1
|
Sawalha K, Gautam N, Sivakumar K, Paydak H, Mehta JL. Metformin: Its salutary effects beyond diabetes mellitus. J Investig Med 2025:10815589251327511. [PMID: 40033492 DOI: 10.1177/10815589251327511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Metformin, an oral hypoglycemic agent, is commonly used in patients with type II diabetes mellitus. Studies have shown its use is associated with a reduction in major cardiovascular events (MACE) in patients with type 2 diabetes such as hospitalization for acute myocardial infarction, stroke, transient ischemic attack, or cardiovascular death. There is also a suggestion that metformin may have effects beyond those relating to lowering of blood sugar. The goal of this review is to assess the effects of metformin in coronary artery disease (CAD), but more importantly, its effects on disease states other than CAD.
Collapse
Affiliation(s)
- Khalid Sawalha
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nitesh Gautam
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kalaivani Sivakumar
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Hakan Paydak
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jawaher L Mehta
- Division of Cardiovascular Disease, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
2
|
Metformin and Breast Cancer: Where Are We Now? Int J Mol Sci 2022; 23:ijms23052705. [PMID: 35269852 PMCID: PMC8910543 DOI: 10.3390/ijms23052705] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the most prevalent cancer and the leading cause of cancer-related death among women worldwide. Type 2 diabetes–associated metabolic traits such as hyperglycemia, hyperinsulinemia, inflammation, oxidative stress, and obesity are well-known risk factors for breast cancer. The insulin sensitizer metformin, one of the most prescribed oral antidiabetic drugs, has been suggested to function as an antitumoral agent, based on epidemiological and retrospective clinical data as well as preclinical studies showing an antiproliferative effect in cultured breast cancer cells and animal models. These benefits provided a strong rationale to study the effects of metformin in routine clinical care of breast cancer patients. However, the initial enthusiasm was tempered after disappointing results in randomized controlled trials, particularly in the metastatic setting. Here, we revisit the current state of the art of metformin mechanisms of action, critically review past and current metformin-based clinical trials, and briefly discuss future perspectives on how to incorporate metformin into the oncologist’s armamentarium for the prevention and treatment of breast cancer.
Collapse
|
3
|
El-Khayat SM, Abouegylah M, Abdallah D, Geweil AG, Elenbaby AM, Zahra OS. The effect of metformin when combined with neoadjuvant chemotherapy in breast cancer patients. Med Oncol 2021; 39:1. [PMID: 34739637 DOI: 10.1007/s12032-021-01599-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/15/2021] [Indexed: 12/29/2022]
Abstract
Metformin has been used to treat type 2 Diabetes Mellitus since long time. It has two proposed anti-neoplastic mechanisms, direct (insulin-independent) and indirect (insulin-dependent) actions. To assess the effect of Metformin on pathological response when combined with neoadjuvant chemotherapy in breast cancer. A prospective study included stage II, III non-diabetic breast cancer patients who received neoadjuvant chemotherapy in our center during the period from May 2017 to March 2019. 59 patients met our inclusion criteria and completed the study, 27 patients received 850 mg Metformin every 12 h with chemotherapy (group A), and 32 patients received chemotherapy without Metformin (group B). Pathological response was assessed by Chevallier classification and residual cancer burden score (RCB). Both groups were well balanced regarding baseline characteristics. The results of our study showed that the rate of pathological complete response (pCR) was 14.8% in group (A) vs. 6.3% in group (B) with a P value of 0.39. RCB class 3 was 40.7% in group (A) vs. 68.8% in group (B) which was statistically significant with a (P value of 0.031). Patients with triple-positive histology who had RCB class 3 were only (14.3%) in group (A) versus (60%) in group B. Patients with body mass index (BMI) ≥ 25 who had RCB 3 were 40% and 66.7% in group (A) and (B), respectively. Metformin may increase the pCR especially in patients with BMI ≥ 25 and patients with triple-positive histology, a larger phase III study is needed to confirm this finding.
Collapse
Affiliation(s)
- Shaimaa M El-Khayat
- Clinical Oncology Department, Medical Research Institute, Alexandria University, 169 El-hureya Street, Qism Bab Sharqi, Alexandria, Alexandria Governorate, Egypt.
| | - Mohamed Abouegylah
- Clinical Oncology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Dina Abdallah
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ahmed Gaber Geweil
- Clinical Oncology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - A M Elenbaby
- Clinical Oncology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Omar Shebl Zahra
- Clinical Oncology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
4
|
Torres-Vanegas JD, Cruz JC, Reyes LH. Delivery Systems for Nucleic Acids and Proteins: Barriers, Cell Capture Pathways and Nanocarriers. Pharmaceutics 2021; 13:428. [PMID: 33809969 PMCID: PMC8004853 DOI: 10.3390/pharmaceutics13030428] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has been used as a potential approach to address the diagnosis and treatment of genetic diseases and inherited disorders. In this line, non-viral systems have been exploited as promising alternatives for delivering therapeutic transgenes and proteins. In this review, we explored how biological barriers are effectively overcome by non-viral systems, usually nanoparticles, to reach an efficient delivery of cargoes. Furthermore, this review contributes to the understanding of several mechanisms of cellular internalization taken by nanoparticles. Because a critical factor for nanoparticles to do this relies on the ability to escape endosomes, researchers have dedicated much effort to address this issue using different nanocarriers. Here, we present an overview of the diversity of nanovehicles explored to reach an efficient and effective delivery of both nucleic acids and proteins. Finally, we introduced recent advances in the development of successful strategies to deliver cargoes.
Collapse
Affiliation(s)
- Julian D. Torres-Vanegas
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| |
Collapse
|
5
|
Samuel SM, Varghese E, Koklesová L, Líšková A, Kubatka P, Büsselberg D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers (Basel) 2020; 12:E2482. [PMID: 32883003 PMCID: PMC7565921 DOI: 10.3390/cancers12092482] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the leaps and bounds in achieving success in the management and treatment of breast cancers through surgery, chemotherapy, and radiotherapy, breast cancer remains the most frequently occurring cancer in women and the most common cause of cancer-related deaths among women. Systemic therapeutic approaches, such as chemotherapy, although beneficial in treating and curing breast cancer subjects with localized breast tumors, tend to fail in metastatic cases of the disease due to (a) an acquired resistance to the chemotherapeutic drug and (b) the development of intrinsic resistance to therapy. The existence of cancer stem cells (CSCs) plays a crucial role in both acquired and intrinsic chemoresistance. CSCs are less abundant than terminally differentiated cancer cells and confer chemoresistance through a unique altered metabolism and capability to evade the immune response system. Furthermore, CSCs possess active DNA repair systems, transporters that support multidrug resistance (MDR), advanced detoxification processes, and the ability to self-renew and differentiate into tumor progenitor cells, thereby supporting cancer invasion, metastasis, and recurrence/relapse. Hence, current research is focusing on targeting CSCs to overcome resistance and improve the efficacy of the treatment and management of breast cancer. Studies revealed that metformin (1, 1-dimethylbiguanide), a widely used anti-hyperglycemic agent, sensitizes tumor response to various chemotherapeutic drugs. Metformin selectively targets CSCs and improves the hypoxic microenvironment, suppresses the tumor metastasis and inflammation, as well as regulates the metabolic programming, induces apoptosis, and reverses epithelial-mesenchymal transition and MDR. Here, we discuss cancer (breast cancer) and chemoresistance, the molecular mechanisms of chemoresistance in breast cancers, and metformin as a chemo-sensitizing/re-sensitizing agent, with a particular focus on breast CSCs as a critical contributing factor to acquired and intrinsic chemoresistance. The review outlines the prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer/chemo-sensitizing drug in the treatment of breast cancer. It intends to provide a rationale for the use of metformin as a combinatory therapy in a clinical setting.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Lenka Koklesová
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Alena Líšková
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
6
|
Xue J, Li L, Li N, Li F, Qin X, Li T, Liu M. Metformin suppresses cancer cell growth in endometrial carcinoma by inhibiting PD-L1. Eur J Pharmacol 2019; 859:172541. [PMID: 31319067 DOI: 10.1016/j.ejphar.2019.172541] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/02/2019] [Accepted: 07/15/2019] [Indexed: 01/09/2023]
Abstract
Endometrial cancer is the most common cancer of the female reproductive system in the developed countries. Metformin is a widely used medication that has been prescribed to treat type 2 diabetes. In recent years, metformin has been found to improve the survival prognosis of cancer patients clinically. We aimed to investigate inhibition of metformin on the proliferation of endometrial carcinoma. Metformin was used to treat endometrial cancer cell lines Ishikawa and RL95-2. The expression of programmed death-ligand 1 (PD-L1) in the treated cells was assessed by western blot. The tumor cell proliferation was evaluated by colony formation assay. The binding between PD-L1 and AMP-activated protein kinase (AMPK) was identified by co-immunoprecipitation. Ishikawa and RL95-2 cells were co-cultured with activated T cells to detect the survival of Ishikawa and RL95-2 cells in the presence or absence of metformin. Our results showed that metformin treatment on endometrial cancer cells Ishikawa and RL95-2 decreased the expression level of PD-L1 protein. Metformin treatment significantly activated T cells against Ishikawa and RL95-2 cells. We demonstrated that the inhibition of PD-L1 by metformin is dependent on the AMPK signaling protein, and that metformin promotes direct binding of the AMPK protein to the PD-L1 protein. We confirmed that metformin, a conventional medication used in diabetes therapy, holds anti-tumor activity in endometrial cancer. The suppression of metformin is relevant to the inhibition of PD-L1 expression and the activation of AMPK signaling protein, providing a novel mechanism in the anti-tumor property of metformin.
