensive list of conventional therapies that have potentially significant anti-myeloma properties and anti-neoplastic effects. Additionally, we offer valuable insights into the resources that can help streamline and accelerate drug repurposing efforts in the field of MM.
Collapse
Affiliation(s)
- Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (O.S.A.-O.); (E.N.)
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA;
| | - Emily Nelson
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (O.S.A.-O.); (E.N.)
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA;
| | | | | | - Dhimant Desai
- Department of Pharmacology, Penn State Neuroscience Institute, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (O.S.A.-O.); (E.N.)
| |
Collapse
|
9
|
Al Assi A, Posty S, Lamarche F, Chebel A, Guitton J, Cottet-Rousselle C, Prudent R, Lafanechère L, Giraud S, Dallemagne P, Suzanne P, Verney A, Genestier L, Castets M, Fontaine E, Billaud M, Cordier-Bussat M. A novel inhibitor of the mitochondrial respiratory complex I with uncoupling properties exerts potent antitumor activity. Cell Death Dis 2024; 15:311. [PMID: 38697987 PMCID: PMC11065874 DOI: 10.1038/s41419-024-06668-9] [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: 07/06/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
Cancer cells are highly dependent on bioenergetic processes to support their growth and survival. Disruption of metabolic pathways, particularly by targeting the mitochondrial electron transport chain complexes (ETC-I to V) has become an attractive therapeutic strategy. As a result, the search for clinically effective new respiratory chain inhibitors with minimized adverse effects is a major goal. Here, we characterize a new OXPHOS inhibitor compound called MS-L6, which behaves as an inhibitor of ETC-I, combining inhibition of NADH oxidation and uncoupling effect. MS-L6 is effective on both intact and sub-mitochondrial particles, indicating that its efficacy does not depend on its accumulation within the mitochondria. MS-L6 reduces ATP synthesis and induces a metabolic shift with increased glucose consumption and lactate production in cancer cell lines. MS-L6 either dose-dependently inhibits cell proliferation or induces cell death in a variety of cancer cell lines, including B-cell and T-cell lymphomas as well as pediatric sarcoma. Ectopic expression of Saccharomyces cerevisiae NADH dehydrogenase (NDI-1) partially restores the viability of B-lymphoma cells treated with MS-L6, demonstrating that the inhibition of NADH oxidation is functionally linked to its cytotoxic effect. Furthermore, MS-L6 administration induces robust inhibition of lymphoma tumor growth in two murine xenograft models without toxicity. Thus, our data present MS-L6 as an inhibitor of OXPHOS, with a dual mechanism of action on the respiratory chain and with potent antitumor properties in preclinical models, positioning it as the pioneering member of a promising drug class to be evaluated for cancer therapy. MS-L6 exerts dual mitochondrial effects: ETC-I inhibition and uncoupling of OXPHOS. In cancer cells, MS-L6 inhibited ETC-I at least 5 times more than in isolated rat hepatocytes. These mitochondrial effects lead to energy collapse in cancer cells, resulting in proliferation arrest and cell death. In contrast, hepatocytes which completely and rapidly inactivated this molecule, restored their energy status and survived exposure to MS-L6 without apparent toxicity.
Collapse
Affiliation(s)
- Alaa Al Assi
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Solène Posty
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France
| | - Frédéric Lamarche
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Amel Chebel
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Jérôme Guitton
- Laboratoire de biochimie et pharmacologie-toxicologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, F-69495, Pierre Bénite, France. Laboratoire de Toxicologie, Faculté de pharmacie ISPBL, Université Lyon 1, 69373, Lyon, France
| | - Cécile Cottet-Rousselle
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Renaud Prudent
- Université Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence Lafanechère
- Université Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Stéphane Giraud
- Center for Drug Discovery and Development, Synergie Lyon Cancer Foundation, Lyon, Cancer Research Center, Centre Léon Bérard, Lyon, France
| | | | - Peggy Suzanne
- Normandie Univ., UNICAEN, CERMN, 14000, Caen, France
| | - Aurélie Verney
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Laurent Genestier
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Marie Castets
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France
| | - Eric Fontaine
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France.
| | - Marc Billaud
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France.
| | - Martine Cordier-Bussat
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France.
| |
Collapse
|
10
|
Kawakita E, Kanasaki K. Cancer biology in diabetes update: Focusing on antidiabetic drugs. J Diabetes Investig 2024; 15:525-540. [PMID: 38456597 PMCID: PMC11060166 DOI: 10.1111/jdi.14152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/25/2023] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
The association of type 2 diabetes with certain cancer risk has been of great interest for years. However, the effect of diabetic medications on cancer development is not fully understood. Prospective clinical trials have not elucidated the long-term influence of hypoglycemic drugs on cancer incidence and the safety for cancer-bearing patients with diabetes, whereas numerous preclinical studies have shown that antidiabetic drugs could have an impact on carcinogenesis processes beyond the glycemic control effect. Because there is no evidence of the safety profile of antidiabetic agents on cancer biology, careful consideration would be required when prescribing any medicines to patients with diabetes and existing tumor. In this review, we discuss the potential influence of each diabetes therapy in cancer 'initiation', 'promotion' and 'progression'.
Collapse
Affiliation(s)
- Emi Kawakita
- Department of Internal Medicine 1, Faculty of MedicineShimane UniversityIzumoJapan
| | - Keizo Kanasaki
- Department of Internal Medicine 1, Faculty of MedicineShimane UniversityIzumoJapan
- The Center for Integrated Kidney Research and Advance, Faculty of MedicineShimane UniversityIzumoJapan
| |
Collapse
|
11
|
Mesquita LA, Spiazzi BF, Piccoli GF, Nogara DA, da Natividade GR, Garbin HI, Wayerbacher LF, Wiercinski VM, Baggio VA, Zingano CP, Schwartsmann G, Lopes G, Petrie JR, Colpani V, Gerchman F. Does metformin reduce the risk of cancer in obesity and diabetes? A systematic review and meta-analysis. Diabetes Obes Metab 2024; 26:1929-1940. [PMID: 38389430 DOI: 10.1111/dom.15509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024]
Abstract
AIM To evaluate the effect of metformin on cancer incidence in subjects with overweight/obesity and/or prediabetes/diabetes. MATERIALS AND METHODS We searched MEDLINE, Embase and CENTRAL for randomized controlled trials (RCTs) in adults with overweight/obesity and/or prediabetes/diabetes that compared metformin to other interventions for ≥24 weeks. Independent reviewers selected and extracted data including population and intervention characteristics and new diagnoses of cancer. We used the RoB 2.0 risk-of-bias tool and the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) framework to assess risk of bias and certainty of evidence. RESULTS From 14 895 records after removal of duplicates, 27 trials were included, providing a total of 10 717 subjects in the metformin group and 10 003 in the control group, with 170 and 208 new cases of cancer, respectively. Using a random-effects model, the relative risk was 1.07 (95% confidence interval 0.87-1.31), with similar results in subgroup analyses by study duration or effect of control intervention on weight. Risk of bias in most studies was low, and no evidence of publication bias was found. Trial sequential analysis provided evidence that the cumulative sample size was large enough to exclude a significant effect of metformin on cancer incidence. CONCLUSIONS Metformin did not reduce cancer incidence in RCTs involving subjects with overweight/obesity and/or prediabetes/diabetes.
Collapse
Affiliation(s)
- Leonardo A Mesquita
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Critical Care Medicine, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Bernardo F Spiazzi
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Giovana F Piccoli
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Daniela A Nogara
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gabriella R da Natividade
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Henrique I Garbin
- Division of Cardiology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Laura F Wayerbacher
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Vanessa M Wiercinski
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Viviane A Baggio
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carolina P Zingano
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilberto Schwartsmann
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilberto Lopes
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - John R Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Verônica Colpani
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando Gerchman
- Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Endocrinology and Metabolism, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
12
|
Salehi AM, Wang L, Gu X, Coates PJ, Norberg Spaak L, Sgaramella N, Nylander K. Patients with oral tongue squamous cell carcinoma and co‑existing diabetes exhibit lower recurrence rates and improved survival: Implications for treatment. Oncol Lett 2024; 27:142. [PMID: 38385115 PMCID: PMC10877229 DOI: 10.3892/ol.2024.14275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Locoregional recurrences and distant metastases are major problems for patients with squamous cell carcinoma of the head and neck (SCCHN). Because SCCHN is a heterogeneous group of tumours with varying characteristics, the present study concentrated on the subgroup of squamous cell carcinoma of the oral tongue (SCCOT) to investigate the use of machine learning approaches to predict the risk of recurrence from routine clinical data available at diagnosis. The approach also identified the most important parameters that identify and classify recurrence risk. A total of 66 patients with SCCOT were included. Clinical data available at diagnosis were analysed using statistical analysis and machine learning approaches. Tumour recurrence was associated with T stage (P=0.001), radiological neck metastasis (P=0.010) and diabetes (P=0.003). A machine learning model based on the random forest algorithm and with attendant explainability was used. Whilst patients with diabetes were overrepresented in the SCCOT cohort, diabetics had lower recurrence rates (P=0.015 after adjusting for age and other clinical features) and an improved 2-year survival (P=0.025) compared with non-diabetics. Clinical, radiological and histological data available at diagnosis were used to establish a prognostic model for patients with SCCOT. Using machine learning to predict recurrence produced a classification model with 71.2% accuracy. Notably, one of the findings of the feature importance rankings of the model was that diabetics exhibited less recurrence and improved survival compared with non-diabetics, even after accounting for the independent prognostic variables of tumour size and patient age at diagnosis. These data imply that the therapeutic manipulation of glucose levels used to treat diabetes may be useful for patients with SCCOT regardless of their diabetic status. Further studies are warranted to investigate the impact of diabetes in other SCCHN subtypes.
Collapse
Affiliation(s)
- Amir M. Salehi
- Department of Medical Biosciences/Pathology, Umeå University, SE 901 85 Umeå, Sweden
| | - Lixiao Wang
- Department of Medical Biosciences/Pathology, Umeå University, SE 901 85 Umeå, Sweden
| | - Xiaolian Gu
- Department of Medical Biosciences/Pathology, Umeå University, SE 901 85 Umeå, Sweden
| | - Philip J. Coates
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno 656 53, Czech Republic
| | - Lena Norberg Spaak
- Department of Medical Biosciences/Pathology, Umeå University, SE 901 85 Umeå, Sweden
| | - Nicola Sgaramella
- Department of Medical Biosciences/Pathology, Umeå University, SE 901 85 Umeå, Sweden
- Department of Oral and Maxillo-Facial Surgery, Mater Dei Hospital, I-70125 Bari, Italy
| | - Karin Nylander
- Department of Medical Biosciences/Pathology, Umeå University, SE 901 85 Umeå, Sweden
| |
Collapse
|
13
|
Bosso M, Haddad D, Al Madhoun A, Al-Mulla F. Targeting the Metabolic Paradigms in Cancer and Diabetes. Biomedicines 2024; 12:211. [PMID: 38255314 PMCID: PMC10813379 DOI: 10.3390/biomedicines12010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Dysregulated metabolic dynamics are evident in both cancer and diabetes, with metabolic alterations representing a facet of the myriad changes observed in these conditions. This review delves into the commonalities in metabolism between cancer and type 2 diabetes (T2D), focusing specifically on the contrasting roles of oxidative phosphorylation (OXPHOS) and glycolysis as primary energy-generating pathways within cells. Building on earlier research, we explore how a shift towards one pathway over the other serves as a foundational aspect in the development of cancer and T2D. Unlike previous reviews, we posit that this shift may occur in seemingly opposing yet complementary directions, akin to the Yin and Yang concept. These metabolic fluctuations reveal an intricate network of underlying defective signaling pathways, orchestrating the pathogenesis and progression of each disease. The Warburg phenomenon, characterized by the prevalence of aerobic glycolysis over minimal to no OXPHOS, emerges as the predominant metabolic phenotype in cancer. Conversely, in T2D, the prevailing metabolic paradigm has traditionally been perceived in terms of discrete irregularities rather than an OXPHOS-to-glycolysis shift. Throughout T2D pathogenesis, OXPHOS remains consistently heightened due to chronic hyperglycemia or hyperinsulinemia. In advanced insulin resistance and T2D, the metabolic landscape becomes more complex, featuring differential tissue-specific alterations that affect OXPHOS. Recent findings suggest that addressing the metabolic imbalance in both cancer and diabetes could offer an effective treatment strategy. Numerous pharmaceutical and nutritional modalities exhibiting therapeutic effects in both conditions ultimately modulate the OXPHOS-glycolysis axis. Noteworthy nutritional adjuncts, such as alpha-lipoic acid, flavonoids, and glutamine, demonstrate the ability to reprogram metabolism, exerting anti-tumor and anti-diabetic effects. Similarly, pharmacological agents like metformin exhibit therapeutic efficacy in both T2D and cancer. This review discusses the molecular mechanisms underlying these metabolic shifts and explores promising therapeutic strategies aimed at reversing the metabolic imbalance in both disease scenarios.
Collapse
Affiliation(s)
- Mira Bosso
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| |
Collapse
|
14
|
Welponer T, Weber DD, Trattner L, Tockner B, Aminzadeh-Gohari S, Leb-Reichl V, Kaufmann A, Zauner R, Wimmer M, Wally V, Felder TK, Strunk D, Koller U, Bauer JW, Kofler B, Guttmann-Gruber C, Piñon Hofbauer J. Metformin shows anti-neoplastic properties by inhibition of oxidative phosphorylation and glycolysis in epidermolysis bullosa-associated aggressive cutaneous squamous cell carcinoma. J Eur Acad Dermatol Venereol 2024; 38:112-123. [PMID: 37669776 DOI: 10.1111/jdv.19488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/18/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND While most cutaneous squamous cell carcinomas (cSCCs) are treatable, certain high-risk cSCCs, such as those in recessive dystrophic epidermolysis bullosa (RDEB) patients, are particularly aggressive. Owing to repeated wounding, inflammation and unproductive healing, RDEB patients have a 68% cumulative risk of developing life-threatening cSCCs by the age of 35, and a 70% risk of death by the age of 45. Despite aggressive treatment, cSCC represents the leading cause of premature mortality in these patients, highlighting an unmet clinical need. Increasing evidence points to a role of altered metabolism in the initiation and maintenance of cSCC, making metabolism a potential therapeutic target. OBJECTIVES We sought to determine the feasibility of targeting tumour cell energetics as a strategy to selectively hinder the growth advantage of aggressive cSCC. METHODS We evaluated the cell energetics profiles of RDEB-SCC cells by analysing available gene expression data against multiple gene signatures and single-gene targets linked to metabolic reprogramming. Additionally, we employed real-time metabolic profiling to measure glycolysis and respiration in these cells. Furthermore, we investigated the anti-neoplastic properties of the metformin against human and murine high-risk cSCCs in vitro and in vivo. RESULTS Gene expression analyses highlighted a divergence in cell energetics profiles between RDEB-SCC and non-malignant RDEB keratinocytes, with tumour cells demonstrating enhanced respiration and glycolysis scores. Real-time metabolic profiling supported these data and additionally highlighted a metabolic plasticity of RDEB-SCC cells. Against this background, metformin exerted an anti-neoplastic potential by hampering both respiration and glycolysis, and by inhibiting proliferation in vitro. Metformin treatment in an analogous model of fast-growing murine cSCC resulted in delayed tumour onset and slower tumour growth, translating to a 29% increase in median overall survival. CONCLUSIONS Our data indicate that metformin exerts anti-neoplastic properties in aggressive cSCCs that exhibit high-risk features by interfering with respiration and glycolytic processes.
Collapse
Affiliation(s)
- T Welponer
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - D D Weber
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - L Trattner
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - B Tockner
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - S Aminzadeh-Gohari
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - V Leb-Reichl
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - A Kaufmann
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - R Zauner
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - M Wimmer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - V Wally
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - T K Felder
- Department of Laboratory Medicine, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - D Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - U Koller
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - J W Bauer
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - B Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - C Guttmann-Gruber
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - J Piñon Hofbauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
15
|
Cavalluzzi MM, Viale M, Rotondo NP, Ferraro V, Lentini G. Drug Repositioning for Ovarian Cancer Treatment: An Update. Anticancer Agents Med Chem 2024; 24:637-647. [PMID: 38367265 DOI: 10.2174/0118715206282904240122063914] [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: 10/14/2023] [Revised: 12/28/2023] [Accepted: 01/06/2024] [Indexed: 02/19/2024]
Abstract
Ovarian cancer (OC) is one of the most prevalent malignancies in female reproductive organs, and its 5-year survival is below 45%. Despite the advances in surgical and chemotherapeutic options, OC treatment is still a challenge, and new anticancer agents are urgently needed. Drug repositioning has gained significant attention in drug discovery, representing a smart way to identify new clinical applications for drugs whose human safety and pharmacokinetics have already been established, with great time and cost savings in pharmaceutical development endeavors. This review offers an update on the most promising drugs repurposable for OC treatment and/or prevention.
Collapse
Affiliation(s)
| | - Maurizio Viale
- U.O.C. Bioterapie, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Valeria Ferraro
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Lentini
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
16
|
Bartolucci G, Pallecchi M, Braconi L, Dei S, Teodori E, Lapolla A, Sartore G, Traldi P. Mass Spectrometry Study about In Vitro and In Vivo Reaction between Metformin and Glucose: A Preliminary Investigation on Alternative Biological Behavior. Int J Mol Sci 2023; 25:180. [PMID: 38203351 PMCID: PMC10779030 DOI: 10.3390/ijms25010180] [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: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Metformin is the most prescribed glucose-lowering drug worldwide; globally, over 100 million patients are prescribed this drug annually. Some different action mechanisms have been proposed for this drug, but, surprisingly, no metabolite of metformin has ever been described. It was considered interesting to investigate the possible reaction of metformin with glucose following the Maillard reaction pattern. The reaction was first performed in in vitro conditions, showing the formation of two adducts that originated by the condensation of the two molecular species with the losses of one or two water molecules. Their structures were investigated by liquid chromatography coupled with mass spectrometry (HPLC-MS), tandem mass spectrometry (MS/MS) and accurate mass measurements (HRMS). The species originated via the reaction of glucose and metformin and were called metformose and dehydrometformose, and some structural hypotheses were conducted. It is worth to emphasize that they were detected in urine samples from a diabetic patient treated with metformin and consequently they must be considered metabolites of the drug, which has never been identified before now. The glucose-related substructure of these compounds could reflect an improved transfer across cell membranes and, consequently, new hypotheses could be made about the biological targets of metformin.
Collapse
Affiliation(s)
- Gianluca Bartolucci
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Università di Firenze, 50100 Firenze, Italy; (M.P.); (L.B.); (S.D.); (E.T.)
| | - Marco Pallecchi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Università di Firenze, 50100 Firenze, Italy; (M.P.); (L.B.); (S.D.); (E.T.)
| | - Laura Braconi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Università di Firenze, 50100 Firenze, Italy; (M.P.); (L.B.); (S.D.); (E.T.)
| | - Silvia Dei
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Università di Firenze, 50100 Firenze, Italy; (M.P.); (L.B.); (S.D.); (E.T.)
| | - Elisabetta Teodori
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino (NEUROFARBA), Università di Firenze, 50100 Firenze, Italy; (M.P.); (L.B.); (S.D.); (E.T.)
| | - Annunziata Lapolla
- Dipartimento di Medicina, Università di Padova, 35100 Padova, Italy; (A.L.); (G.S.)
| | - Giovanni Sartore
- Dipartimento di Medicina, Università di Padova, 35100 Padova, Italy; (A.L.); (G.S.)
| | - Pietro Traldi
- Istituto di Ricerca Pediatrica Città della Speranza, 35100 Padova, Italy
| |
Collapse
|
17
|
Ruan G, Wu F, Shi D, Sun H, Wang F, Xu C. Metformin: update on mechanisms of action on liver diseases. Front Nutr 2023; 10:1327814. [PMID: 38192642 PMCID: PMC10773879 DOI: 10.3389/fnut.2023.1327814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
Substantial attention has been paid to the various effects of metformin on liver diseases; the liver is the targeted organ where metformin exerts its antihyperglycemic properties. In non-alcoholic fatty liver disease (NAFLD), studies have shown that metformin affects the ATP/AMP ratio to activate AMPK, subsequently governing lipid metabolism. The latest research showed that low-dose metformin targets the lysosomal AMPK pathway to decrease hepatic triglyceride levels through the PEN2-ATP6AP1 axis in an AMP-independent manner. Metformin regulates caspase-3, eukaryotic initiation factor-2a (eIF2a), and insulin receptor substrate-1 (IRS-1) in palmitate-exposed HepG2 cells, alleviating endoplasmic reticulum (ER) stress. Recent observations highlighted the critical association with intestinal flora, as confirmed by the finding that metformin decreased the relative abundance of Bacteroides fragilis while increasing Akkermansia muciniphila and Bifidobacterium bifidum. The suppression of intestinal farnesoid X receptor (FXR) and the elevation of short-chain fatty acids resulted in the upregulation of tight junction protein and the alleviation of hepatic inflammation induced by lipopolysaccharide (LPS). Additionally, metformin delayed the progression of cirrhosis by regulating the activation and proliferation of hepatic stellate cells (HSCs) via the TGF-β1/Smad3 and succinate-GPR91 pathways. In hepatocellular carcinoma (HCC), metformin impeded the cell cycle and enhanced the curative effect of antitumor medications. Moreover, metformin protects against chemical-induced and drug-induced liver injury (DILI) against hepatotoxic drugs. These findings suggest that metformin may have pharmacological efficacy against liver diseases.
Collapse
Affiliation(s)
- Gaoyi Ruan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangquan Wu
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, China
| | - Dibang Shi
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongxia Sun
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, China
| | - Changlong Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
18
|
Pagoni M, Cava C, Sideris DC, Avgeris M, Zoumpourlis V, Michalopoulos I, Drakoulis N. miRNA-Based Technologies in Cancer Therapy. J Pers Med 2023; 13:1586. [PMID: 38003902 PMCID: PMC10672431 DOI: 10.3390/jpm13111586] [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: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
The discovery of therapeutic miRNAs is one of the most exciting challenges for pharmaceutical companies. Since the first miRNA was discovered in 1993, our knowledge of miRNA biology has grown considerably. Many studies have demonstrated that miRNA expression is dysregulated in many diseases, making them appealing tools for novel therapeutic approaches. This review aims to discuss miRNA biogenesis and function, as well as highlight strategies for delivering miRNA agents, presenting viral, non-viral, and exosomic delivery as therapeutic approaches for different cancer types. We also consider the therapeutic role of microRNA-mediated drug repurposing in cancer therapy.
Collapse
Affiliation(s)
- Maria Pagoni
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15701 Athens, Greece
| | - Claudia Cava
- Department of Science, Technology and Society, University School for Advanced Studies IUSS Pavia, 27100 Pavia, Italy;
| | - Diamantis C. Sideris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece;
| | - Margaritis Avgeris
- Laboratory of Clinical Biochemistry—Molecular Diagnostics, Second Department of Pediatrics, School of Medicine, “P. & A. Kyriakou” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassilios Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece;
| | - Ioannis Michalopoulos
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece;
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15701 Athens, Greece
| |
Collapse
|
19
|
Satpathy C, Kumar Mishra T, Singh S, Jha AK. Reverse cardio-oncology: A budding concept. Indian Heart J 2023; 75:398-402. [PMID: 37774949 PMCID: PMC10774571 DOI: 10.1016/j.ihj.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023] Open
Abstract
Having established the significance of cardiovascular side-effects of anti-neoplastic drugs, present day cardio-oncology has forayed into newer territories buoyed by research into the multiple connections that exist between cardiovascular disease and cancer. An emerging concept of reverse cardio-oncology focuses on the heightened risk of cancer in patients with cardiovascular disease. Common mechanistics of cancer and heart failure (HF) like chronic inflammation and clonal haematopoesis as well as common predisposing factors like obesity and diabetes underline the relation between both cardiovascular disease and various cancers.This review discusses the potential magnitude of the problem, the underlying pathophysiological mechanisms and classification of this novel subject.