Collapse
Affiliation(s)
- Jing Xue
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, No 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, No 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Na Li
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, No 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Feifei Li
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, No 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Xiaoyan Qin
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, No 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Tao Li
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, No 324 Jingwu Road, Jinan, 250021, Shandong, China
| | - Ming Liu
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, No 324 Jingwu Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
7
|
Deng J, Peng M, Wang Z, Zhou S, Xiao D, Deng J, Yang X, Peng J, Yang X. Novel application of metformin combined with targeted drugs on anticancer treatment. Cancer Sci 2018; 110:23-30. [PMID: 30358009 PMCID: PMC6317954 DOI: 10.1111/cas.13849] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022] Open
Abstract
The success of targeted drug therapies for treating cancer patients has attracted broad attention both in the academic community and social society. However, rapidly developed acquired resistance is becoming a newly recognized major challenge to the continuing efficiency of these therapies. Metformin is a well‐known natural compound with low toxicity derived from the plant French lilac. Our previous work has highlighted research progress of the combination of clinically applied chemotherapies and metformin by different mechanisms. We have also launched a study to combine metformin with the small molecule targeted drug gefitinib to treat bladder cancer using intravesical administration. Thus, in this minireview, we summarize recent achievements combining metformin with various targeted therapies. This work directs the potential clinical future by selecting available cancer patients and providing precise medicine by the combination of metformin and targeted drugs to overcome resistance and enhance therapeutic efficacies.
Collapse
Affiliation(s)
- Jun Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiren Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Sichun Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Jiating Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Xue Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingyuan Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
8
|
Martin-Castillo B, Pernas S, Dorca J, Álvarez I, Martínez S, Pérez-Garcia JM, Batista-López N, Rodríguez-Sánchez CA, Amillano K, Domínguez S, Luque M, Stradella A, Morilla I, Viñas G, Cortés J, Cuyàs E, Verdura S, Fernández-Ochoa Á, Fernández-Arroyo S, Segura-Carretero A, Joven J, Pérez E, Bosch N, Garcia M, López-Bonet E, Saidani S, Buxó M, Menendez JA. A phase 2 trial of neoadjuvant metformin in combination with trastuzumab and chemotherapy in women with early HER2-positive breast cancer: the METTEN study. Oncotarget 2018; 9:35687-35704. [PMID: 30479698 PMCID: PMC6235018 DOI: 10.18632/oncotarget.26286] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022] Open
Abstract
The METTEN study assessed the efficacy, tolerability, and safety of adding metformin to neoadjuvant chemotherapy plus trastuzumab in early HER2-positive breast cancer (BC). Women with primary, non-metastatic HER2-positive BC were randomized (1:1) to receive metformin (850 mg twice-daily) for 24 weeks concurrently with 12 cycles of weekly paclitaxel plus trastuzumab, followed by four cycles of 3-weekly FE75C plus trastuzumab (arm A), or equivalent regimen without metformin (arm B), followed by surgery. Primary endpoint was the rate of pathological complete response (pCR) in the per-protocol efficacy population. pCR rate was numerically higher in the metformin-containing arm A (19 of 29 patients [65.5%, 95% CI: 47.3-80.1]) than in arm B (17 of 29 patients [58.6%, 95% CI: 40.7-74.5]; OR 1.34 [95% CI: 0.46-3.89], P = 0.589). The rate of breast-conserving surgery was 79.3% and 58.6% in arm A and B (P = 0.089), respectively. Blood metformin concentrations (6.2 μmol/L, 95% CI: 3.6-8.8) were within the therapeutic range. Seventy-six percent of patients completed the metformin-containing regimen; 13% of patients in arm A dropped out because of metformin-related gastrointestinal symptoms. The most common adverse events (AEs) of grade ≥3 were neutropenia in both arms and diarrhea in arm A. None of the serious AEs was deemed to be metformin-related. Addition of anti-diabetic doses of metformin to a complex neoadjuvant regimen was well tolerated and safe. Because the study was underpowered relative to its primary endpoint, the efficacy data should be interpreted with caution.
Collapse
Affiliation(s)
| | - Sonia Pernas
- Department of Medical Oncology, Breast Unit, Catalan Institute of Oncology-Hospital Universitari de Bellvitge-Bellvitge Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Joan Dorca
- Medical Oncology, Catalan Institute of Oncology, Girona, Spain
| | - Isabel Álvarez
- Medical Oncology Service, Hospital Universitario Donostia, Donostia-San Sebastián, Spain
- Biodonostia Health Research Institute, Donostia-San Sebastián, Spain
| | - Susana Martínez
- Medical Oncology Department, Hospital de Mataró, Mataró, Barcelona, Spain
| | | | - Norberto Batista-López
- Medical Oncology Service, Hospital Universitario de Canarias, La Laguna, Tenerife, Spain
| | - César A. Rodríguez-Sánchez
- Medical Oncology Service, Hospital Universitario de Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Kepa Amillano
- Medical Oncology, Hospital Universitari Sant Joan, Reus, Spain
| | - Severina Domínguez
- Medical Oncology Service, Hospital Universitario Araba, Vitoria-Gasteiz, Spain
| | - Maria Luque
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Agostina Stradella
- Department of Medical Oncology, Breast Unit, Catalan Institute of Oncology-Hospital Universitari de Bellvitge-Bellvitge Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Idoia Morilla
- Department of Medical Oncology, Breast Unit, Catalan Institute of Oncology-Hospital Universitari de Bellvitge-Bellvitge Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gemma Viñas
- Medical Oncology, Catalan Institute of Oncology, Girona, Spain
| | - Javier Cortés
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Elisabet Cuyàs
- Metabolism and Cancer Group, Girona Biomedical Research Institute, Girona, Spain
| | - Sara Verdura
- Metabolism and Cancer Group, Girona Biomedical Research Institute, Girona, Spain
| | - Álvaro Fernández-Ochoa
- Department of Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Centre (CIDAF), Health Science Technological Park, Granada, Spain
| | - Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, University of Granada, Granada, Spain
- Research and Development of Functional Food Centre (CIDAF), Health Science Technological Park, Granada, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Elsa Pérez
- Department of Radiology-IDI, Dr. Josep Trueta Hospital of Girona, Girona, Spain
| | - Neus Bosch
- Unit of Clinical Research, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Margarita Garcia
- Clinical Research Unit, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Eugeni López-Bonet
- Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona, Spain
| | - Samiha Saidani
- Unit of Clinical Research, Catalan Institute of Oncology, Girona, Spain
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Maria Buxó
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Javier A. Menendez
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
- Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism and Cancer Group, Catalan Institute of Oncology, Girona, Spain
| |
Collapse
|
9
|
Lee JH, Son YH, Kwon YW, Park SY, Koo BS, Jung SH. Anticancer Effects of a Korean Herbal Medicine Formula (H9) via AMPK and HER2-PI3K/Akt Signaling in Breast Cancer Cells. Phytother Res 2017; 31:1765-1775. [DOI: 10.1002/ptr.5906] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 07/18/2017] [Accepted: 08/07/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Ju-Hee Lee
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Young Hoon Son
- Department of Biochemistry, College of Medicine; Seoul National University; Seoul 03080 Korea
| | - Young-Won Kwon
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Sung Yun Park
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Byung-Soo Koo
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Seung Hyun Jung
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| |
Collapse
|
10
|
Pusceddu S, Prinzi N, Lo Russo G, Femia D, Milione M, Perrone F, Tamborini E, Concas L, Pulice I, Vernieri C, Corti F, Buzzoni R, de Braud F. Rationale and protocol of MetNET-2 trial: Lanreotide Autogel plus metformin in advanced gastrointestinal or lung neuroendocrine tumors. Future Oncol 2017; 13:1677-1683. [PMID: 28580793 DOI: 10.2217/fon-2017-0132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Metformin (MET) has recently emerged as a potentially active agent in cancer prevention and treatment. MET is thought to exert its antitumor effects either via modification of systemic metabolism or through cell-autonomous effects (e.g., activation of AMPK and inhibition of the mTOR pathway). Preliminary findings of the PRIME-NET study suggest that the addition of MET to treatment with everolimus (EVE) and/or somatostatin analogs (SSAs) can provide clinical benefit in diabetic neuroendocrine tumor (NET) patients. In light of this and other retrospective evidence of MET's anticancer activity in NETs, prospective studies are needed. A pilot, single-arm, open-label, prospective study (MetNET-2 trial, NCT02823691) was designed to evaluate the safety of MET in combination with lanreotide in well-differentiated gastrointestinal (WD GI) and lung NETs.
Collapse
Affiliation(s)
- Sara Pusceddu
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Giuseppe Lo Russo
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Daniela Femia
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Massimo Milione
- Department of Pathology, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Federica Perrone
- Department of Pathology, Laboratory of Experimental Molecular Pathology, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Elena Tamborini
- Department of of Diagnostic Pathology & Laboratory Medicine ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Laura Concas
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Iolanda Pulice
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Claudio Vernieri
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Roberto Buzzoni
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Unit 1, ENET Center of Excellence, Fondazione IRCCS Istituto Nazionale Tumori, Via G. Venezian 1, 20133 Milan, Italy
| |
Collapse
|
11
|
Lyu H, Yang XH, Edgerton SM, Thor AD, Wu X, He Z, Liu B. The erbB3- and IGF-1 receptor-initiated signaling pathways exhibit distinct effects on lapatinib sensitivity against trastuzumab-resistant breast cancer cells. Oncotarget 2016; 7:2921-35. [PMID: 26621843 PMCID: PMC4823081 DOI: 10.18632/oncotarget.6404] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 11/16/2015] [Indexed: 01/04/2023] Open
Abstract
Both erbB3 and IGF-1 receptor (IGF-1R) have been shown to play an important role in trastuzumab resistance. However, it remains unclear whether erbB3- and IGF-1R-initiated signaling pathways possess distinct effects on the sensitivity of lapatinib, a dual tyrosine kinase inhibitor against both EGFR and erbB2, in trastuzumab-resistant breast cancer. Here, we show that the trastuzumab-resistant SKBR3-pool2 and BT474-HR20 breast cancer sublines, as compared the parental SKBR3 and BT474 cells, respectively, exhibit refractoriness to lapatinib. Knockdown of erbB3 inhibited Akt in SKBR3-pool2 and BT474-HR20 cells, significantly increased lapatinib efficacy, and dramatically re-sensitized the cells to lapatinib-induced apoptosis. In contrast, specific knockdown of IGF-1R did not alter the cells' responsiveness to lapatinib. While the levels of phosphorylated Src (P-Src) were reduced upon IGF-1R downregulation, the P-Akt levels remained unchanged. Furthermore, a specific inhibitor of Akt, but not Src, significantly enhanced lapatinib-mediated anti-proliferative/anti-survival effects on SKBR3-pool2 and BT474-HR20 cells. These data indicate that erbB3 signaling is critical for both trastuzumab and lapatinib resistances mainly through the PI-3K/Akt pathway, whereas IGF-1R-initiated Src activation results in trastuzumab resistance without affecting lapatinib sensitivity. Our findings may facilitate the development of precision therapeutic regimens for erbB2-positive breast cancer patients who become resistant to erbB2-targeted therapy.