Collapse
Affiliation(s)
- Chhabi Satpathy
- Department of Cardiology, MKCG Medical College and Hospital, Berhampur, Odisha, India
| | - Trinath Kumar Mishra
- Department of Cardiology, MKCG Medical College and Hospital, Berhampur, Odisha, India.
| | - Subhasish Singh
- Department of Cardiology, MKCG Medical College and Hospital, Berhampur, Odisha, India
| | - Anshu Kumar Jha
- Department of Cardiology, MKCG Government Medical College, Berhampur, Odisha, India
| |
Collapse
|
20
|
Li H, Chen Y, Hu L, Yang W, Gao Z, Liu M, Tao H, Li J. Will metformin use lead to a decreased risk of thyroid cancer? A systematic review and meta-analyses. Eur J Med Res 2023; 28:392. [PMID: 37773165 PMCID: PMC10542235 DOI: 10.1186/s40001-023-01287-0] [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: 05/09/2023] [Accepted: 08/12/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND It has been reported that metformin use may reduce the risk of thyroid cancer, but existing studies have generated inconsistent results. The purpose of this study was to investigate such association between metformin use and the risk of thyroid cancer. METHODS Studies of metformin use for the risk of thyroid cancer were searched in Web of Science, PubMed, Embase, Cochrane Library, China National Knowledge Infrastructure, China Biomedical Database, Wanfang Data, and Chinese Scientific Journals Database (VIP) from the establishment date to December 2022. Newcastle-Ottawa scale is adopted for assessing the methodological quality of included studies, and the inter-study heterogeneity was assessed by using the I-squared statistic. Combined odds ratios (ORs) with the corresponding 95% confidence intervals (CIs) were calculated through either fixed-effects or random-effects model according to the heterogeneity. Besides, subgroup analyses, sensitivity analyses and test for publication bias were conducted. RESULTS Five studies involving 1,713,528 participants were enrolled in the qualitative and quantitative synthesis. The result of the meta-analyses showed that metformin use was associated with a statistically significant lower risk of thyroid cancer (pooled OR = 0.68, 95% CI = 0.50-0.91, P = 0.011). Moreover, in the subgroup analysis, we found that the use of metformin may also aid in the prevention of thyroid cancer in Eastern population (pooled OR = 0.55, 95% CI = 0.35-0.88, P = 0.012) rather than Western population (pooled OR = 0.89, 95% CI = 0.52-1.54, P = 0.685). Sensitivity analysis suggested the results of this meta-analyses were relatively stable. No publication bias was detected. CONCLUSION Metformin use is beneficial for reducing the risk of thyroid cancer. For further investigation, more well-designed studies are still needed to elucidate the association between metformin use and the risk of thyroid cancer.
Collapse
Affiliation(s)
- Hailong Li
- Department of Clinical Medicine, Sun Yat-Sen University, No.74 Nonglin Road, Guangzhou, 510030, Guangdong, People's Republic of China
| | - Yue Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Lei Hu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Wenzhi Yang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Zongshi Gao
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China
| | - Mengqing Liu
- Department of Clinical Medicine, School of Chaohu Clinical Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Hui Tao
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China.
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China.
| | - Jie Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China.
| |
Collapse
|
21
|
Dickerman BA, García-Albéniz X, Logan RW, Denaxas S, Hernán MA. Evaluating Metformin Strategies for Cancer Prevention: A Target Trial Emulation Using Electronic Health Records. Epidemiology 2023; 34:690-699. [PMID: 37227368 PMCID: PMC10524586 DOI: 10.1097/ede.0000000000001626] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
BACKGROUND Metformin users appear to have a substantially lower risk of cancer than nonusers in many observational studies. These inverse associations may be explained by common flaws in observational analyses that can be avoided by explicitly emulating a target trial. METHODS We emulated target trials of metformin therapy and cancer risk using population-based linked electronic health records from the UK (2009-2016). We included individuals with diabetes, no history of cancer, no recent prescription for metformin or other glucose-lowering medication, and hemoglobin A1c (HbA1c) <64 mmol/mol (<8.0%). Outcomes included total cancer and 4 site-specific cancers (breast, colorectal, lung, and prostate). We estimated risks using pooled logistic regression with adjustment for risk factors via inverse-probability weighting. We emulated a second target trial among individuals regardless of diabetes status. We compared our estimates with those obtained using previously applied analytic approaches. RESULTS Among individuals with diabetes, the estimated 6-year risk differences (metformin - no metformin) were -0.2% (95% CI = -1.6%, 1.3%) in the intention-to-treat analysis and 0.0% (95% CI = -2.1%, 2.3%) in the per-protocol analysis. The corresponding estimates for all site-specific cancers were close to zero. Among individuals regardless of diabetes status, these estimates were also close to zero and more precise. By contrast, previous analytic approaches yielded estimates that appeared strongly protective. CONCLUSIONS Our findings are consistent with the hypothesis that metformin therapy does not meaningfully influence cancer incidence. The findings highlight the importance of explicitly emulating a target trial to reduce bias in the effect estimates derived from observational analyses.
Collapse
Affiliation(s)
- Barbra A. Dickerman
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
| | - Xabier García-Albéniz
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
- RTI Health Solutions, Barcelona, Spain
| | - Roger W. Logan
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
| | - Spiros Denaxas
- Institute of Health Informatics Research, University
College London, London, UK
- Health Data Research UK (HDR UK) London, University College
London, London, UK
- The Alan Turing Institute, London, UK
| | - Miguel A. Hernán
- CAUSALab, Harvard T.H. Chan School of Public Health,
Boston, Massachusetts, US
- Department of Epidemiology, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
- Department of Biostatistics, Harvard T.H. Chan School of
Public Health, Boston, Massachusetts, US
| |
Collapse
|
22
|
Abdelmoneim M, Aboalela MA, Naoe Y, Matsumura S, Eissa IR, Bustos-Villalobos I, Sibal PA, Takido Y, Kodera Y, Kasuya H. The Impact of Metformin on Tumor-Infiltrated Immune Cells: Preclinical and Clinical Studies. Int J Mol Sci 2023; 24:13353. [PMID: 37686159 PMCID: PMC10487782 DOI: 10.3390/ijms241713353] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The tumor microenvironment (TME) plays a pivotal role in the fate of cancer cells, and tumor-infiltrating immune cells have emerged as key players in shaping this complex milieu. Cancer is one of the leading causes of death in the world. The most common standard treatments for cancer are surgery, radiation therapy, and chemotherapeutic drugs. In the last decade, immunotherapy has had a potential effect on the treatment of cancer patients with poor prognoses. One of the immune therapeutic targeted approaches that shows anticancer efficacy is a type 2 diabetes medication, metformin. Beyond its glycemic control properties, studies have revealed intriguing immunomodulatory properties of metformin. Meanwhile, several studies focus on the impact of metformin on tumor-infiltrating immune cells in various tumor models. In several tumor models, metformin can modulate tumor-infiltrated effector immune cells, CD8+, CD4+ T cells, and natural killer (NK) cells, as well as suppressor immune cells, T regulatory cells, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs). In this review, we discuss the role of metformin in modulating tumor-infiltrating immune cells in different preclinical models and clinical trials. Both preclinical and clinical studies suggest that metformin holds promise as adjunctive therapy in cancer treatment by modulating the immune response within the tumor microenvironment. Nonetheless, both the tumor type and the combined therapy have an impact on the specific targets of metformin in the TME. Further investigations are warranted to elucidate the precise mechanisms underlying the immunomodulatory effects of metformin and to optimize its clinical application in cancer patients.
Collapse
Affiliation(s)
- Mohamed Abdelmoneim
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; (M.A.A.); (I.R.E.)
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| | - Mona Alhussein Aboalela
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; (M.A.A.); (I.R.E.)
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| | - Yoshinori Naoe
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| | - Shigeru Matsumura
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| | - Ibrahim Ragab Eissa
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; (M.A.A.); (I.R.E.)
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| | - Itzel Bustos-Villalobos
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| | - Patricia Angela Sibal
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; (M.A.A.); (I.R.E.)
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| | - Yuhei Takido
- Department of Neurosurgery, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Yasuhiro Kodera
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; (M.A.A.); (I.R.E.)
| | - Hideki Kasuya
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan (S.M.)
| |
Collapse
|
23
|
Manna EDF, Serrano D, Aurilio G, Bonanni B, Lazzeroni M. Chemoprevention and Lifestyle Modifications for Risk Reduction in Sporadic and Hereditary Breast Cancer. Healthcare (Basel) 2023; 11:2360. [PMID: 37628558 PMCID: PMC10454363 DOI: 10.3390/healthcare11162360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Female breast cancer is the most commonly diagnosed malignancy worldwide. Risk assessment helps to identify women at increased risk of breast cancer and allows the adoption of a comprehensive approach to reducing breast cancer incidence through personalized interventions, including lifestyle modification, chemoprevention, intensified surveillance with breast imaging, genetic counseling, and testing. Primary prevention means acting on modifiable risk factors to reduce breast cancer occurrence. Chemoprevention with tamoxifen, raloxifene, anastrozole, and exemestane has already shown benefits in decreasing breast cancer incidence in women at an increased risk for breast cancer. For healthy women carrying BRCA 1 or BRCA 2 pathogenic/likely pathogenic (P/LP) germline variants, the efficacy of chemoprevention is still controversial. Adopting chemoprevention strategies and the choice among agents should depend on the safety profile and risk-benefit ratio. Unfortunately, the uptake of these agents has been low. Lifestyle modifications can reduce breast cancer incidence, and the recommendations for BRCA 1 or BRCA 2 P/LP germline variant carriers are comparable to the general population. This review summarizes the most recent evidence regarding the efficacy of chemoprevention and lifestyle interventions in women with sporadic and hereditary breast cancer.