Collapse
Affiliation(s)
- Hui Lyu
- Cancer Research Institute and Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Xiao He Yang
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Kannapolis, NC, USA
| | - Susan M Edgerton
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ann D Thor
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Xiaoying Wu
- Department of Pathology, Xiangya Hospital, School of Basic Medical Science, Central South University, Changsha, China
| | - Zhimin He
- Cancer Research Institute and Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bolin Liu
- Cancer Research Institute and Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
12
|
Alkhatib Y, Abdel Rahman Z, Kuriakose P. Clinical impact of metformin in diabetic diffuse large B-cell lymphoma patients: a case-control study. Leuk Lymphoma 2016; 58:1130-1134. [PMID: 27701994 DOI: 10.1080/10428194.2016.1239822] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Molecular studies have shown metformin to have a promising effect in lymphoma; however, there is lack of studies translating this effect into clinical settings. This was a case-control study to assess the clinical effect of metformin in diabetic diffuse large B-cell lymphoma (DLBCL) patients. Case subjects were diabetic on metformin with a new diagnosis of DLBCL. A total of 24 case subjects were identified, and for each case a control was matched. Outcomes of this study were to assess overall response rate, complete remission rate, progression free survival, and overall survival between the two groups. There was a significant increase in overall response rate, complete remission rate, and improved progression free survival in the metformin group compared to the control group, however, no significant overall survival difference was observed. Metformin use might be associated with an improved response rates and progression-free survival in diabetic DLBCL patients.
Collapse
Affiliation(s)
- Yaser Alkhatib
- a Division of Hematology and Medical Oncology , Henry Ford Hospital , Detroit , MI , USA
| | - Zaid Abdel Rahman
- b Department of Internal Medicine , Henry Ford Hospital , Detroit , MI , USA
| | - Philip Kuriakose
- a Division of Hematology and Medical Oncology , Henry Ford Hospital , Detroit , MI , USA
| |
Collapse
|
13
|
Zhao Y, Wang W, Guo S, Wang Y, Miao L, Xiong Y, Huang L. PolyMetformin combines carrier and anticancer activities for in vivo siRNA delivery. Nat Commun 2016; 7:11822. [PMID: 27264609 PMCID: PMC4897747 DOI: 10.1038/ncomms11822] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/04/2016] [Indexed: 02/08/2023] Open
Abstract
Metformin, a widely implemented anti-diabetic drug, exhibits potent anticancer efficacies. Herein a polymeric construction of Metformin, PolyMetformin (PolyMet) is successfully synthesized through conjugation of linear polyethylenimine (PEI) with dicyandiamide. The delocalization of cationic charges in the biguanide groups of PolyMet reduces the toxicity of PEI both in vitro and in vivo. Furthermore, the polycationic properties of PolyMet permits capture of siRNA into a core-membrane structured lipid-polycation-hyaluronic acid (LPH) nanoparticle for systemic gene delivery. Advances herein permit LPH-PolyMet nanoparticles to facilitate VEGF siRNA delivery for VEGF knockdown in a human lung cancer xenograft, leading to enhanced tumour suppressive efficacy. Even in the absence of RNAi, LPH-PolyMet nanoparticles act similarly to Metformin and induce antitumour efficacy through activation of the AMPK and inhibition of the mTOR. In essence, PolyMet successfully combines the intrinsic anticancer efficacy of Metformin with the capacity to carry siRNA to enhance the therapeutic activity of an anticancer gene therapy.
Collapse
Affiliation(s)
- Yi Zhao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Wei Wang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Shutao Guo
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Yuhua Wang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Lei Miao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Yang Xiong
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Leaf Huang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
14
|
Zambrano J, Yeh ES. Autophagy and Apoptotic Crosstalk: Mechanism of Therapeutic Resistance in HER2-Positive Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2016; 10:13-23. [PMID: 26997868 PMCID: PMC4790584 DOI: 10.4137/bcbcr.s32791] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 12/16/2022]
Abstract
While breast cancer patients benefit from the use of HER2 inhibitors, many fail therapy and become resistant to treatment, indicating a critical need to prevent treatment failure. A number of studies have emerged that highlight the catabolic process of autophagy in breast cancer as a mechanism of resistance to chemotherapy and targeted inhibitors. Furthermore, recent research has begun to dissect how autophagy signaling crosstalks with apoptotic signaling. Thus, a possible strategy in fighting resistance is to couple targeting of apoptotic and autophagy signaling pathways. In this review, we discuss how cellular response by autophagy circumvents cell death to promote resistance of breast cancers to HER2 inhibitors, as well as the potential avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Joelle Zambrano
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
15
|
Kalogirou C, Schäfer D, Krebs M, Kurz F, Schneider A, Riedmiller H, Kneitz B, Vergho D. Metformin-Derived Growth Inhibition in Renal Cell Carcinoma Depends on miR-21-Mediated PTEN Expression. Urol Int 2015; 96:106-15. [PMID: 26496641 DOI: 10.1159/000441011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/10/2015] [Indexed: 11/19/2022]
Abstract
PURPOSE Metformin (MF) acts as a tumour-suppressor in renal cell carcinoma (RCC) by inhibiting the AKT/mTOR pathway via AMPK activation. Here, we explore the influence of miR-21 and its target gene PTEN on MF effects in CAKI-1 and CAKI-2 cells. METHODS Proliferation assays (MTS) and qRT-PCR after transient transfection with pre- and anti-miR-21 and MF treatment were conducted. AMPK-dependency was assessed via transfection of siAMPK. The expression of PTEN, AKT and miR-21 after transient pre-miR-21 transfection and MF treatment was analysed. RESULTS We demonstrate that CAKI-1 cells, which were found to be less sensitive towards MF, showed a significant higher miR-21 and lower PTEN expression than CAKI-2. This was confirmed in a primary RCC collective (n = 28): miR-21 and PTEN expression correlated negatively. MF treatment lowered miR-21 AMPK-dependently and increased PTEN expression in the cell lines. Ectopic miR-21 regulation modulated MF sensitivity. Western blot analysis showed that pre-miR-21 transfection and MF treatment regulated PTEN expression with impact on pAKT levels in the cells. CONCLUSIONS We show that differing MF sensitivity in RCC cells is associated with and mediated through the regulation of miR-21/PTEN expression with an impact on subsequent AKT signalling. This provides imaginable clinical implications regarding MF therapy of RCC patients for the future.
Collapse
Affiliation(s)
- Charis Kalogirou
- Department of Urology and Paediatric Urology, Julius Maximilians University Medical Centre of Wx00FC;rzburg, Wx00FC;rzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Fujimori T, Kato K, Fujihara S, Iwama H, Yamashita T, Kobayashi K, Kamada H, Morishita A, Kobara H, Mori H, Okano K, Suzuki Y, Masaki T. Antitumor effect of metformin on cholangiocarcinoma: In vitro and in vivo studies. Oncol Rep 2015; 34:2987-96. [PMID: 26398221 DOI: 10.3892/or.2015.4284] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/31/2015] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma (CCA) is the most common biliary malignancy and the second most common hepatic malignancy after hepatocellular carcinoma (HCC). Treatment with the anti-diabetic drug metformin has been associated with reduced cancer incidence in patients with type 2 diabetes. Thus, the present study evaluated the effects of metformin on human CCA cell proliferation in vitro and in vivo and identified the microRNAs associated with its antitumor effects. Metformin inhibited the proliferation of the CCA cell lines HuCCT-1 and TFK-1 and blocked the G0 to G1 cell cycle transition, accompanied by AMP kinase pathway activation. Metformin treatment also led to marked decreases in cyclin D1 and cyclin-dependent kinase (Cdk) 4 protein levels and retinoblastoma protein phosphorylation. However, this drug did not affect p27kip protein expression. In addition, it reduced the phosphorylation of Axl, EphA10, ALK and PYK, as well as tumor proliferation in athymic nude mice with xenograft tumors. Furthermore, it markedly altered microRNA expression. These findings suggest that metformin may have clinical use in the treatment of CCA.
Collapse
Affiliation(s)
- Takayuki Fujimori
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Kiyohito Kato
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Shintaro Fujihara
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hisakazu Iwama
- Life Science Research Center, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Takuma Yamashita
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Kiyoyuki Kobayashi
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hideki Kamada
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hirohito Mori
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Keiichi Okano
- Department of Gastroenterological Surgery, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Yasuyuki Suzuki
- Department of Gastroenterological Surgery, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University, Faculty of Medicine, Graduate School of Medicine, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| |
Collapse
|
17
|
Fujihara S, Kato K, Morishita A, Iwama H, Nishioka T, Chiyo T, Nishiyama N, Miyoshi H, Kobayashi M, Kobara H, Mori H, Okano K, Suzuki Y, Masaki T. Antidiabetic drug metformin inhibits esophageal adenocarcinoma cell proliferation in vitro and in vivo. Int J Oncol 2015; 46:2172-80. [PMID: 25709052 DOI: 10.3892/ijo.2015.2903] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/02/2015] [Indexed: 01/30/2023] Open
Abstract
Esophageal carcinoma is the eighth most common cancer worldwide and the sixth leading cause of cancer-related deaths, with one of the worst prognoses of any form of cancer. Treatment with the anti-diabetic drug metformin has been associated with reduced cancer incidence in patients with type 2 diabetes. This study therefore evaluated the effects of metformin on the proliferation, in vitro and in vivo, of human esophageal adenocarcinoma cells, as well as the microRNAs associated with the antitumor effects of metformin. Metformin inhibited the proliferation of the esophageal adenocarcinoma cell lines OE19, OE33, SK-GT4 and OACM 5.1C, blocking the G0 to G1 transition in the cell cycle. This was accompanied by strong reductions in G1 cyclins, especially cyclin D1, cyclin-dependent kinase (Cdk)4, and Cdk6, and decreases in retinoblastoma protein phosphorylation. In addition, metformin reduced the phosphorylation of epidermal growth factor receptor and insulin-like growth factor and insulin-like growth factor-1 receptor, as well as angiogenesis-related proteins, such as vascular endothelial growth factor, tissue inhibitor of metalloproteinases (TIMP)-1, and TIMP-2. Metformin also markedly altered microRNA expression. Treatment with metformin of athymic nude mice bearing xenograft tumors reduced tumor proliferation. These findings suggest that metformin may have clinical use in the treatment of esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Shintaro Fujihara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Kiyohito Kato
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hisakazu Iwama
- Life Science Resarch Center, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Tomoko Nishioka
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Taiga Chiyo
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Noriko Nishiyama
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hisaaki Miyoshi
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Mitsuyoshi Kobayashi
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Hirohito Mori
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Keiichi Okano
- Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Yasuyuki Suzuki
- Gastroenterological Surgery, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
| |
Collapse
|
18
|
Chen CI, Kuan CF, Fang YA, Liu SH, Liu JC, Wu LL, Chang CJ, Yang HC, Hwang J, Miser JS, Wu SY. Cancer risk in HBV patients with statin and metformin use: a population-based cohort study. Medicine (Baltimore) 2015; 94:e462. [PMID: 25674734 PMCID: PMC4602747 DOI: 10.1097/md.0000000000000462] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic infection with hepatitis B virus (HBV) often causes chronic inflammation of the liver with an increased incidence of hepatocellular carcinoma (HCC). HBV-infected individuals may also have an increased incidence of nonliver cancers. Taking statin or metformin may decrease inflammation and infiltration, which may, as a result, reduce the risk of liver cancer or other major cancers in patients with HBV infection. The purpose of this study was to evaluate the hypothesis that statin and metformin could reduce the incidence of liver cancer (HCC) or nonliver cancers in patients with HBV.Using the Taiwan Longitudinal Health Insurance Database 2000 to 2008, this cohort study comprised patients with a recorded diagnosis of HBV (N = 71,847) between January 1, 2000 and December 31, 2008. Each patient was followed-up until the end of 2008. The occurrence of HCC or a nonliver cancer was evaluated in patients who either were or were not taking statin or metformin. Cox proportional hazard regressions were used to evaluate the cancer incidence after adjusting for known confounding factors.In total, 71,824 HBV-infected patients comprised the study cohort. Our study showed that either metformin or statin use was associated with a reduction in the incidence of cancer. This was most prominent in patients taking both statin and metformin. The adjusted hazard ratios (HRs) for patients using only statin were 0.52 (95% confidence interval [CI], 0.48-0.57) for all cancers, 0.28 (95% CI, 0.23-0.35) for liver cancer, and 0.63 (95% CI, 0.57-0.70) for nonliver cancers. Patients taking only metformin had risk-adjusted HRs of 0.82 (95% CI, 0.75-0.90) for all cancers, 0.97 (95% CI, 0.84-1.14) for liver cancer, and 0.75 (95% CI, 0.67-0.84) for nonliver cancers. A dose-dependent effect of statin use for chemoprevention was observed for all cancers, including both liver cancer and nonliver cancers. A dose-dependent effect of metformin was also seen in liver cancer and nonliver cancers without stratification into different cumulative daily doses of statin use.This population-based cohort study investigated the protective effect of statin and metformin against cancer events in patients with HBV infection. Our study demonstrated that either statin or metformin served as independent chemopreventive agents with a dose-response effect in reducing the incidence of cancer with a dose-response effect of the agents and an additive or synergistic effect of combining statin and metformin use in reducing the incidence of many cancers.
Collapse
Affiliation(s)
- Chang-I Chen
- From the Graduate Institute of Medical Science (C-IC); Center of Excellence for Cancer Research (C-IC, Y-AF); Cancer Center (C-IC), Wan Fang Hospital, Taipei Medical University, Taipei; Department of Health care Administration (C-FK), Central Taiwan University of Science and Technology, Taichung; Graduate Institute of Toxicology (S-HL), College of Medicine, National Taiwan University, Taipei; Division of Cardiovascular Medicine (J-CL), Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City; Department of Ophthalmology (L-LW), National Taiwan University Hospital; Section of Endocrinology and Metabolism (C-JC), Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei; Institute of Biomedical Informatics (H-CY), National Yang Ming University; Department of Biochemistry (JH), School of Medicine, Taipei Medical University, Taipei, Taiwan; City of Hope National Medical Center (JSM), Duarte, CA; College of Medical Science and Technology (JSM), Taipei Medical University; Graduate Institute of Toxicology (SYW), College of Medicine, National Taiwan University, Taipei; Department of Internal Medicine (SYW), School of Medicine, College of Medicine; Department of Radiation Oncology (SYW), Wan Fang Hospital, Taipei Medical University, Taipei; Department of Biotechnology (SYW), Hung Kuang University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Herrigel DJ, Moss RA. Diabetes mellitus as a novel risk factor for gastrointestinal malignancies. Postgrad Med 2015; 126:106-18. [PMID: 25414939 DOI: 10.3810/pgm.2014.10.2825] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Evidence of an emerging etiologic link between diabetes mellitus and several gastrointestinal malignancies is presented. Although a correlation between pancreatic cancer and diabetes mellitus has long been suspected, the potential role diabetes mellitus plays in the pathogenicity of both hepatocellular carcinoma and colon cancer is becoming increasingly well defined. Further supporting the prospect of etiologic linkage, the association of diabetes mellitus with colon cancer is consistently demonstrated to be independent of obesity. An increasing incidence of diabetes and obesity in the United States has led to a recent surge in incidence of hepatocellular cancer on the background of nonalcoholic fatty liver disease, and this disease is expected to commensurately grow in incidence. Widespread recognition of this emerging risk factor may lead to a change in screening practices. Although the mechanisms underlying the correlation are still under investigation, the role of insulin, the insulin-like growth factor-I, and related binding and signaling pathways as regulators of cell growth and cell proliferation are implicated in carcinogenesis and tumor growth. The potential role of metformin and other medications for diabetes mellitus in the chemoprevention, carcinogenesis, and treatment of gastrointestinal malignancies is also presented.
Collapse
Affiliation(s)
- Dana J Herrigel
- Department of Internal Medicine, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ
| | | |
Collapse
|
20
|
Yan C, Wei H, Minjuan Z, Yan X, Jingyue Y, Wenchao L, Sheng H. The mTOR inhibitor rapamycin synergizes with a fatty acid synthase inhibitor to induce cytotoxicity in ER/HER2-positive breast cancer cells. PLoS One 2014; 9:e97697. [PMID: 24866893 PMCID: PMC4035285 DOI: 10.1371/journal.pone.0097697] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/23/2014] [Indexed: 12/20/2022] Open
Abstract
Patients with ER/HER2-positive breast cancer have a poor prognosis and are less responsive to selective estrogen receptor modulators; this is presumably due to the crosstalk between ER and HER2. Fatty acid synthase (FASN) is essential for the survival and maintenance of the malignant phenotype of breast cancer cells. An intimate relationship exists between FASN, ER and HER2. We hypothesized that FASN may be the downstream effector underlying ER/HER2 crosstalk through the PI3K/AKT/mTOR pathway in ER/HER2-positive breast cancer. The present study implicated the PI3K/AKT/mTOR pathway in the regulation of FASN expression in ER/HER2-positive breast cancer cells and demonstrated that rapamycin, an mTOR inhibitor, inhibited FASN expression. Cerulenin, a FASN inhibitor, synergized with rapamycin to induce apoptosis and inhibit cell migration and tumorigenesis in ER/HER2-positive breast cancer cells. Our findings suggest that inhibiting the mTOR-FASN axis is a promising new strategy for treating ER/HER2-positive breast cancer.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cerulenin/pharmacology
- Drug Synergism
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Fatty Acid Synthase, Type I/antagonists & inhibitors
- Fatty Acid Synthase, Type I/genetics
- Fatty Acid Synthase, Type I/metabolism
- Female
- Fluorescent Antibody Technique, Indirect
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mice
- Mice, Nude
- Protein Kinase Inhibitors/pharmacology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sirolimus/pharmacology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chen Yan
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Huang Wei
- Department of Cardiology, Xi'jing hospital, Fourth military medical university, Xi'an, PR China
| | - Zheng Minjuan
- Department of Ultrasound, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Xue Yan
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yang Jingyue
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Liu Wenchao
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
- * E-mail: (LW); (HS)
| | - Han Sheng
- State Key Laboratory of Military Stomatology, Department of Information Center, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
- * E-mail: (LW); (HS)
| |
Collapse
|
21
|
Garofalo C, Capristo M, Manara MC, Mancarella C, Landuzzi L, Belfiore A, Lollini PL, Picci P, Scotlandi K. Metformin as an adjuvant drug against pediatric sarcomas: hypoxia limits therapeutic effects of the drug. PLoS One 2013; 8:e83832. [PMID: 24391834 PMCID: PMC3877110 DOI: 10.1371/journal.pone.0083832] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/08/2013] [Indexed: 01/27/2023] Open
Abstract
Metformin, a well-known insulin-sensitizer commonly used for type 2 diabetes therapy, has recently emerged as potentially very attractive drug also in oncology. It is cheap, it is relatively safe and many reports have indicated effects in cancer prevention and therapy. These desirable features are particularly interesting for pediatric sarcomas, a group of rare tumors that have been shown to be dependent on IGF and insulin system for pathogenesis and progression. Metformin exerts anti-mitogenic activity in several cancer histotypes through several molecular mechanisms. In this paper, we analyzed its effects against osteosarcoma, Ewing sarcoma and rhabdomyosarcoma, the three most common pediatric sarcomas. Despite in vitro metformin gave remarkable antiproliferative and chemosensitizing effects both in sensitive and chemoresistant cells, its efficacy was not confirmed against Ewing sarcoma xenografts neither as single agent nor in combination with vincristine. This discrepancy between in vitro and in vivo effects may be due to hypoxia, a common feature of solid tumors. We provide evidences that in hypoxia conditions metformin was not able to activate AMPK and inhibit mTOR signaling, which likely prevents the inhibitory effects of metformin on tumor growth. Thus, although metformin may be considered a useful complement of conventional chemotherapy in normoxia, its therapeutic value in highly hypoxic tumors may be more limited. The impact of hypoxia should be considered when novel therapies are planned for pediatric sarcomas.