Collapse
Affiliation(s)
- Eliza Del Fiol Manna
- Division of Cancer Prevention and Genetics, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.S.); (G.A.); (B.B.); (M.L.)
| | | | | | | | | |
Collapse
|
24
|
Yarmolinsky J, Bouras E, Constantinescu A, Burrows K, Bull CJ, Vincent EE, Martin RM, Dimopoulou O, Lewis SJ, Moreno V, Vujkovic M, Chang KM, Voight BF, Tsao PS, Gunter MJ, Hampe J, Pellatt AJ, Pharoah PDP, Schoen RE, Gallinger S, Jenkins MA, Pai RK, Gill D, Tsilidis KK. Genetically proxied glucose-lowering drug target perturbation and risk of cancer: a Mendelian randomisation analysis. Diabetologia 2023; 66:1481-1500. [PMID: 37171501 PMCID: PMC10317892 DOI: 10.1007/s00125-023-05925-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/13/2023] [Indexed: 05/13/2023]
Abstract
AIMS/HYPOTHESIS Epidemiological studies have generated conflicting findings on the relationship between glucose-lowering medication use and cancer risk. Naturally occurring variation in genes encoding glucose-lowering drug targets can be used to investigate the effect of their pharmacological perturbation on cancer risk. METHODS We developed genetic instruments for three glucose-lowering drug targets (peroxisome proliferator activated receptor γ [PPARG]; sulfonylurea receptor 1 [ATP binding cassette subfamily C member 8 (ABCC8)]; glucagon-like peptide 1 receptor [GLP1R]) using summary genetic association data from a genome-wide association study of type 2 diabetes in 148,726 cases and 965,732 controls in the Million Veteran Program. Genetic instruments were constructed using cis-acting genome-wide significant (p<5×10-8) SNPs permitted to be in weak linkage disequilibrium (r2<0.20). Summary genetic association estimates for these SNPs were obtained from genome-wide association study (GWAS) consortia for the following cancers: breast (122,977 cases, 105,974 controls); colorectal (58,221 cases, 67,694 controls); prostate (79,148 cases, 61,106 controls); and overall (i.e. site-combined) cancer (27,483 cases, 372,016 controls). Inverse-variance weighted random-effects models adjusting for linkage disequilibrium were employed to estimate causal associations between genetically proxied drug target perturbation and cancer risk. Co-localisation analysis was employed to examine robustness of findings to violations of Mendelian randomisation (MR) assumptions. A Bonferroni correction was employed as a heuristic to define associations from MR analyses as 'strong' and 'weak' evidence. RESULTS In MR analysis, genetically proxied PPARG perturbation was weakly associated with higher risk of prostate cancer (for PPARG perturbation equivalent to a 1 unit decrease in inverse rank normal transformed HbA1c: OR 1.75 [95% CI 1.07, 2.85], p=0.02). In histological subtype-stratified analyses, genetically proxied PPARG perturbation was weakly associated with lower risk of oestrogen receptor-positive breast cancer (OR 0.57 [95% CI 0.38, 0.85], p=6.45×10-3). In co-localisation analysis, however, there was little evidence of shared causal variants for type 2 diabetes liability and cancer endpoints in the PPARG locus, although these analyses were likely underpowered. There was little evidence to support associations between genetically proxied PPARG perturbation and colorectal or overall cancer risk or between genetically proxied ABCC8 or GLP1R perturbation with risk across cancer endpoints. CONCLUSIONS/INTERPRETATION Our drug target MR analyses did not find consistent evidence to support an association of genetically proxied PPARG, ABCC8 or GLP1R perturbation with breast, colorectal, prostate or overall cancer risk. Further evaluation of these drug targets using alternative molecular epidemiological approaches may help to further corroborate the findings presented in this analysis. DATA AVAILABILITY Summary genetic association data for select cancer endpoints were obtained from the public domain: breast cancer ( https://bcac.ccge.medschl.cam.ac.uk/bcacdata/ ); and overall prostate cancer ( http://practical.icr.ac.uk/blog/ ). Summary genetic association data for colorectal cancer can be accessed by contacting GECCO (kafdem at fredhutch.org). Summary genetic association data on advanced prostate cancer can be accessed by contacting PRACTICAL (practical at icr.ac.uk). Summary genetic association data on type 2 diabetes from Vujkovic et al (Nat Genet, 2020) can be accessed through dbGAP under accession number phs001672.v3.p1 (pha004945.1 refers to the European-specific summary statistics). UK Biobank data can be accessed by registering with UK Biobank and completing the registration form in the Access Management System (AMS) ( https://www.ukbiobank.ac.uk/enable-your-research/apply-for-access ).
Collapse
Affiliation(s)
- James Yarmolinsky
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Emmanouil Bouras
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Andrei Constantinescu
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kimberley Burrows
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Caroline J Bull
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- School of Translational Health Sciences, University of Bristol, Bristol, UK
| | - Emma E Vincent
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- School of Translational Health Sciences, University of Bristol, Bristol, UK
| | - Richard M Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, UK
| | - Olympia Dimopoulou
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sarah J Lewis
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Victor Moreno
- Biomarkers and Susceptibility Unit, Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Marijana Vujkovic
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Benjamin F Voight
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip S Tsao
- VA Palo Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Marc J Gunter
- Nutrition and Metabolism Section, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Jochen Hampe
- Department of Medicine I, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | | | - Paul D P Pharoah
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Robert E Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Steven Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, London, UK
| | - Kostas K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, London, UK
| |
Collapse
|
25
|
Chen Y, Mushashi F, Son S, Bhatti P, Dummer T, Murphy RA. Diabetes medications and cancer risk associations: a systematic review and meta-analysis of evidence over the past 10 years. Sci Rep 2023; 13:11844. [PMID: 37481610 PMCID: PMC10363143 DOI: 10.1038/s41598-023-38431-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 07/07/2023] [Indexed: 07/24/2023] Open
Abstract
Diabetes medications may modify the risk of certain cancers. We systematically searched MEDLINE, Embase, Web of Science, and Cochrane CENTRAL from 2011 to March 2021 for studies evaluating associations between diabetes medications and the risk of breast, lung, colorectal, prostate, liver, and pancreatic cancers. A total of 92 studies (3 randomized controlled trials, 64 cohort studies, and 25 case-control studies) were identified in the systematic review, involving 171 million participants. Inverse relationships with colorectal (n = 18; RR = 0.85; 95% CI = 0.78-0.92) and liver cancers (n = 10; RR = 0.55; 95% CI = 0.46-0.66) were observed in biguanide users. Thiazolidinediones were associated with lower risks of breast (n = 6; RR = 0.87; 95% CI = 0.80-0.95), lung (n = 6; RR = 0.77; 95% CI = 0.61-0.96) and liver (n = 8; RR = 0.83; 95% CI = 0.72-0.95) cancers. Insulins were negatively associated with breast (n = 15; RR = 0.90; 95% CI = 0.82-0.98) and prostate cancer risks (n = 7; RR = 0.74; 95% CI = 0.56-0.98). Positive associations were found between insulin secretagogues and pancreatic cancer (n = 5; RR = 1.26; 95% CI = 1.01-1.57), and between insulins and liver (n = 7; RR = 1.74; 95% CI = 1.08-2.80) and pancreatic cancers (n = 8; RR = 2.41; 95% CI = 1.08-5.36). Overall, biguanide and thiazolidinedione use carried no risk, or potentially lower risk of some cancers, while insulin secretagogue and insulin use were associated with increased pancreatic cancer risk.
Collapse
Affiliation(s)
- Yixian Chen
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | - Surim Son
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Parveen Bhatti
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
- Cancer Control Research, BC Cancer, Vancouver, British Columbia, Canada
| | - Trevor Dummer
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rachel A Murphy
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
- Cancer Control Research, BC Cancer, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Najafi F, Rajati F, Sarokhani D, Bavandpour M, Moradinazar M. The Relationship between Metformin Consumption and Cancer Risk: An Updated Umbrella Review of Systematic Reviews and Meta-Analyses. Int J Prev Med 2023; 14:90. [PMID: 37854987 PMCID: PMC10580191 DOI: 10.4103/ijpvm.ijpvm_62_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/15/2022] [Indexed: 10/20/2023] Open
Abstract
Background Considering that metformin is widely used in the treatment of diabetes, and its protective role against various malignancies, the strength and validity of the available evidence from related systematic reviews and meta-analysis were evaluated. Methods Scopus, PubMed, Embase, Cochrane, Web of science databases, and Google Scholar and manual screening of retrieved references were systematically searched from their inception dates to 24 March 2020 by extracting the effect size (Odds ratios (OR) and relative risk (RR) in each study. To present the forest plot of effect of metformin on each cancer, Stata version 14.2 was used. Results This study included 36 meta-analysis studies and 620 original research studies (26 randomized control trials studies and 594 observational studies (cohort, case-control)) covering 15 different cancers. Overall, metformin medication prevented different cancers, including ovarian cancer (OR = 0.76, 95% CI: 0.62,0.93), cervical cancer (OR = 0.60, 95% CI: 0.43, 0.83), endometrial cancer (OR = 1.05, 95% CI: 0.82,1.35), liver cancer (OR = 0.59, 95% CI: 0.47,0.74), pancreatic cancer (OR = 0.59, 95%CI 0.50,0.69), head and neck cancer (OR = 0.71, 95% CI: 0.61,0.83), stomach cancer (OR = 0.72, 95% CI: 0.26,1.99), colorectal cancer (OR = 0.73, 95% CI: 0.59,0.91), colorectal adenoma cancer (OR = 0.75, 95% CI: 0.65,0.86), colon cancer (OR = 0.79, 95% CI: 0.69,0.91), esophagus cancer (OR = 0.90, 95% CI: 0.83,0.98), lung cancer (OR = 0.92, CI95%:0.85,0.99), breast cancer (OR = 0.93, 95% CI: 0.84,1.02), prostate cancer (OR = 0.94, 95% CI: 0.85-1.04), and bladder cancer (OR = 0.94 95% CI: 0.64,1.38). Conclusions Treatment with metformin can significantly decrease the chance of all cancers with larger preventive effect on hepatocellular carcinoma and smaller preventive effect on lung and breast cancers.
Collapse
Affiliation(s)
- Farid Najafi
- Research Center for Environmental Determinants of Health, School of Public Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Behavioral Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Rajati
- Research Center for Environmental Determinants of Health, School of Public Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Behavioral Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Diana Sarokhani
- Behavioral Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Maryam Bavandpour
- Behavioral Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Moradinazar
- Research Center for Environmental Determinants of Health, School of Public Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Behavioral Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
27
|
Wahid Satar SNA, Norhayati MN, Yaacob LH. Recurrent hypoglycemia in a patient with hepatocellular carcinoma. ELECTRONIC JOURNAL OF GENERAL MEDICINE 2023. [DOI: 10.29333/ejgm/12903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Hypoglycemia is a common complication of diabetes mellitus. Patients may present with any kind of symptoms, either autonomic or neuroglycopenic. Here, we report a case of a 70-year-old man with type 2 diabetes mellitus with a newly diagnosed hepatocellular carcinoma presenting with recurrent episodes of hypoglycemia. The insulin treatment was withheld initially. However, the patient's diabetic control worsened after starting treatment for his hepatocellular carcinoma. Hence, the insulin treatment was resumed. This case highlighted the challenge in balancing the management of hypoglycaemia and poor diabetic control in a patient who has diabetes mellitus and hepatocellular carcinoma. Primary care practitioners need to have high clinical suspicion to monitor these patients to prevent hypoglycemia and manage them accordingly.
Collapse
Affiliation(s)
- Siti Nor Ain Wahid Satar
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, MALAYSIA
| | - Mohd Noor Norhayati
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, MALAYSIA
| | - Lili Husniati Yaacob
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, MALAYSIA
| |
Collapse
|
28
|
Roest S, Gürgöze MT, Cherikh WS, Stehlik J, Boersma EH, Zijlstra F, Manintveld OC. Influence of chronic kidney disease and other risk factors pre-heart transplantation on malignancy incidence post-heart transplantation. Front Cardiovasc Med 2023; 10:1145996. [PMID: 37077737 PMCID: PMC10106779 DOI: 10.3389/fcvm.2023.1145996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/06/2023] [Indexed: 04/05/2023] Open
Abstract
AimsChronic kidney disease (CKD) pre-heart transplantation (HTx) has been proposed as a risk factor for malignancy risk post-HTx. Using multicenter registry data, our aim was to calculate the death-adjusted annual incidence of malignancies post-HTx, corroborate the association between CKD pre-HTx and malignancy risk post-HTx, and determine other risk factors for post-HTx malignancies.Methods and materialsWe used data from patients transplanted in North American HTx centers between January 2000 and June 2017 and registered in the International Society for Heart and Lung Transplantation Thoracic Organ Transplant Registry. We excluded recipients with missing data on post-HTx malignancies, heterotopic heart transplant, retransplantation, multi-organ transplantation, and patients with a total artificial heart pre-HTx.ResultsOverall, 34,873 patients were included to determine the annual incidence of malignancies, 33,345 patients were included in the risk analyses. The incidence of any malignancy, solid-organ malignancy, post-transplant lymphoproliferative disease (PTLD), and skin cancer adjusted for death 15 years post-HTx, was 26.6%, 10.9%, 3.6%, and 15.8% respectively. Besides widely acknowledged risk factors, CKD stage ≥4 pre-HTx was associated with the development of all malignancies post-HTx (HR 1.17 compared to CKD stage 1, p = 0.023), as well as solid-organ malignancies (HR 1.35, p = 0.01), but not for PTLD (HR 0.73, p = 0.057), and skin cancer (HR 1.06, p = 0.59).ConclusionRisk of malignancy post-HTx remains high. CKD stages ≥4 pre-HTx was associated with an increased risk for any malignancy and solid-organ malignancy post-HTx. Strategies to mitigate the impact of pre-HTx patient factors on the risk of post-HTx malignancy are needed.