Collapse
Affiliation(s)
- Cecilia Garofalo
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Mariantonietta Capristo
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Maria Cristina Manara
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Caterina Mancarella
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Lorena Landuzzi
- Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | | | - Pier-Luigi Lollini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Piero Picci
- Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Katia Scotlandi
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
- * E-mail:
| |
Collapse
|
22
|
Hajjar J, Habra MA, Naing A. Metformin: an old drug with new potential. Expert Opin Investig Drugs 2013; 22:1511-7. [PMID: 23978196 DOI: 10.1517/13543784.2013.833604] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Metformin is the most commonly prescribed antidiabetic oral agent. It has also been used off-label for polycystic ovarian syndrome, steatohepatitis, and HIV-associated metabolic abnormalities. However, this oldie is a newbie for the oncologist. Population studies have suggested that metformin decreased the incidence and mortality rates of cancer in diabetic patients. With better understanding of its mechanisms of antitumor activity, metformin may become a new drug for cancer in combination with chemotherapy or targeted therapy.
Collapse
Affiliation(s)
- Joud Hajjar
- Virginia Commonwealth University, Department of Internal Medicine, Division of Rheumatology, Allergy and Immunology , Richmond, VA , USA
| | | | | |
Collapse
|
23
|
El-Khattouti A, Selimovic D, Haikel Y, Hassan M. Crosstalk between apoptosis and autophagy: molecular mechanisms and therapeutic strategies in cancer. J Cell Death 2013; 6:37-55. [PMID: 25278778 PMCID: PMC4147769 DOI: 10.4137/jcd.s11034] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Both apoptosis and autophagy are highly conserved processes that besides their role in the maintenance of the organismal and cellular homeostasis serve as a main target of tumor therapeutics. Although their important roles in the modulation of tumor therapeutic strategies have been widely reported, the molecular actions of both apoptosis and autophagy are counteracted by cancer protective mechanisms. While apoptosis is a tightly regulated process that is implicated in the removal of damaged or unwanted cells, autophagy is a cellular catabolic pathway that is involved in lysosomal degradation and recycling of proteins and organelles, and thereby is considered an important survival/protective mechanism for cancer cells in response to metabolic stress or chemotherapy. Although the relationship between autophagy and cell death is very complicated and has not been characterized in detail, the molecular mechanisms that control this relationship are considered to be a relevant target for the development of a therapeutic strategy for tumor treatment. In this review, we focus on the molecular mechanisms of apoptosis, autophagy, and those of the crosstalk between apoptosis and autophagy in order to provide insight into the molecular mechanisms that may be essential for the balance between cell survival and death as well as their role as targets for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Denis Selimovic
- Institut National de la Santé et de la Recherche Médicale, U 977, 67000 Strasbourg, France. ; Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, U 977, 67000 Strasbourg, France. ; Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, U 977, 67000 Strasbourg, France. ; Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
24
|
Cufí S, Vazquez-Martin A, Oliveras-Ferraros C, Corominas-Faja B, Urruticoechea A, Martin-Castillo B, Menendez JA. Autophagy-related gene 12 (ATG12) is a novel determinant of primary resistance to HER2-targeted therapies: utility of transcriptome analysis of the autophagy interactome to guide breast cancer treatment. Oncotarget 2013; 3:1600-14. [PMID: 23307622 PMCID: PMC3681498 DOI: 10.18632/oncotarget.742] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The autophagic process, which can facilitate breast cancer resistance to endocrine, cytotoxic,
and molecularly targeted agents, is mainly regulated at the post-translational level. Although
recent studies have suggested a possible transcriptome regulation of the autophagic genes, little is
known about either the analysis tools that can be applied or the functional importance of putative
candidate genes emerging from autophagy-dedicated transcriptome studies. In this context, we
evaluated whether the constitutive activation of the autophagy machinery, as revealed by a
transcriptome analysis using an autophagy-focused polymerase chain reaction (PCR) array, might allow
for the identification of novel autophagy-specific biomarkers for intrinsic (primary) resistance to
HER2-targeted therapies. Quantitative real-time PCR (qRT-PCR)-based profiling of 84 genes involved
in autophagy revealed that, when compared to trastuzumab-sensitive SKBR3 cells, the positive
regulator of autophagic vesicle formation ATG12 (autophagy-related gene 12) was the
most differentially up-regulated gene in JIMT1 cells, a model of intrinsic cross-resistance to
trastuzumab and other HER1/2-targeting drugs. An analysis of the transcriptional status of
ATG12 in > 50 breast cancer cell lines suggested that the
ATG12 transcript is commonly upregulated in trastuzumab-unresponsive
HER2-overexpressing breast cancer cells. A lentiviral-delivered small hairpin RNA stable knockdown
of the ATG12 gene fully suppressed the refractoriness of JIMT1 cells to
trastuzumab, erlotinib, gefitinib, and lapatinib in vitro. ATG12 silencing
significantly reduced JIMT1 tumor growth induced by subcutaneous injection in nude mice. Remarkably,
the outgrowth of trastuzumab-unresponsive tumors was prevented completely when trastuzumab treatment
was administered in an ATG12-silenced genetic background. We demonstrate for the
first time the usefulness of low-density, autophagy-dedicated qRT-PCR-based platforms for monitoring
primary resistance to HER2-targeted therapies by transcriptionally screening the autophagy
interactome. The degree of predictive accuracy warrants further investigation in the clinical
situation.
Collapse
Affiliation(s)
- Sílvia Cufí
- Metabolism and Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology-Girona, ICO-Girona
| | | | | | | | | | | | | |
Collapse
|
25
|
Li JL, Han SL, Fan X. Modulating autophagy: a strategy for cancer therapy. CHINESE JOURNAL OF CANCER 2013; 30:655-68. [PMID: 21959043 PMCID: PMC4012266 DOI: 10.5732/cjc.011.10185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Autophagy is a process in which long-lived proteins, damaged cell organelles, and other cellular particles are sequestered and degraded. This process is important for maintaining the cellular microenvironment when the cell is under stress. Many studies have shown that autophagy plays a complex role in human diseases, especially in cancer, where it is known to have paradoxical effects. Namely, autophagy provides the energy for metabolism and tumor growth and leads to cell death that promotes tumor suppression. The link between autophagy and cancer is also evident in that some of the genes that regulate Carcinogenesis, oncogenes and tumor suppressor genes, participate in or impact the autophagy process. Therefore, modulating autophagy will be a valuable topic for cancer therapy. Many studies have shown that autophagy can inhibit the tumor growth when autophagy modulators are combined with radiotherapy and/or chemotherapy. These findings suggest that autophagy may be a potent target for cancer therapy.
Collapse
Affiliation(s)
- Jun-Lin Li
- Department of General Surgery, The Central Hospital of Yongzhou City, Yongzhou, Hunan, People's Republic of China.
| | | | | |
Collapse
|
26
|
Corominas-Faja B, Quirantes-Piné R, Oliveras-Ferraros C, Vazquez-Martin A, Cufí S, Martin-Castillo B, Micol V, Joven J, Segura-Carretero A, Menendez JA. Metabolomic fingerprint reveals that metformin impairs one-carbon metabolism in a manner similar to the antifolate class of chemotherapy drugs. Aging (Albany NY) 2012; 4:480-98. [PMID: 22837425 PMCID: PMC3433934 DOI: 10.18632/aging.100472] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Metabolomic fingerprint of breast cancer cells treated with the antidiabetic drug metformin revealed a significant accumulation of 5-formimino-tetrahydrofolate, one of the tetrahydrofolate forms carrying activated one-carbon units that are essential for the de novo synthesis of purines and pyrimidines. De novo synthesis of glutathione, a folate-dependent pathway interconnected with one-carbon metabolism was concomitantly depleted in response to metformin. End-product reversal studies demonstrated that thymidine alone leads to a significant but incomplete protection from metformin's cytostatic effects. The addition of the substrate hypoxanthine for the purine salvage pathway produces major rightward shifts in metformin's growth inhibition curves. Metformin treatment failed to activate the DNA repair protein ATM kinase and the metabolic tumor suppressor AMPK when thymidine and hypoxanthine were present in the extracellular milieu. Our current findings suggest for the first time that metformin can function as an antifolate chemotherapeutic agent that induces the ATM/AMPK tumor suppressor axis secondarily following the alteration of the carbon flow through the folate-related one-carbon metabolic pathways.