Collapse
Affiliation(s)
- Stefan Roest
- Department of Cardiology, Thorax Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Muhammed T. Gürgöze
- Department of Cardiology, Thorax Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Wida S. Cherikh
- United Network for Organ Sharing, Richmond, VA, United States
| | - Josef Stehlik
- Divisionof Cardiovascular Medicine, U.T.A.H. Cardiac Transplant Program, University of Utah Health Salt Lake City, Utah, UT, United States
| | - Eric H. Boersma
- Department of Cardiology, Thorax Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Felix Zijlstra
- Department of Cardiology, Thorax Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Olivier C. Manintveld
- Department of Cardiology, Thorax Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Correspondence: Olivier C. Manintveld
| |
Collapse
|
29
|
Hsu Y, Hsu CY, Kao YS. The Association between Metformin and the Cancer-Specific Mortality Rate in Nasopharyngeal Cancer Patients: Real-World Evidence. Curr Oncol 2023; 30:3940-3950. [PMID: 37185411 PMCID: PMC10136714 DOI: 10.3390/curroncol30040298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Objectives: Nasopharyngeal cancer is a common cancer in East and South Asia. The radiotherapy and chemotherapy regimen has advanced in recent years. However, many patients still suffer from local recurrence and distant metastasis; thus, identifying medication that can be combined with standard treatment to improve the treatment outcomes in nasopharyngeal cancer patients is an unmet need. Methods: We included nasopharyngeal cancer patients from the Taiwan National Health Insurance Database (NHIRD). The primary endpoint was set as the cancer-specific mortality rate. Metformin cohorts and non-Metformin cohorts were matched by sex, age, and the year of the index date. Propensity score matching with a ratio of 1:1 was applied. Results: A total of 6078 subjects were included in the study, with 3039 patients in each group. Male participants outnumbered female participants. Most of the patients were aged 50 to 64; the mean age was 60.4 ± 10.4 years in Metformin non-users, and that of Metformin users was 59.9 ± 10.5 years. Metformin users had a lower risk of death due to nasopharyngeal cancer (adjusted HR = 0.80; 95% CI = 0.71, 0.90) than controls. Conclusions: We concluded that Metformin might be effective at reducing the cancer-specific mortality rate in nasopharyngeal cancer patients. Further randomized control trials should be completed.
Collapse
|
30
|
Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15041192. [PMID: 36831534 PMCID: PMC9953833 DOI: 10.3390/cancers15041192] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer deaths among men in Western countries. Mitochondria, the "powerhouse" of cells, undergo distinctive metabolic and structural dynamics in different types of cancer. PCa cells experience peculiar metabolic changes during their progression from normal epithelial cells to early-stage and, progressively, to late-stage cancer cells. Specifically, healthy cells display a truncated tricarboxylic acid (TCA) cycle and inefficient oxidative phosphorylation (OXPHOS) due to the high accumulation of zinc that impairs the activity of m-aconitase, the enzyme of the TCA cycle responsible for the oxidation of citrate. During the early phase of cancer development, intracellular zinc levels decrease leading to the reactivation of m-aconitase, TCA cycle and OXPHOS. PCa cells change their metabolic features again when progressing to the late stage of cancer. In particular, the Warburg effect was consistently shown to be the main metabolic feature of late-stage PCa cells. However, accumulating evidence sustains that both the TCA cycle and the OXPHOS pathway are still present and active in these cells. The androgen receptor axis as well as mutations in mitochondrial genes involved in metabolic rewiring were shown to play a key role in PCa cell metabolic reprogramming. Mitochondrial structural dynamics, such as biogenesis, fusion/fission and mitophagy, were also observed in PCa cells. In this review, we focus on the mitochondrial metabolic and structural dynamics occurring in PCa during tumor development and progression; their role as effective molecular targets for novel therapeutic strategies in PCa patients is also discussed.
Collapse
|
31
|
Zhang Y, Zhou F, Guan J, Zhou L, Chen B. Action Mechanism of Metformin and Its Application in Hematological Malignancy Treatments: A Review. Biomolecules 2023; 13:250. [PMID: 36830619 PMCID: PMC9953052 DOI: 10.3390/biom13020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
Hematologic malignancies (HMs) mainly include acute and chronic leukemia, lymphoma, myeloma and other heterogeneous tumors that seriously threaten human life and health. The common effective treatments are radiotherapy, chemotherapy and hematopoietic stem cell transplantation (HSCT), which have limited options and are prone to tumor recurrence and (or) drug resistance. Metformin is the first-line drug for the treatment of type 2 diabetes (T2DM). Recently, studies identified the potential anti-cancer ability of metformin in both T2DM patients and patients that are non-diabetic. The latest epidemiological and preclinical studies suggested a potential benefit of metformin in the prevention and treatment of patients with HM. The mechanism may involve the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway by metformin as well as other AMPK-independent pathways to exert anti-cancer properties. In addition, combining current conventional anti-cancer drugs with metformin may improve the efficacy and reduce adverse drug reactions. Therefore, metformin can also be used as an adjuvant therapeutic agent for HM. This paper highlights the anti-hyperglycemic effects and potential anti-cancer effects of metformin, and also compiles the in vitro and clinical trials of metformin as an anti-cancer and chemosensitizing agent for the treatment of HM. The need for future research on the use of metformin in the treatment of HM is indicated.
Collapse
Affiliation(s)
| | | | | | | | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
32
|
Acton EK, Willis AW, Hennessy S. Core concepts in pharmacoepidemiology: Key biases arising in pharmacoepidemiologic studies. Pharmacoepidemiol Drug Saf 2023; 32:9-18. [PMID: 36216785 DOI: 10.1002/pds.5547] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/13/2022] [Accepted: 09/22/2022] [Indexed: 02/06/2023]
Abstract
Pharmacoepidemiology has an increasingly important role in informing and improving clinical practice, drug regulation, and health policy. Therefore, unrecognized biases in pharmacoepidemiologic studies can have major implications when study findings are translated to the real world. We propose a simple taxonomy for researchers to use as a starting point when thinking through some of the most pervasive biases in pharmacoepidemiology. We organize this discussion according to biases best assessed with respect to the study population (including confounding by indication, channeling bias, healthy user bias, and protopathic bias), the study design (including prevalent user bias and immortal time bias), and the data source (including misclassification bias and missing data/loss to follow up). This tutorial defines, provides a curated list of recommended references, and illustrates through relevant case examples these key biases to consider when planning, conducting, or evaluating pharmacoepidemiologic studies.
Collapse
Affiliation(s)
- Emily K Acton
- Center for Real-world Effectiveness and Safety of Therapeutics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Translational Center of Excellence for Neuroepidemiology and Neurology Outcomes Research, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Allison W Willis
- Center for Real-world Effectiveness and Safety of Therapeutics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Translational Center of Excellence for Neuroepidemiology and Neurology Outcomes Research, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sean Hennessy
- Center for Real-world Effectiveness and Safety of Therapeutics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
Kim NE, Kang EH, Ha E, Lee JY, Lee JH. Association of type 2 diabetes mellitus with lung cancer in patients with chronic obstructive pulmonary disease. Front Med (Lausanne) 2023; 10:1118863. [PMID: 37181380 PMCID: PMC10172489 DOI: 10.3389/fmed.2023.1118863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Background Patients with chronic obstructive pulmonary disease (COPD) have an increased risk of developing lung cancer. Some studies have also suggested that diabetes mellitus (DM) may increase the risk of developing lung cancer. This study aimed to investigate whether type 2 DM (T2DM) is associated with an increased risk of lung cancer in patients with COPD. Materials and methods We conducted a retrospective analysis on two cohorts: the National Health Insurance Service-National Sample Cohort (NHIS-NSC) of Korea and the Common Data Model (CDM) database of a university hospital. Among patients newly diagnosed with COPD in each cohort, those with a lung cancer diagnosis were included, and a control group was selected through propensity score matching. We used the Kaplan-Meier analysis and Cox proportional hazard models to compare lung cancer incidence between patients with COPD and T2DM and those without T2DM. Results In the NHIS-NSC and CDM cohorts, we enrolled 3,474 and 858 patients with COPD, respectively. In both cohorts, T2DM was associated with an increased risk of lung cancer [NHIS-NSC: adjusted hazard ratio (aHR), 1.20; 95% confidence interval (CI), 1.02-1.41; and CDM: aHR, 1.45; 95% CI, 1.02-2.07). Furthermore, in the NHIS-NSC, among patients with COPD and T2DM, the risk of lung cancer was higher in current smokers than in never-smokers (aHR, 1.45; 95% CI, 1.09-1.91); in smokers with ≥30 pack-years than in never-smokers (aHR, 1.82; 95% CI, 1.49-2.25); and in rural residents than in metropolitan residents (aHR, 1.33; 95% CI, 1.06-1.68). Conclusion Our findings suggest that patients with COPD and T2DM may have an increased risk of developing lung cancer compared to those without T2DM.
Collapse
Affiliation(s)
- Nam Eun Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun-Hwa Kang
- Informatization Department, Ewha Womans University Medical Center, Seoul, Republic of Korea
| | - Eunhee Ha
- Graduate Program in System Health Science and Engineering, Department of Environmental Medicine, College of Medicine, Ewha Medical Research Institute, Ewha Womans University, Seoul, Republic of Korea
| | - Ji-Young Lee
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
- *Correspondence: Ji-Young Lee,
| | - Jin Hwa Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
- Jin Hwa Lee,
| |
Collapse
|
34
|
Rezaei M, Shams Z, Rasouli BS, Amirfard KD, Sadrabadi MS, Gheysarzadeh A, Haghani K, Bakhtiyari S. New Association Between Diabetes Mellitus and Pancreatic Cancer. Curr Diabetes Rev 2023; 19:e180122200320. [PMID: 35040413 DOI: 10.2174/1573399818666220118095952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/11/2021] [Accepted: 11/24/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Diabetes mellitus is a global issue that has affected the lives of many people all over the world. This disorder, which is also called the mother of all diseases, possesses high pathogenicity and results in the emergence of many disorders. One of the known correlated diseases is pancreatic cancer which can be accompanied by diabetes mellitus. Therefore, finding the association between these diseases and common genes is urgent. OBJECTIVE In this study, in order to survey the relationship between diabetes mellitus and pancreatic cancer, the common genes of these disorders were analyzed by bioinformatics tools. METHODS For this purpose, we screened 17 shared genes from microarray data downloaded from the Gene Expression Omnibus (GEO) database. In addition, the relationship between identified genes was constructed by STRING and DAVID tools. RESULTS In total, 112 genes were identified to be differentially expressed. Among these, 17 genes were found to be common, including two genes that were down-regulated and others that were upregulated. Other analyses showed that most of the genes were enriched in Vibrio cholera infection and the mTOR signaling pathway. The biological processes of such genes included oxygen and gas transport, phagosome acidification, and GTPase activity. CONCLUSION In this study, 17 common genes that had not previously been considered in diabetes and pancreatic cancer were screened, which can be further considered for clinical approaches and in vitro studies.
Collapse
Affiliation(s)
- Monireh Rezaei
- Department of Medical Genetics, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Zinat Shams
- Department of Biological Science, Kharazmi University, Tehran, Iran
| | - Bahareh Sadat Rasouli
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | | | | | - Ali Gheysarzadeh
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Karimeh Haghani
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Salar Bakhtiyari
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
35
|
Goto M, Perencevich EN. Metformin and Infections: What Is the Next Step in This Decades-Long Story? Clin Infect Dis 2022; 76:1245-1246. [PMID: 36560871 DOI: 10.1093/cid/ciac903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Michihiko Goto
- Division of Infectious Diseases, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
- Center for Access & Delivery Research and Evaluation, Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
| | - Eli N Perencevich
- Center for Access & Delivery Research and Evaluation, Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
- Division of General Internal Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
36
|
Oura K, Morishita A, Tani J, Masaki T. Antitumor Effects and Mechanisms of Metabolic Syndrome Medications on Hepatocellular Carcinoma. J Hepatocell Carcinoma 2022; 9:1279-1298. [PMID: 36545268 PMCID: PMC9760577 DOI: 10.2147/jhc.s392051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/04/2022] [Indexed: 12/15/2022] Open
Abstract
Liver cancer has a high incidence and mortality rate worldwide, with hepatocellular carcinoma (HCC) being the most common histological type. With the decrease in the number of newly infected patients and the spread of antiviral therapy, hepatitis virus-negative chronic liver diseases including steatohepatitis are increasingly accounting for a large proportion of HCC, and an important clinical characteristic is the high prevalence of metabolic syndrome including hypertension, type 2 diabetes (T2D), dyslipidemia, and obesity. Since patients with steatohepatitis are less likely to undergo surveillance for early detection of HCC, they may be diagnosed at an advanced stage and have worse prognosis. Therefore, treatment strategies for patients with HCC caused by steatohepatitis, especially in advanced stages, become increasingly important. Further, hypertension, T2D, and dyslipidemia may occur as side effects during systemic treatment, and there will be increasing opportunities to prescribe metabolic syndrome medications, not only for originally comorbid diseases, but also for adverse events during HCC treatment. Interestingly, epidemiological studies have shown that patients taking some metabolic syndrome medications are less likely to develop various types of cancers, including HCC. Basic studies have also shown that these drugs have direct antitumor effects on HCC. In particular, angiotensin II receptor blockers (a drug group for treating hypertension), biguanides (a drug group for treating T2D), and statins (a drug group for treating dyslipidemia) have shown to elucidate antitumor effects against HCC. In this review, we focus on the antitumor effects of metabolic syndrome medications on HCC and their mechanisms based on recent literature. New therapeutic agents are also increasingly being reported. Analysis of the antitumor effects of metabolic syndrome medications on HCC and their mechanisms will be doubly beneficial for HCC patients with metabolic syndrome, and the use of these medications may be a potential strategy against HCC.
Collapse
Affiliation(s)
- Kyoko Oura
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan,Correspondence: Kyoko Oura, Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki, Kida, Kagawa, Japan, Tel +81-87-891-2156, Fax +81-87-891-2158, Email
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
37
|
Dhawan A, Pifer PM, Sandulache VC, Skinner HD. Metabolic targeting, immunotherapy and radiation in locally advanced non-small cell lung cancer: Where do we go from here? Front Oncol 2022; 12:1016217. [PMID: 36591457 PMCID: PMC9794617 DOI: 10.3389/fonc.2022.1016217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
In the US, there are ~250,000 new lung cancer diagnoses and ~130,000 deaths per year, and worldwide there are an estimated 1.6 million deaths per year from this deadly disease. Lung cancer is the most common cause of cancer death worldwide, and it accounts for roughly a quarter of all cancer deaths in the US. Non-small cell lung cancer (NSCLC) represents 80-85% of these cases. Due to an enormous tobacco cessation effort, NSCLC rates in the US are decreasing, and the implementation of lung cancer screening guidelines and other programs have resulted in a higher percentage of patients presenting with potentially curable locoregional disease, instead of distant disease. Exciting developments in molecular targeted therapy and immunotherapy have resulted in dramatic improvement in patients' survival, in combination with new surgical, pathological, radiographical, and radiation techniques. Concurrent platinum-based doublet chemoradiation therapy followed by immunotherapy has set the benchmark for survival in these patients. However, despite these advances, ~50% of patients diagnosed with locally advanced NSCLC (LA-NSCLC) survive long-term. In patients with local and/or locoregional disease, chemoradiation is a critical component of curative therapy. However, there remains a significant clinical gap in improving the efficacy of this combined therapy, and the development of non-overlapping treatment approaches to improve treatment outcomes is needed. One potential promising avenue of research is targeting cancer metabolism. In this review, we will initially provide a brief general overview of tumor metabolism as it relates to therapeutic targeting. We will then focus on the intersection of metabolism on both oxidative stress and anti-tumor immunity. This will be followed by discussion of both tumor- and patient-specific opportunities for metabolic targeting in NSCLC. We will then conclude with a discussion of additional agents currently in development that may be advantageous to combine with chemo-immuno-radiation in NSCLC.
Collapse
Affiliation(s)
- Annika Dhawan
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States
| | - Phillip M. Pifer
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States
| | - Vlad C. Sandulache
- Bobby R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Heath D. Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States,*Correspondence: Heath D. Skinner,
| |
Collapse
|
38
|
Keys MT, Thinggaard M, Larsen LA, Pedersen DA, Hallas J, Christensen K. Reassessing the evidence of a survival advantage in Type 2 diabetes treated with metformin compared with controls without diabetes: a retrospective cohort study. Int J Epidemiol 2022; 51:1886-1898. [PMID: 36287641 DOI: 10.1093/ije/dyac200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 10/05/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Previous research has suggested that individuals with Type 2 diabetes and initiated on metformin monotherapy present with a survival advantage compared with the general population without diabetes. This finding has generated considerable interest in the prophylactic use of metformin against age-related morbidity. METHODS Utilizing Danish National Health Registers, we assessed differences in survival associated with metformin monotherapy for Type 2 diabetes compared with no diagnosis of diabetes in both singleton and discordant twin populations between 1996 and 2012. Data were analysed in both nested case-control and matched cohort study designs, with incidence rate ratios (IRRs) and hazard ratios estimated using conditional logistic regression and Cox proportional hazards regression, respectively. RESULTS In case-control pairs matched on birth year and sex or co-twin (sex, birth year and familial factors), incident Type 2 diabetes with treatment by metformin monotherapy initiation compared with no diagnosis of diabetes was associated with increased mortality in both singletons (IRR = 1.52, 95% CI: 1.37, 1.68) and discordant twin pairs (IRR = 1.90, 95% CI: 1.35, 2.67). After adjusting for co-morbidities and social indicators, these associations were attenuated to 1.32 (95% CI: 1.16, 1.50) and 1.64 (95% CI: 1.10, 2.46), respectively. Increased mortality was observed across all levels of cumulative use and invariant to a range of study designs and sensitivity analyses. CONCLUSIONS Treatment initiation by metformin monotherapy in Type 2 diabetes was not associated with survival equal or superior to that of the general population without diabetes. Our contrasting findings compared with previous research are unlikely to be the result of differences in epidemiological or methodological parameters.
Collapse
Affiliation(s)
- Matthew Thomas Keys
- Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense, Denmark.,The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Mikael Thinggaard
- Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense, Denmark.,The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Lisbeth Aagaard Larsen
- Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense, Denmark.,The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Dorthe Almind Pedersen
- Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense, Denmark.,The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jesper Hallas
- Clinical Pharmacology and Pharmacy, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Kaare Christensen
- Department of Epidemiology, Biostatistics, and Biodemography, University of Southern Denmark, Odense, Denmark.,The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Danish Ageing Research Centre, Department of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Abdulkareem NM, Bhat R, Powell RT, Chikermane S, Yande S, Trinh L, Abdelnasser HY, Tabassum M, Ruiz A, Sobieski M, Nguyen ND, Park JH, Johnson CA, Kaipparettu BA, Bond RA, Johnson M, Stephan C, Trivedi MV. Screening of GPCR drugs for repurposing in breast cancer. Front Pharmacol 2022; 13:1049640. [PMID: 36561339 PMCID: PMC9763283 DOI: 10.3389/fphar.2022.1049640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Drug repurposing can overcome both substantial costs and the lengthy process of new drug discovery and development in cancer treatment. Some Food and Drug Administration (FDA)-approved drugs have been found to have the potential to be repurposed as anti-cancer drugs. However, the progress is slow due to only a handful of strategies employed to identify drugs with repurposing potential. In this study, we evaluated GPCR-targeting drugs by high throughput screening (HTS) for their repurposing potential in triple-negative breast cancer (TNBC) and drug-resistant human epidermal growth factor receptor-2-positive (HER2+) breast cancer (BC), due to the dire need to discover novel targets and drugs in these subtypes. We assessed the efficacy and potency of drugs/compounds targeting different GPCRs for the growth rate inhibition in the following models: two TNBC cell lines (MDA-MB-231 and MDA-MB-468) and two HER2+ BC cell lines (BT474 and SKBR3), sensitive or resistant to lapatinib + trastuzumab, an effective combination of HER2-targeting therapies. We identified six drugs/compounds as potential hits, of which 4 were FDA-approved drugs. We focused on β-adrenergic receptor-targeting nebivolol as a candidate, primarily because of the potential role of these receptors in BC and its excellent long-term safety profile. The effects of nebivolol were validated in an independent assay in all the cell line models. The effects of nebivolol were independent of its activation of β3 receptors and nitric oxide production. Nebivolol reduced invasion and migration potentials which also suggests its inhibitory role in metastasis. Analysis of the Surveillance, Epidemiology and End Results (SEER)-Medicare dataset found numerically but not statistically significant reduced risk of all-cause mortality in the nebivolol group. In-depth future analyses, including detailed in vivo studies and real-world data analysis with more patients, are needed to further investigate the potential of nebivolol as a repurposed therapy for BC.