Collapse
|
27
|
Menendez JA, Oliveras-Ferraros C, Cufí S, Corominas-Faja B, Joven J, Martin-Castillo B, Vazquez-Martin A. Metformin is synthetically lethal with glucose withdrawal in cancer cells. Cell Cycle 2012; 11:2782-92. [PMID: 22809961 DOI: 10.4161/cc.20948] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Glucose deprivation is a distinctive feature of the tumor microecosystem caused by the imbalance between poor supply and an extraordinarily high consumption rate. The metabolic reprogramming from mitochondrial respiration to aerobic glycolysis in cancer cells (the "Warburg effect") is linked to oncogenic transformation in a manner that frequently implies the inactivation of metabolic checkpoints such as the energy rheostat AMP-activated protein kinase (AMPK). Because the concept of synthetic lethality in oncology can be applied not only to genetic and epigenetic intrinsic differences between normal and cancer cells but also to extrinsic ones such as altered microenvironment, we recently hypothesized that stress-energy mimickers such as the AMPK agonist metformin should produce metabolic synthetic lethality in a glucose-starved cell culture milieu imitating the adverse tumor growth conditions in vivo. Under standard high-glucose conditions, metformin supplementation mostly caused cell cycle arrest without signs of apoptotic cell death. Under glucose withdrawal stress, metformin supplementation circumvented the ability of oncogenes (e.g., HER2) to protect breast cancer cells from glucose-deprivation apoptosis. Significantly, representative cell models of breast cancer heterogeneity underwent massive apoptosis (by >90% in some cases) when glucose-starved cell cultures were supplemented with metformin. Our current findings may uncover crucial issues regarding the cell-autonomous metformin's anti-cancer actions: (1) The offently claimed clinically irrelevant, non-physiological concentrations needed to observe the metformin's anti-cancer effects in vitro merely underlie the artifactual interference of erroneous glucose-rich experimental conditions that poorly reflect glucose-starved in vivo conditions; (2) the preferential killing of cancer stem cells (CSC) by metformin may simply expose the best-case scenario for its synthetically lethal activity because an increased dependency on Warburg-like aerobic glycolysis (hyperglycolytic phenotype) is critical to sustain CSC stemness and immortality; (3) the microenvironment-mediated contextual synthetic lethality of metformin should be expected to enormously potentiate the anti-cancer effect of anti-angiogenesis agents that promote severe oxygen and glucose deprivation in certain areas of the tumor tissues.
Collapse
Affiliation(s)
- Javier A Menendez
- Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia, Spain.
| | | | | | | | | | | | | |
Collapse
|
28
|
Del Barco S, Vazquez-Martin A, Cufí S, Oliveras-Ferraros C, Bosch-Barrera J, Joven J, Martin-Castillo B, Menendez JA. Metformin: multi-faceted protection against cancer. Oncotarget 2012; 2:896-917. [PMID: 22203527 PMCID: PMC3282095 DOI: 10.18632/oncotarget.387] [Citation(s) in RCA: 232] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The biguanide metformin, a widely used drug for the treatment of type 2 diabetes, may exert cancer chemopreventive effects by suppressing the transformative and hyperproliferative processes that initiate carcinogenesis. Metformin's molecular targets in cancer cells (e.g., mTOR, HER2) are similar to those currently being used for directed cancer therapy. However, metformin is nontoxic and might be extremely useful for enhancing treatment efficacy of mechanism-based and biologically targeted drugs. Here, we first revisit the epidemiological, preclinical, and clinical evidence from the last 5 years showing that metformin is a promising candidate for oncology therapeutics. Second, the anticancer effects of metformin by both direct (insulin-independent) and indirect (insulin-dependent) mechanisms are discussed in terms of metformin-targeted processes and the ontogenesis of cancer stem cells (CSC), including Epithelial-to-Mesenchymal Transition (EMT) and microRNAs-regulated dedifferentiation of CSCs. Finally, we present preliminary evidence that metformin may regulate cellular senescence, an innate safeguard against cellular immortalization. There are two main lines of evidence that suggest that metformin's primary target is the immortalizing step during tumorigenesis. First, metformin activates intracellular DNA damage response checkpoints. Second, metformin attenuates the anti-senescence effects of the ATP-generating glycolytic metabotype-the Warburg effect-, which is required for self-renewal and proliferation of CSCs. If metformin therapy presents an intrinsic barrier against tumorigenesis by lowering the threshold for stress-induced senescence, metformin therapeutic strategies may be pivotal for therapeutic intervention for cancer. Current and future clinical trials will elucidate whether metformin has the potential to be used in preventive and treatment settings as an adjuvant to current cancer therapeutics.
Collapse
Affiliation(s)
- Sonia Del Barco
- Medical Oncology, Catalan Institute of Oncology, Girona, Catalonia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Oliveras-Ferraros C, Cufí S, Vazquez-Martin A, Menendez OJ, Bosch-Barrera J, Martin-Castillo B, Joven J, Menendez JA. Metformin rescues cell surface major histocompatibility complex class I (MHC-I) deficiency caused by oncogenic transformation. Cell Cycle 2012; 11:865-70. [PMID: 22333588 DOI: 10.4161/cc.11.5.19252] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Active avoidance by tumor cells from attack and elimination by immune cells is an emerging cancer hallmark that is achieved primarily through decreasing the levels of major histocompatibility complex class I (MHC-I) at the cancer cells' surface. Deficiencies in MHC-I antigen-restricted immunosurveillance may be intertwined with an altered, Warburg-like cancer cell-intrinsic metabolism, another emerging hallmark of cancer that involves a switch from mitochondrial respiration to glycolysis to efficiently support large-scale biosynthetic programs that are required for active cell proliferation. We recently envisioned that intervention strategies aimed at reversing the bioenergetic signature of cancer cells (e.g., the antidiabetic biguanide metformin) should correct oncogene (e.g., HER2)-driven MHC-I defects, thus preventing immune escape of oncogene transformants. First, we explored how metformin treatment impacted mitochondrial biogenesis in cultured breast cancer cells overexpressing the membrane tyrosine kinase receptor HER2, the best-characterized downregulator of MHC-I. Metformin exposure was found to dose-dependently increase the expression levels of cytochrome c oxidase I and mitochondrial succinate dehydrogenase, which are encoded by mitochondrial and nuclear DNA, respectively. Second, we explored whether metformin-enhanced mitochondrial biogenesis might significantly alter the MHC-I status in breast carcinoma cells. MHC-I expression, as assessed by flow cytometry using an anti-HLA-ABC monoclonal antibody, was fully restored (up to ~25-fold upregulation) in MHC-I-negative HER2 gene-amplified carcinoma cells. These findings may help delineate a previously unrecognized mechanism through which metformin (and metformin-like drugs) may enable a cancer patient's own immune system to mount an efficient anti-metastasis response that can prevent or delay disease recurrence. Restored antigenicity and immunogenicity of tumor cells may represent a previously unrecognized primary mode of action underlying the cancer-preventive effects of metformin.
Collapse
|
30
|
Aljada A, Mousa SA. Metformin and neoplasia: implications and indications. Pharmacol Ther 2011; 133:108-15. [PMID: 21924289 DOI: 10.1016/j.pharmthera.2011.09.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 08/20/2011] [Indexed: 01/08/2023]
Abstract
Metformin has been shown to exert anti-neoplastic and chemopreventive activities in epidemiological and animal studies. This review article discusses the epidemiological studies and examines the possible mechanisms by which metformin exerts its anti-carcinogenic activities in breast, colon, ovarian, lung, and prostate cancers. We performed a systematic review of the clinical studies examining the anti-neoplastic activities of metformin and the potential mechanisms associated with these activities. Several observational and biological studies revealed a significant association between metformin and reduction in cancer incidence. The mechanisms by which metformin exerts these effects are unknown. This action may be mediated through activation of AMP-activated protein kinase (AMPK), inhibition of the mammalian target of rapamycin (mTOR) pathway, and inhibition of insulin like growth factors (IGFs), and many others. Further laboratory investigation and large, prospective population clinical trials are required to elucidate metformin anti-neoplastic and chemo-preventive actions.
Collapse
Affiliation(s)
- Ahmad Aljada
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, Saudia Arabia.
| | | |
Collapse
|
31
|
|
32
|
Apontes P, Leontieva OV, Demidenko ZN, Li F, Blagosklonny MV. Exploring long-term protection of normal human fibroblasts and epithelial cells from chemotherapy in cell culture. Oncotarget 2011; 2:222-33. [PMID: 21447859 PMCID: PMC3260824 DOI: 10.18632/oncotarget.248] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Killing of proliferating normal cells limits chemotherapy of cancer. Several
strategies to selectively protect normal cells were previously suggested. Here
we further explored the protection of normal cells from cell cycle-specific
chemotherapeutic agents such as mitotic inhibitors (MI). We focused on a
long-term cell recovery (rather than on a short-term cell survival) after a
3-day exposure to MI (paclitaxel and nocodazole). In three normal human cell
types (RPE, NKE, WI-38t cells) but not in cancer cells with mutant p53,
pre-treatment with nutlin-3a, a non-genotoxic inducer of wt p53, caused G1
and/or G2 arrest, thus preventing lethal mitotic arrest caused by MI and
allowing normal cells to recover after removal of MI. Rapamycin, an inhibitor of
the nutrient-sensing mTOR pathway, potentiated the protective effect of
nutlin-3a in normal cells. Also, a combination of rapamycin and metformin, an
anti-diabetic drug, induced G1 and G2 arrest selectively in normal cells and
thereby protected them from MI. A combination of metformin and rapamycin also
protected normal cells in low glucose conditions, whereas in contrast it was
cytotoxic for cancer cells. Based on these data and the analysis of the
literature, we suggest that a rational combination of metformin and rapamycin
can potentiate chemotherapy with mitotic inhibitors against cancer, while
protecting normal cells, thus further increasing the therapeutic window.
Collapse
Affiliation(s)
- Pasha Apontes
- Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | | | | | | | |
Collapse
|
33
|
Nahta R, O'Regan RM. Evolving strategies for overcoming resistance to HER2-directed therapy: targeting the PI3K/Akt/mTOR pathway. Clin Breast Cancer 2011; 10 Suppl 3:S72-8. [PMID: 21115425 DOI: 10.3816/cbc.2010.s.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancers, which account for 25%-30% of breast cancers, are characterized by an aggressive course and a high propensity for recurrence in the 4 years following diagnosis. The use of trastuzumab-based chemotherapy in the adjuvant setting has markedly improved the outcome for patients with early stage HER2+ breast cancer. Likewise the use of trastuzumab in combination with chemotherapy in patients with metastatic HER2+ breast cancers has prolonged survival, with current expected median survival of about 3 years. Despite these major improvements in outcome, approximately 10% of patients develop a distant recurrence following adjuvant trastuzumab-based chemotherapy, and all patients with metastatic disease eventually develop disease progression. Known mechanisms of resistance to trastuzumab include increased signaling through upstream growth factors, phosphatase and tensin (PTEN) deficiency and alterations of the HER2 receptor. Many of these mechanisms are being targeted in the clinic in an attempt to improve outcome for patients with HER2+ breast cancers. The phosphatidylinositol 3-kinase (PI3K) pathway plays a key role in trastuzumab-resistance, through these and other mechanisms, and represents a logical target for drug development for trastuzumab-resistant breast cancers. The use of mammalian target of rapamycin (mTOR) inhibition has been demonstrated to potentially reverse resistance to trastuzumab in patients with HER2+, metastatic breast cancers. Phase I and II trials have produced encouraging results when the mTOR inhibitor, everolimus, was combined with trastuzumab with or without chemotherapy, in patients with trastuzumab-resistant HER2+ metastatic breast cancer. These results are being confirmed in ongoing phase III trials in the first-line and trastuzumab-resistant settings. The mechanism of how mTOR inhibitors reverse resistance to trastuzumab remains largely unexplained. Other agents targeting the PI3K pathway in trastuzumab-resistant breast cancers are in early phase clinical trials.
Collapse
Affiliation(s)
- Rita Nahta
- Department of Pharmacology, Emory University,Winship Cancer Institute, 1365 Clifton Road, Atlanta, GA 30322, USA
| | | |
Collapse
|
34
|
Bloomgarden ZT. World congress on insulin resistance, diabetes, and cardiovascular disease: Part 1. Diabetes Care 2011; 34:e115-120. [PMID: 21709286 PMCID: PMC3120184 DOI: 10.2337/dc11-0840] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
35
|
Decensi A, Puntoni M, Pruneri G, Guerrieri-Gonzaga A, Lazzeroni M, Serrano D, Macis D, Johansson H, Pala O, Luini A, Veronesi P, Galimberti V, Dotti MC, Viale G, Bonanni B. Lapatinib activity in premalignant lesions and HER-2-positive cancer of the breast in a randomized, placebo-controlled presurgical trial. Cancer Prev Res (Phila) 2011; 4:1181-9. [PMID: 21685235 DOI: 10.1158/1940-6207.capr-10-0337] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dual epidermal growth factor receptor (EGFR) and HER2 targeting with the tyrosine kinase inhibitor lapatinib is approved for treating advanced HER2-positive breast cancer and can prevent estrogen receptor (ER)-negative mammary tumors in HER2 transgenic mouse models. Ki-67 labeling index (LI) has prognostic and predictive value and can be used to screen drugs' therapeutic and preventive potential in a clinical model of short-term presurgical therapy of breast cancer. We conducted a randomized, placebo-controlled trial of lapatinib (1500 mg/d) administered orally for three weeks between biopsy and surgery in 60 women with HER-2-positive breast cancer to assess lapatinib biomarker (including the primary endpoint, Ki-67 LI) and clinical activity in invasive breast cancer, adjacent ductal intraepithelial neoplasia (DIN, which comprises ductal carcinoma in situ and atypical ductal hyperplasia), and distant ductal hyperplasia without atypia (DH). Ki-67 LI increased progressively in association with disease stage, increasing in the placebo arm, for example, by medians of 3% in DH to 20% in DIN to 30% in invasive cancer. Ki-67 LI in cancer tissue decreased by a mean (±SD) of 9.3% (±34.2) in the lapatinib arm and increased by 15.1% (±30.9) in the placebo arm (P = 0.008). Compared with placebo, lapatinib reduced Ki-67 significantly more in ER-negative tumors (by 34.8%; P = 0.01) but not significantly more in ER-positive tumors (by 12.3%; P = 0.2) and reduced Ki-67 more (nonsignificantly) in cytosol PTEN-overexpressing tumors (P = 0.057). The prevalence of DIN in post-treatment surgical specimens of both arms was similar (70%-76%), with a median Ki-67 of 15% (range, 5%-35%) on lapatinib versus 20% (5%-60%) on placebo (P = 0.067). The prevalence of DH also was similar in both arms (>90%), with a median Ki-67 of 1% (1%-7%) on lapatinib versus 3% (1%-5%) on placebo (P = 0.006). Other results of lapatinib versus placebo, respectively, were as follows: Median tumor diameter at surgery of 18 mm (11 mm-57 mm) versus 24 mm (10 mm-37 mm; P = 0.009); partial response of 13.6% versus 3.7%, stable disease of 59.1% versus 40.7%, and progression of 27.3% versus 55.6% (P-trend = 0.035). In conclusion, short-term lapatinib decreased cell proliferation in DIN, DH, and invasive HER-2-positive (especially ER-negative) breast cancer, thus providing the rationale for further clinical development of lapatinib for breast cancer prevention in high-risk patients, including those with HER-2-positive DIN.
Collapse
Affiliation(s)
- Andrea Decensi
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Guppy A, Jamal-Hanjani M, Pickering L. Anticancer effects of metformin and its potential use as a therapeutic agent for breast cancer. Future Oncol 2011; 7:727-36. [DOI: 10.2217/fon.11.49] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Metformin is an orally available, biguanide derivative that is widely used in the treatment of Type 2 diabetes. Recent preclinical data have demonstrated that it can also act as an anticancer agent by activation of AMPK and subsequent inhibition of mTOR. Metformin is currently being investigated in several Phase II/III clinical trials. This article will review the current evidence for its mechanism of action, efficacy in preclinical and clinical models, and toxicity. Ongoing and planned studies evaluating the impact of metformin on breast cancer outcomes are also discussed.
Collapse
Affiliation(s)
- Amy Guppy
- Mount Vernon Cancer Centre, Northwood, Middlesex HA6 2RN, UK
| | | | - Lisa Pickering
- St George’s Hospital Healthcare NHS Trust, Blackshaw Road, London SW17 9QT, UK
| |
Collapse
|
37
|
Huang X, Wang S, Lee CK, Yang X, Liu B. HDAC inhibitor SNDX-275 enhances efficacy of trastuzumab in erbB2-overexpressing breast cancer cells and exhibits potential to overcome trastuzumab resistance. Cancer Lett 2011; 307:72-79. [PMID: 21497990 DOI: 10.1016/j.canlet.2011.03.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 03/17/2011] [Accepted: 03/20/2011] [Indexed: 12/22/2022]
Abstract
Trastuzumab (or Herceptin), as the first erbB2-targeted therapy, has been successfully used to treat breast cancer patients with erbB2-overexpressing tumors. However, resistances to trastuzumab frequently occur, and novel strategies/agents are urgently needed to abrogate the resistant phenotype. Our current study explores the potential of SNDX-275, a class I HDAC inhibitor, to overcome trastuzumab resistance and investigates the combinational effects of SNDX-275 and trastuzumab on both sensitive and resistant breast cancer cells. Cell proliferation assays showed that SNDX-275 significantly enhanced trastuzumab-induced growth inhibition in trastuzumab-sensitive, erbB2-overexpressing breast cancer cells. Importantly, SNDX-275 at its therapeutic range re-sensitized trastuzumab-resistant cells to trastuzumab-mediated growth inhibition. SNDX-275 in combination with trastuzumab resulted in a dramatic reduction of erbB3 and its phosphorylation (P-erbB3), and inhibition of Akt signaling. Apoptotic-ELISA and western blot analyses confirmed that the combinations of SNDX-275 and trastuzumab as compared to SNDX-275 alone significantly enhanced DNA fragmentation and induced more PARP cleavage and caspase-3 activation in both trastuzumab-sensitive and -resistant breast cancer cells. Furthermore, co-immunoprecipitation assays revealed that SNDX-275 mainly attenuated the interactions of erbB2 and erbB3 receptors, but had no significant effect on erbB2/IGF-1R or erbB3/IGF-1R associations in the trastuzumab-resistant breast cancer cells. These data indicated that SNDX-275 enhanced trastuzumab efficacy against erbB2-overexpressing breast cancer cells, and exhibited potential to overcome trastuzumab resistance via disrupting erbB2/erbB3 interactions and inactivating PI-3K/Akt signaling. SNDX-275 may be included in erbB2-targeted regimen as a novel strategy to treat breast cancer patients whose tumors overexpress erbB2.
Collapse
Affiliation(s)
- Xiaoping Huang
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, CO, United States
| | - Shuiliang Wang
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, CO, United States
| | - Choon-Kee Lee
- The Myeloma and Amyloidosis Program, Department of Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States
| | - XiaoHe Yang
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Bolin Liu
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, CO, United States.
| |
Collapse
|
38
|
Oliveras-Ferraros C, Vazquez-Martin A, Cufí S, Torres-Garcia VZ, Sauri-Nadal T, Barco SD, Lopez-Bonet E, Brunet J, Martin-Castillo B, Menendez JA. Inhibitor of Apoptosis (IAP) survivin is indispensable for survival of HER2 gene-amplified breast cancer cells with primary resistance to HER1/2-targeted therapies. Biochem Biophys Res Commun 2011; 407:412-9. [PMID: 21402055 DOI: 10.1016/j.bbrc.2011.03.039] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 03/09/2011] [Indexed: 12/24/2022]
Abstract
Primary resistance of HER2 gene-amplified breast carcinomas (BC) to HER-targeted therapies can be explained in terms of overactive HER2-independent downstream pro-survival pathways. We here confirm that constitutive overexpression of Inhibitor of Apoptosis (IAP) survivin is indispensable for survival of HER2-positive BC cells with intrinsic cross-resistance to multiple HER1/2 inhibitors. The IC₅₀ values for the HER1/2 Tyrosine Kinase Inhibitors (TKIs) gefitinib, erlotinib and lapatinib were up to 40-fold higher in trastuzumab-unresponsive JIMT-1 cells than in trastuzumab-naïve SKBR3 cells. ELISA-based and immunoblotting assays demonstrated that trastuzumab-refractory JIMT-1 cells constitutively expressed ~ 4 times more survivin protein than trastuzumab-responsive SKBR3 cells. In response to trastuzumab, JIMT-1 cells accumulated ~10 times more survivin than SKBR3 cells. HER1/2 TKIs failed to down-regulate survivin expression in JIMT-1 cells whereas equimolar doses of HER1/HER2 TKIs drastically depleted survivin protein in SKBR3 cells. ELISA-based detection of histone-associated DNA fragments confirmed that trastuzumab-refractory JIMT-1 cells were intrinsically protected against the apoptotic effects of HER1/2 TKIs. Of note, when we knocked-down survivin expression using siRNA and then added trastuzumab, cell proliferation and colony formation were completely suppressed in JIMT-1 cells. Our current findings may be extremely helpful to design successful combinatorial strategies aimed to circumvent the occurrence of de novo resistance to HER2-directed drugs using survivin antagonists.