Collapse
Affiliation(s)
- Noor Mazin Abdulkareem
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, United states
| | - Raksha Bhat
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, United states
| | - Reid T. Powell
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, United states
| | - Soumya Chikermane
- Department of Pharmaceutical Health Outcomes and Policy, University of Houston, Houston, TX, United states
| | - Soham Yande
- Department of Pharmaceutical Health Outcomes and Policy, University of Houston, Houston, TX, United states
| | - Lisa Trinh
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, United states
| | - Hala Y. Abdelnasser
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, United states
| | - Mantasha Tabassum
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, United states
| | - Alexis Ruiz
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, United states
| | - Mary Sobieski
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, United states
| | - Nghi D. Nguyen
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, United states
| | - Jun Hyoung Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United states
| | - Camille A. Johnson
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, United states
| | - Benny A. Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United states
| | - Richard A. Bond
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, United states
| | - Michael Johnson
- Department of Pharmaceutical Health Outcomes and Policy, University of Houston, Houston, TX, United states
| | - Clifford Stephan
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, United states
| | - Meghana V. Trivedi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, United states,Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, United states,*Correspondence: Meghana V. Trivedi,
| |
Collapse
|
40
|
Shen Q, Zhang C, Jiang X, Li J, Liu F, Zhang X, En G, Pang B. Emerging current trends and research focus related to pancreatic cancer metabolism: A bibliometric and visualized analysis. Front Oncol 2022; 12:1009700. [PMID: 36505775 PMCID: PMC9732726 DOI: 10.3389/fonc.2022.1009700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022] Open
Abstract
Background As a malignant digestive system tumor, pancreatic cancer has unique metabolic characteristics. In recent years, the study of pancreatic cancer metabolism is in full swing, which provides a new direction for the treatment of pancreatic cancer patients. However, there is no systematic report of pancreatic cancer metabolism. In this paper, bibliometrics and visualization methods were used to analyze the number of publications, countries/regions, authors, institutions, journals, co-cited references, and keywords of pancreatic cancer metabolism articles, to summarize the research trends and predict research hotspots. Methods We searched, screened and downloaded articles on pancreatic cancer metabolism through the Web of Science Core Collection (WoSCC). Using CiteSpace, VOSviewer and Bibliometrix Package to analyze publications, countries/regions, authors, institutions, journals, co-cited references, and keywords of pancreatic cancer metabolism to identify research trends and predict research hotspots. Results According to the inclusion and exclusion criteria, a total of 5,255 articles were retrieved during the period 1943-2022. The number of publications on pancreatic cancer metabolism is increasing year by year. The United States (n=1602, 30.49%), China (n=1074, 20.44%), and Italy (n=313, 5.96%) are the three countries with the largest number of publications and citations, and there is close cooperation between countries. LI J (n=55) is the most prolific author. FUDAN UNIV (n=348) is the most published institution. CANCERS (n=118), PLOS ONE (n=93), and CANCER RESEARCH (n=80) are the most popular journals in this field. "Nutriment-deficient environment", "cancer chemoprevention" and "targeting cancer stem cell" are the main areas of focus. "immunotherapy", "ferroptosis" and "targeted therapy" are hot keywords in recent years. Taking pancreatic cancer metabolism as an entry point to study the role of traditional Chinese medicine (TCM) mainly focuses on curcumin and resveratrol, lack of broader and deeper research on TCM. Conclusions The number of publications on pancreatic cancer metabolism has generally increased, and scholars have generally paid more attention to this field. "immunotherapy", "ferroptosis" and "targeted therapy" are the current research hotspots. The in-depth study of pancreatic cancer metabolism will provide new ideas for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Qian Shen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanlong Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junchen Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fudong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiyuan Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ge’er En
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Pang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
41
|
The Effects of 6 Common Antidiabetic Drugs on Anti-PD1 Immune Checkpoint Inhibitor in Tumor Treatment. J Immunol Res 2022; 2022:2651790. [PMID: 36033393 PMCID: PMC9410852 DOI: 10.1155/2022/2651790] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/16/2022] [Accepted: 07/26/2022] [Indexed: 12/02/2022] Open
Abstract
Diabetes and cancer are common diseases and are frequently diagnosed in the same individual. These patients need to take antidiabetic drugs while receiving antitumor drugs therapy. Recently, immunotherapy offers significant advances for cancer treatment. However, it is unclear whether antidiabetic drugs affect immunotherapy. Here, by employing syngeneic mouse colon cancer model and melanoma model, we studied the effects of 6 common antidiabetic drugs on anti-PD1 immune checkpoint inhibitor in tumor treatment, including acarbose, sitagliptin, metformin, glimepiride, pioglitazone, and insulin. We found that acarbose and sitagliptin enhanced the tumor inhibition of anti-PD1, and metformin had no effect on the tumor inhibition of anti-PD1, whereas glimepiride, pioglitazone, and insulin weakened the tumor inhibition of anti-PD1. Our study suggests that cancer patients receiving anti-PD1 antibody therapy need serious consideration when choosing antidiabetic drugs. In particular, acarbose significantly inhibited tumor growth and further enhanced the therapeutic effect of anti-PD1, which can be widely used in tumor therapy. Based on this study, further clinical trials are expected.
Collapse
|
42
|
Hong J, Maacha S, Pidkovka N, Bates A, Salaria SN, Washington MK, Belkhiri A. AXL Promotes Metformin-Induced Apoptosis Through Mediation of Autophagy by Activating ROS-AMPK-ULK1 Signaling in Human Esophageal Adenocarcinoma. Front Oncol 2022; 12:903874. [PMID: 35936716 PMCID: PMC9354051 DOI: 10.3389/fonc.2022.903874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
AXL receptor tyrosine kinase promotes an invasive phenotype and chemotherapy resistance in esophageal adenocarcinoma (EAC). AXL has been implicated in the regulation of autophagy, but the underlying molecular mechanism remains poorly understood. Herein, we investigate the mechanistic role of AXL in autophagy as well as metformin-induced effects on the growth and survival of EAC. We demonstrate that AXL mediates autophagic flux through activation of AMPK-ULK1 signaling in a reactive oxygen species (ROS)-dependent mechanism by glucose starvation. AXL positively regulates basal cellular ROS levels without significantly affecting mitochondrial ROS production in EAC cells. Pharmacological inhibition of cellular ROS using Trolox abrogates glucose starvation-induced AMPK signaling and autophagy. We demonstrate that AXL expression is required for metformin-induced apoptosis in EAC cells in vitro. The apoptosis induction by metformin is markedly attenuated by inhibition of autophagy through genetic silencing of Beclin1 or ATG7 autophagy mediators, thereby confirming the requirement of intact autophagy for enhancing metformin-induced apoptosis in EAC cells. Our data indicate that metformin-induced autophagy displays a pro-apoptotic function in EAC cells. We show that the metformin-induced suppression of tumor growth in vivo is highly dependent on AXL expression in a tumor xenograft mouse model of EAC. We demonstrate that AXL promotes metformin-induced apoptosis through activation of autophagy in EAC. AXL may be a valuable biomarker to identify tumors that are sensitive to metformin. Therefore, AXL expression could inform the selection of patients for future clinical trials to evaluate the therapeutic efficacy of metformin in EAC.
Collapse
Affiliation(s)
- Jun Hong
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Selma Maacha
- Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Nataliya Pidkovka
- Department of Health Science, South College, Nashville, TN, United States
| | - Andreia Bates
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Safia N. Salaria
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Mary K. Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
- *Correspondence: Abbes Belkhiri,
| |
Collapse
|
43
|
Chao R, Nishida M, Yamashita N, Tokumasu M, Zhao W, Kudo I, Udono H. Nutrient Condition in the Microenvironment Determines Essential Metabolisms of CD8+ T Cells for Enhanced IFNγ Production by Metformin. Front Immunol 2022; 13:864225. [PMID: 35844589 PMCID: PMC9277540 DOI: 10.3389/fimmu.2022.864225] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/01/2022] [Indexed: 11/27/2022] Open
Abstract
Metformin (Met), a first-line drug for type 2 diabetes, lowers blood glucose levels by suppressing gluconeogenesis in the liver, presumably through the liver kinase B1-dependent activation of AMP-activated protein kinase (AMPK) after inhibiting respiratory chain complex I. Met is also implicated as a drug to be repurposed for cancers; its mechanism is believed identical to that of gluconeogenesis inhibition. However, AMPK activation requires high Met concentrations at more than 1 mM, which are unachievable in vivo. The immune-mediated antitumor response might be the case in a low dose Met. Thus, we proposed activating or expanding tumor-infiltrating CD8+ T cells (CD8TILs) in a mouse model by orally administering Met in free drinking water. Here we showed that Met, at around 10 μM and a physiologically relevant concentration, enhanced production of IFNγ,TNFα and expression of CD25 of CD8+ T cells upon TCR stimulation. Under a glucose-rich condition, glycolysis was exclusively involved in enhancing IFNγ production. Under a low-glucose condition, fatty acid oxidation or autophagy-dependent glutaminolysis, or both, was also involved. Moreover, phosphoenolpyruvate carboxykinase 1 (PCK1), converting oxaloacetate to phosphoenolpyruvate, became essential. Importantly, the enhanced IFNγ production was blocked by a mitochondrial ROS scavenger and not by an inhibitor of AMPK. In addition, IFNγ production by CD8TILs relied on pyruvate translocation to the mitochondria and PCK1. Our results revealed a direct effect of Met on IFNγ production of CD8+ T cells that was dependent on differential metabolic pathways and determined by nutrient conditions in the microenvironment.
Collapse
|
44
|
Pollard JA, Furutani E, Liu S, Esrick E, Cohen LE, Bledsoe J, Liu CW, Lu K, de Haro MJR, Surrallés J, Malsch M, Kuniholm A, Galvin A, Armant M, Kim AS, Ballotti K, Moreau L, Zhou Y, Babushok D, Boulad F, Carroll C, Hartung H, Hont A, Nakano T, Olson T, Sze SG, Thompson AA, Wlodarski MW, Gu X, Libermann TA, D’Andrea A, Grompe M, Weller E, Shimamura A. Metformin for treatment of cytopenias in children and young adults with Fanconi anemia. Blood Adv 2022; 6:3803-3811. [PMID: 35500223 PMCID: PMC9631552 DOI: 10.1182/bloodadvances.2021006490] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 04/15/2022] [Indexed: 11/26/2022] Open
Abstract
Fanconi anemia (FA), a genetic DNA repair disorder characterized by marrow failure and cancer susceptibility. In FA mice, metformin improves blood counts and delays tumor development. We conducted a single institution study of metformin in nondiabetic patients with FA to determine feasibility and tolerability of metformin treatment and to assess for improvement in blood counts. Fourteen of 15 patients with at least 1 cytopenia (hemoglobin < 10 g/dL; platelet count < 100 000 cells/µL; or an absolute neutrophil count < 1000 cells/µL) were eligible to receive metformin for 6 months. Median patient age was 9.4 years (range 6.0-26.5 ). Thirteen of 14 subjects (93%) tolerated maximal dosing for age; 1 subject had dose reduction for grade 2 gastrointestinal symptoms. No subjects developed hypoglycemia or metabolic acidosis. No subjects had dose interruptions caused by toxicity, and no grade 3 or higher adverse events attributed to metformin were observed. Hematologic response based on modified Myelodysplastic Syndrome International Working Group criteria was observed in 4 of 13 evaluable patients (30.8%; 90% confidence interval, 11.3-57.3). Median time to response was 84.5 days (range 71-128 days). Responses were noted in neutrophils (n = 3), platelets (n = 1), and red blood cells (n = 1). No subjects met criteria for disease progression or relapse during treatment. Correlative studies explored potential mechanisms of metformin activity in FA. Plasma proteomics showed reduction in inflammatory pathways with metformin. Metformin is safe and tolerable in nondiabetic patients with FA and may provide therapeutic benefit. This trial was registered at as #NCT03398824.