Collapse
Affiliation(s)
- Cristina Oliveras-Ferraros
- Unit of Translational Research, Catalan Institute of Oncology-Girona, Avenida de Francia S/N, E-17007 Girona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Minematsu T, Giacomini KM. Interactions of tyrosine kinase inhibitors with organic cation transporters and multidrug and toxic compound extrusion proteins. Mol Cancer Ther 2011; 10:531-9. [PMID: 21252289 PMCID: PMC3063525 DOI: 10.1158/1535-7163.mct-10-0731] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The drug-drug interaction (DDI) potential of tyrosine kinase inhibitors (TKI) as interacting drugs via transporter inhibition has not been fully assessed. Here, we estimated the half maximal inhibitory concentration (IC(50)) values for 8 small-molecule TKIs (imatinib, dasatinib, nilotinib, gefitinib, erlotinib, sunitinib, lapatinib, and sorafenib) on [(14)C]metformin transport by human organic cation transporters (OCT), OCT1, OCT2, and OCT3, and multidrug and toxic compound extrusion (MATE) proteins, MATE1 and MATE2-K, using human embryonic kidney cells stably expressing these transporters. We then compared the estimated IC(50) values to the maximum clinical concentrations of unbound TKIs in plasma (unbound C(max,sys,p)). Results showed that imatinib, nilotinib, gefitinib, and erlotinib exerted selectively potent inhibitory effects, with unbound C(max,sys,p)/IC(50) values ≥0.1, on MATE1, OCT3, MATE2-K, and OCT1, respectively. In comparison to the common form of OCT1, the OCT1 polymorphism, M420del, was more sensitive to drug inhibition by erlotinib. Major metabolites of several TKIs showed IC(50) values similar to those for unchanged TKIs. Taken together, these findings suggest the potential of clinical transporter-mediated DDIs between specific TKIs and OCTs and MATEs, which may affect the disposition, efficacy, and toxicity of metformin and other drugs that are substrates of these transporters. The study provides the basis for further clinical DDI studies with TKIs.
Collapse
Affiliation(s)
- Tsuyoshi Minematsu
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1550 4th Street, San Francisco, CA 94158, USA
| | | |
Collapse
|
40
|
Metformin and cancer: new applications for an old drug. Med Oncol 2011; 29:1314-27. [PMID: 21301998 DOI: 10.1007/s12032-011-9846-7] [Citation(s) in RCA: 221] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Accepted: 01/26/2011] [Indexed: 12/26/2022]
Abstract
Metformin, one of most widely prescribed oral hypoglycemic agents, has recently received increased attention because of its potential antitumorigenic effects that are thought to be independent of its hypoglycemic effects. Several potential mechanisms have been suggested for the ability of metformin to suppress cancer growth in vitro and vivo: (1) activation of LKB1/AMPK pathway, (2) induction of cell cycle arrest and/or apoptosis, (3) inhibition of protein synthesis, (4) reduction in circulating insulin levels, (5) inhibition of the unfolded protein response (UPR), (6) activation of the immune system, and (7) eradication of cancer stem cells. There is also a growing number of evidence, mostly in the form of retrospective clinical studies that suggest that metformin may be associated with a decreased risk of developing cancer and with a better response to chemotherapy. There are currently several ongoing randomized clinical trials that incorporate metformin as an adjuvant to classic chemotherapy and aim to evaluate its potential benefits in this setting. This review highlights basic aspects of the molecular biology of metformin and summarizes new advances in basic science as well as intriguing results from recent clinical studies.
Collapse
|
41
|
Clinical outcomes after radical prostatectomy in diabetic patients treated with metformin. Urology 2010; 76:1240-4. [PMID: 20627287 DOI: 10.1016/j.urology.2010.03.059] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 03/17/2010] [Accepted: 03/23/2010] [Indexed: 11/23/2022]
Abstract
OBJECTIVES To investigate the relationship between diabetes and metformin use with outcomes after radical prostatectomy (RP) for clinically localized cancer. METHODS A total of 112 diabetic metformin users and 98 diabetic non-metformin users treated with RP from 1990 to 2009 were identified. Nondiabetic controls were match using their 5-year risk of biochemical recurrence (BCR) as calculated by the preoperative Kattan nomogram. RESULTS A total of 616 patients were evaluated in this study. There was no significant difference between nondiabetic and diabetic patients, including metformin users, with respect to age, clinical stage, preoperative prostate-specific antigen (PSA) score, pathologic Gleason score, and pathologic stage. Diabetic patients, including metformin users, were more likely to be of African American or Hispanic background than were nondiabetic controls (P = .001). The estimated 5-year BCR-free survival was 75.0% for nondiabetic patients, compared with 66.1% for metformin users and 59.3% for diabetic non-metformin users (P = .004). In multivariate analysis, metformin use was not significantly associated with risk of BCR (HR = 0.94; 95% CI = 0.6-1.5, P = .817). However, being diabetic, regardless of metformin use, resulted in a 55% increase in risk of BCR (HR = 1.55; 95% CI = 1.03-2.33, P = .034). CONCLUSIONS Diabetes, regardless of metformin use, was significantly associated with an increased likelihood of BCR after RP. Metformin use did not prove to be of any benefit. These observations underscore the importance for further studies evaluating the metabolic pathways that affect prostate cancer biology.
Collapse
|
42
|
Perego P, Cossa G, Zuco V, Zunino F. Modulation of cell sensitivity to antitumor agents by targeting survival pathways. Biochem Pharmacol 2010; 80:1459-65. [PMID: 20688050 DOI: 10.1016/j.bcp.2010.07.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 07/20/2010] [Accepted: 07/23/2010] [Indexed: 01/17/2023]
Abstract
The advent of drugs targeting tumor-associated prosurvival alterations of cancer cells has changed the interest of antitumor drug development from cytotoxic drugs to target-specific agents. Although single-agent therapy with molecularly targeted agents has shown limited success in tumor growth control, a promising strategy is represented by the development of rational combinations of target-specific agents and conventional antitumor drugs. Activation of survival/antiapoptotic pathways is a common feature of cancer cells that converge in the development of cellular resistance to cytotoxic agents. The survival pathways implicated in cellular response to drug treatment are primarily PI3K/Akt and Ras/MAPK, which also mediate the signalling activated by growth factors and play a role in the regulation of critical processes including cell proliferation, metabolism, apoptosis and angiogenesis. Inhibitors of PI3K, Akt and mTOR have been shown to sensitize selected tumor cells to cytotoxic drugs through multiple downstream effects. Moreover, the MAPK pathway, also implicated in the regulation of gene expression in response to stress stimuli, can interfere with the chemotherapy-induced proapoptotic signals. Targeting Hsp90, which acts as a molecular chaperone for survival factors including Akt, may have the potential advantage to simultaneously block multiple oncogenic pathways. Overall, the available evidence supports the interest of rationally designed approaches to enhance the efficacy of conventional antitumor treatments through the inhibition of survival pathways and the notion that the concomitant targeting of multiple pathways may be a successful strategy to deal with tumor heterogeneity and to overcome drug resistance of tumor cells.
Collapse
Affiliation(s)
- Paola Perego
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy
| | | | | | | |
Collapse
|
43
|
Abstract
The article examines the importance of managing weight to reduce risk for developing cancer and for survival among cancer patients and presents a set of strategies that can be useful to guide clinical advice to patients for whom weight control is an important adjunct to risk management or to improve quality of life and disease-free survival after diagnosis. Weight, weight gain, and obesity account for approximately 20% of all cancer cases. Evidence on the relation of each to cancer is summarized, including esophageal, thyroid, colon, renal, liver, melanoma, multiple myeloma, rectum, gallbladder, leukemia, lymphoma, and prostate in men; and postmenopausal breast and endometrium in women. Different mechanisms drive etiologic pathways for these cancers. Weight loss, particularly among postmenopausal women, reduces risk for breast cancer. Among cancer patients, data are less robust, but we note a long history of poor outcomes after breast cancer among obese women. While evidence on obesity and outcomes for other cancers is mixed, growing evidence points to benefits of physical activity for breast and colon cancers. Dosing of chemotherapy and radiation therapy among obese patients is discussed and the impact on therapy-related toxicity is noted. Guidelines for counseling patients for weight loss and increased physical activity are presented and supported by strong evidence that increased physical activity leads to improved quality of life among cancer survivors. The “Five A's” model guides clinicians through a counseling session: assess, advise, agree, assist, arrange. The burden of obesity on society continues to increase and warrants closer attention by clinicians for both cancer prevention and improved outcomes after diagnosis.
Collapse
Affiliation(s)
- Kathleen Y Wolin
- Department of Surgery, Washington University School of Medicine, and Siteman Cancer Center, Barnes Jewish Hospital, 660 S Euclid Avenue, St. Louis, Missouri 63110, USA.
| | | | | |
Collapse
|
44
|
Martin-Castillo B, Dorca J, Vazquez-Martin A, Oliveras-Ferraros C, Lopez-Bonet E, Garcia M, Del Barco S, Menendez JA. Incorporating the antidiabetic drug metformin in HER2-positive breast cancer treated with neo-adjuvant chemotherapy and trastuzumab: an ongoing clinical-translational research experience at the Catalan Institute of Oncology. Ann Oncol 2009; 21:187-9. [PMID: 19884247 DOI: 10.1093/annonc/mdp494] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|