Collapse
Affiliation(s)
- Jessica A. Pollard
- Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Elissa Furutani
- Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Shanshan Liu
- Biostatistics and Research Design Center, Institutional Centers for Clinical and Translational Research, Harvard Medical School, Boston, MA
| | - Erica Esrick
- Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Laurie E. Cohen
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Department of Endocrinology, and
| | - Jacob Bledsoe
- Department of Pathology, Boston Children’s Hospital, Boston, MA
| | - Chih-Wei Liu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Maria Jose Ramirez de Haro
- Joint Research Unit UAB-Sant Pau Biomedical Research Institute,Institut de Recerca Hospital de la Santa Creu i Sant Pau-IIB Sant Pau, Universitat Autònoma de Barcelona, Barcelona Spain
- Center for Biomedical Network Research on Rare Diseases, Madrid, Spain
| | - Jordi Surrallés
- Joint Research Unit UAB-Sant Pau Biomedical Research Institute,Institut de Recerca Hospital de la Santa Creu i Sant Pau-IIB Sant Pau, Universitat Autònoma de Barcelona, Barcelona Spain
- Center for Biomedical Network Research on Rare Diseases, Madrid, Spain
| | - Maggie Malsch
- Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
- Clinical Research Operations Center, Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, MA
| | - Ashley Kuniholm
- Clinical Research Operations Center, Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, MA
| | - Ashley Galvin
- Clinical Research Operations Center, Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, MA
| | - Myriam Armant
- Trans Laboratory, Boston Children’s Hospital, Boston, MA
| | - Annette S. Kim
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Kaitlyn Ballotti
- Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
| | - Lisa Moreau
- Comprehensive Center for Fanconi Anemia, Dana-Farber Cancer Institute, Boston, MA
| | - Yu Zhou
- Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
| | - Daria Babushok
- Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA
| | - Farid Boulad
- Pediatric Hematology-Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Clint Carroll
- Pediatric Hematology-Oncology, The Children's Hospital at TriStar Centennial, Nashville, TN
| | - Helge Hartung
- Pediatric Hematology-Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Amy Hont
- Pediatric Hematology-Oncology, Children’s National Medical Center, Washington, DC
| | - Taizo Nakano
- Pediatric Hematology-Oncology, Children’s Hospital Colorado, Denver, CO
| | - Tim Olson
- Pediatric Hematology-Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Sei-Gyung Sze
- Department of Pediatrics, Maine Medical Center, Tufts University School of Medicine, Portland, ME
| | - Alexis A. Thompson
- Pediatric Hematology-Oncology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL
| | - Marcin W. Wlodarski
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Xuesong Gu
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Towia A. Libermann
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Alan D’Andrea
- Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Markus Grompe
- Oregon Stem Cell Center, Department of Pediatrics, Papé Family Institute, Oregon Health and Science University, Portland, OR; and
| | - Edie Weller
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Biostatistics and Research Design Center, Institutional Centers for Clinical and Translational Research, Harvard Medical School, Boston, MA
| | - Akiko Shimamura
- Pediatric Hematology-Oncology, Boston Children’s Hospital, Boston, MA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| |
Collapse
|
45
|
van Hattum JW, de Ruiter BM, Oddens JR, de Reijke TM, Wilmink JW, Molenaar RJ. The Effect of Metformin on Bladder Cancer Incidence and Outcomes: A Systematic Review and Meta-Analysis. Bladder Cancer 2022; 8:211-228. [PMID: 38993366 PMCID: PMC11181685 DOI: 10.3233/blc-211653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/17/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND Effective oral treatment options for urothelial bladder cancer (BC) are lacking. Metformin, the most frequently used oral drug in type II diabetes mellitus, has putative anticancer properties and could, therefore, influence BC incidence and treatment outcomes. We systematically reviewed the current literature regarding the effect of metformin on BC incidence and oncological outcomes in non-muscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC). METHODS This review was conducted according to the PRISMA guidelines. Literature was gathered through a systematic search in PubMed/Medline, EMBASE and the Cochrane library. Risk of bias was determined using the Cochrane risk-of-bias tool for randomized trials and the Newcastle-Ottawa Scale for non-randomized trials. Hazard ratios (HRs) were extracted and pooled in a random-effects meta-analysis. RESULTS We reviewed 13 studies, including 3,315,320 patients, considering the risk of developing BC after metformin exposure and 9 studies, including 4,006 patients, on oncological outcomes of patients with BC. Metformin did not affect BC incidence (HR 0.97, 95% CI 0.87 -1.09) or oncological outcomes for NMIBC but did show a reduced risk of recurrence (HR 0.52, 95% CI 0.32 -0.84), cancer-specific mortality (HR 0.58, 95% CI 0.43 -0.78) and overall mortality (HR 0.66, 95% CI 0.47 -0.92) in MIBC. CONCLUSIONS The role of metformin in the prevention and treatment of BC in patients remains unclear. Although a beneficial effect of metformin on treatment outcomes of certain stages of BC may exist, a definitive conclusion cannot be drawn. Prospective clinical trials are needed to assess the efficacy of metformin for BC treatment.
Collapse
Affiliation(s)
- Jons W. van Hattum
- Department of Urology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Ben Max de Ruiter
- Department of Urology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Jorg R. Oddens
- Department of Urology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Theo M. de Reijke
- Department of Urology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Johanna W. Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Remco J. Molenaar
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
46
|
Zhu S, Bai Q, Li L, Xu T. Drug repositioning in drug discovery of T2DM and repositioning potential of antidiabetic agents. Comput Struct Biotechnol J 2022; 20:2839-2847. [PMID: 35765655 PMCID: PMC9189996 DOI: 10.1016/j.csbj.2022.05.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 12/19/2022] Open
Abstract
Repositioning or repurposing drugs account for a substantial part of entering approval pipeline drugs, which indicates that drug repositioning has huge market potential and value. Computational technologies such as machine learning methods have accelerated the process of drug repositioning in the last few decades years. The repositioning potential of type 2 diabetes mellitus (T2DM) drugs for various diseases such as cancer, neurodegenerative diseases, and cardiovascular diseases have been widely studied. Hence, the related summary about repurposing antidiabetic drugs is of great significance. In this review, we focus on the machine learning methods for the development of new T2DM drugs and give an overview of the repurposing potential of the existing antidiabetic agents.
Collapse
Affiliation(s)
- Sha Zhu
- Key Lab of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Qifeng Bai
- Key Lab of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
- Corresponding author.
| | | | | |
Collapse
|
47
|
Taylor SE, Chan DK, Yang D, Bruno T, Lieberman R, Siddiqui J, Soong TR, Coffman L, Buckanovich RJ. Shifting the Soil: Metformin Treatment Decreases the Protumorigenic Tumor Microenvironment in Epithelial Ovarian Cancer. Cancers (Basel) 2022; 14:2298. [PMID: 35565427 PMCID: PMC9104826 DOI: 10.3390/cancers14092298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 12/04/2022] Open
Abstract
Controversy persists regarding metformin's role in cancer therapy. Our recent work suggested metformin acts by impacting the tumor microenvironment (TME), normalizing the epigenetic profile of cancer-associated mesenchymal stem cells (CA-MSC). As CA-MSC can negatively impact tumor immune infiltrates, we evaluated metformin's impact on the human TME, focusing on the interplay of stroma and immune infiltrates. Tumor samples from (i) 38 patients treated with metformin and chemotherapy and (ii) 44 non-metformin matched controls were included in a tissue microarray (TMA). The TMA was used to compare the presence of CA-MSC, desmoplasia and immune infiltrates in the TME. In vitro and in vivo models examined metformin's role in alteration of the CA-MSC phenotype. The average percentage of CA-MSC was significantly lower in metformin-treated than in chemotherapy alone-treated tumors (p = 0.006). There were fewer regulatory T-cells in metformin-treated tumors (p = 0.043). Consistent with CA-MSC's role in excluding T-cells from tumor islets, the T-cells were primarily present within the tumor stroma. Evaluation of metformin's impact in vitro suggested that metformin cannot reverse a CA-MSC phenotype; however, the in vivo model where metformin was introduced prior to the establishment of the CA-MSC phenotype supported that metformin can partially prevent the reprogramming of normal MSC into CA-MSC. Metformin treatment led to a decrease in both the presence of protumorigenic CA-MSC and in immune exclusion of T cells, leading to a more immune-permissive environment. This suggests clinical utility in prevention and in treatment for early-stage disease and putatively in immune therapy.
Collapse
Affiliation(s)
- Sarah E. Taylor
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (D.K.C.); (L.C.); (R.J.B.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA;
| | - Daniel K. Chan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (D.K.C.); (L.C.); (R.J.B.)
| | - Dongli Yang
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA;
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tulia Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Richard Lieberman
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Javed Siddiqui
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Thing Rinda Soong
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Lan Coffman
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (D.K.C.); (L.C.); (R.J.B.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA;
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ronald J. Buckanovich
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (D.K.C.); (L.C.); (R.J.B.)
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA;
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
48
|
Metformin combined with local irradiation provokes abscopal effects in a murine rectal cancer model. Sci Rep 2022; 12:7290. [PMID: 35508498 PMCID: PMC9068771 DOI: 10.1038/s41598-022-11236-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/18/2022] [Indexed: 11/20/2022] Open
Abstract
Although preoperative chemoradiation therapy can down-stage locally advanced rectal cancer (LARC), it has little effect on distant metastases. Metformin exerts an anti-cancer effect partly through the activation of host immunity. LuM1, a highly lung metastatic subclone of colon 26, was injected subcutaneously (sc) in BALB/c mice and treated with metformin and/or local radiation (RT). Lung metastases and the primary tumors were evaluated and the phenotypes of immune cells in the spleen and lung metastases were examined with flow cytometry and immunohistochemistry. Local RT, but not metformin, partially delayed the growth of sc tumor which was augmented with metformin. Lung metastases were unchanged in metformin or RT alone, but significantly reduced in the combined therapy. The ratios of splenic T cells tended to be low in the RT group, which were increased by the addition of metformin. IFN-γ production of the splenic CD4(+) and CD8(+) T cells was enhanced and CD49b (+) CD335(+) activated NK cells was increased after combined treatment group. Density of NK cells infiltrating in lung metastases was increased after combination treatment. Metformin effectively enhances local and abscopal effects of RT though the activation of cell-mediated immunity and might be clinically useful for LARC.
Collapse
|
49
|
Metformin Enhancement of Therapeutic Effects of 5-Fluorouracil and Oxaliplatin in Colon Cancer Cells and Nude Mice. Biomedicines 2022; 10:biomedicines10050955. [PMID: 35625692 PMCID: PMC9138369 DOI: 10.3390/biomedicines10050955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Studies have demonstrated that metformin has antitumor effects in addition to therapeutic effects on hyperglycemia; however, few studies have explored the effects of metformin in chemotherapy. Therefore, we hypothesized that the administration of metformin would enhance the therapeutic effects of 5-fluorouracil and oxaliplatin (FuOx) to inhibit the growth of colorectal cancer (CRC) cells in vitro and in vivo. The results of our in vitro experiments demonstrated that metformin significantly increased the effects of FuOx with respect to cell proliferation (p < 0.05), colony formation (p < 0.05), and migration (p < 0.01) and induced cell cycle arrest in the G0/G1 phase in HT29 cells and the S phase in SW480 and SW620 cells (p < 0.05). Flow cytometry analysis revealed that metformin combined with FuOx induced late apoptosis (p < 0.05) by mediating mitochondria-related Mcl-1 and Bim protein expression. Furthermore, in vivo, metformin combined with FuOx more notably reduced tumor volume than FuOx or metformin alone did in BALB/c mice (p < 0.05). These findings demonstrate that metformin may act as an adjunctive agent to enhance the chemosensitivity of CRC cells to FuOx. However, further clinical trials are warranted to validate the clinical implications of the findings.
Collapse
|
50
|
Gan X, Cao C, He Y, Hu X, Peng X, Su Y. The metformin has no significant anticancer effect on patients with advanced or unresectable cancer: systematic review and meta-analysis. Curr Pharm Des 2022; 28:1351-1358. [PMID: 35352646 DOI: 10.2174/1381612828666220329113434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/14/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND At present, the antitumor effect of metformin is controversial. Previous meta-analyses included observational studies, of which the results can be influenced by many confounders, affecting the result of meta-analyses and weakening the strength of evidence. Therefore, we conducted a meta-analysis to confirm the effect of metformin use on patients with advanced or unresectable cancers, including randomized clinical trials (RCTs). METHODS We searched for RCTs in accordance with the inclusion and exclusion criteria. A meta-analysis was conducted to combine hazard ratios (HRs) or risk ratios (RRs) and their 95% confidence intervals (CIs), using a random-effects model. RESULTS Finally, 7 eligible RCTs were included in meta-analysis. Overall, the combined results revealed that treatment with metformin did not improve the overall survival (OS) of patients (HR, 1.12; 95%CI, 0.91-1.37, P>0.05), and there was no clear evidence that metformin use was related to improved progression-free survival (PFS) (HR,1.17; 95%CI, 0.97-1.40; P>0.05). The pooled RR for grade III or IV adverse events was 0.92 (95%CI, 0.52-1.60; P>0.05), indicating that the use of metformin was not significantly related to increased toxicity. CONCLUSION Metformin does not significantly improve the survival of patients with advanced or unresectable cancer, regardless of cancer type and region. Open Science Framework: DOI 10.17605/OSF.IO/SPKE8.
Collapse
Affiliation(s)
- Xinyan Gan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chang Cao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan He
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaolin Hu
- Department of Nursing, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yonglin Su
- Rehabilitation Medicine Center and Department of Biotherapy, West China Hospital, Sichuan University, Chengdu, China;
| |
Collapse
|