Metformin in combination with pemetrexed significantly strengthens the apoptosis of the three tested NSCLC cell lines. (A) Representative apoptosis results of A549, HCC827, and H1975 cell lines under different treatments. (B-D) Quantitative analysis of cell apoptosis in A549, HCC827, H1975 cell lines treated with metformin, pemetrexed and the combination. (E-G) Metformin and pemetrexed alone or the combination group separately showed no difference on A549, HCC827, H1975 cell lines. *P < 0.05 compared with the corresponding control group; # P < 0.05 compared with Metf alone; ▲ P < 0.05 compared with Pem alone. 

Metformin in combination with pemetrexed significantly strengthens the apoptosis of the three tested NSCLC cell lines. (A) Representative apoptosis results of A549, HCC827, and H1975 cell lines under different treatments. (B-D) Quantitative analysis of cell apoptosis in A549, HCC827, H1975 cell lines treated with metformin, pemetrexed and the combination. (E-G) Metformin and pemetrexed alone or the combination group separately showed no difference on A549, HCC827, H1975 cell lines. *P < 0.05 compared with the corresponding control group; # P < 0.05 compared with Metf alone; ▲ P < 0.05 compared with Pem alone. 

Source publication
Article
Full-text available
The aim of this study was to investigate whether metformin in combination with pemetrexed has an effect on the treatment of non-small-cell lung cancer (NSCLC) models and to explore the related molecular mechanism. The half maximal inhibitory concentration (IC50) and combination index (CI) of metformin and pemetrexed were detected by the CCK8 assay...

Contexts in source publication

Context 1
... Figure 2A-D, we found that apoptosis rates of the A549, HCC827, and H1975 cell lines treated by metformin were 10.40 ± 0.57%, 16.28 ± 1.21%, and 12.68 ± 1.67%. The apoptosis rates of the A549, HCC827, and H1975 cell lines treated by pemetrexed were 14.26 ± 1.17%, 14.65 ± 0.84%, and 13.22 ± 1.60%. ...
Context 2
... combination therapy had a further synergistic effect on enhancing apoptosis of the three NSCLC cell lines compared with the single drug treatment (P < 0.001). In Figure 2E, there were no obvious differences in cell apoptosis among the A549, HCC827, and H1975 cells treated with metformin alone as well as pemetrexed alone (Fig. 2F). Similarly, for the combination treatment, there were no obvious differences in cell apop- tosis among A549, HCC827, and H1975 cells (Fig. 2G). ...
Context 3
... and 28.54 ± 4.07%. The combination therapy had a further synergistic effect on enhancing apoptosis of the three NSCLC cell lines compared with the single drug treatment (P < 0.001). In Figure 2E, there were no obvious differences in cell apoptosis among the A549, HCC827, and H1975 cells treated with metformin alone as well as pemetrexed alone (Fig. 2F). Similarly, for the combination treatment, there were no obvious differences in cell apop- tosis among A549, HCC827, and H1975 cells (Fig. ...
Context 4
... the single drug treatment (P < 0.001). In Figure 2E, there were no obvious differences in cell apoptosis among the A549, HCC827, and H1975 cells treated with metformin alone as well as pemetrexed alone (Fig. 2F). Similarly, for the combination treatment, there were no obvious differences in cell apop- tosis among A549, HCC827, and H1975 cells (Fig. ...

Citations

... Previous studies reported that pemetrexed could inhibit NSCLC progression by reducing cell proliferation via inducing cycle arrest [24,25]. Moreover, ROS-mediated oxidative stress also contributed to cytotoxicity of pemetrexed to NSCLC cells [26,27]. ...
Article
Full-text available
Pemetrexed is a folate analog metabolic inhibitor that is given for therapy of non-small cell lung cancer (NSCLC). Drug resistance affects the efficacy of pemetrexed in NSCLC. Lentinan is a polysaccharide extracted from Shiitake mushrooms which has antitumor roles in multiple cancers, including lung cancer. However, the effects of lentinan on pemetrexed resistance in NSCLC remain unclear. In present study, The pemetrexed-resistant NSCLC cells were established and exposed to pemetrexed and lentinan. Oxidative stress was investigated via mitochondrial membrane potential (JC-1 staining), levels of MDA and SOD.The phosphorylation and total of PI3K and Akt levels were actuated using specific activator 740Y-P and measured through western blot. We observed that Lentinan decreased IC50 of pemetrexed in resistant NSCLC cells. Lentinan aggravated pemetrexed-induced proliferation inhibition of resistant NSCLC cells via reducing PCNA levels. Lentinan exacerbated pemetrexed-triggered oxidative stress through increasing ROS and MDA levels, and reducing mitochondrial membrane potential and SOD levels. Lentinan inhibited PI3K/Akt signaling activation in pemetrexed-treated cells. Activated PI3K/Akt pathway using activator 740Y-P reversed the effects of lentinan on pemetrexed-mediated proliferation inhibition and oxidative stress. Our findings uncover that Lentinan mitigates pemetrexed resistance in NSCLC through inhibiting cell proliferation and inducing oxidative stress by suppressing PI3K/Akt signaling.
... AMPK activity can impede the mTOR signaling pathway, ultimately suppressing tumor cell proliferation. [40][41][42][43] With the shift in the therapeutic focus of NSCLC towards immune checkpoint inhibition, it is imperative to investigate the interaction between metformin and the immune system. In murine models, metformin was found to directly affect CD-81 T cells, which prevented apoptosis of CD81 TILs in the tumor microenvironment, independent of programmed cell death protein 1 (PD-1) and TIM-3 expression. ...
Article
Full-text available
Background: Currently, the anticancer effects of metformin on different types of lung cancer have been frequently studied. However, the relationship between metformin and prognosis in nondiabetic patients with lung cancer remains controversial. To systematically evaluate the efficacy of metformin adjunctive therapy as the treatment for nondiabetic patients with advanced non-small cell lung cancer (NSCLC) to provide an evidence-based reference for clinical medication. Materials and methods: The literatures related to Phase II or III randomized controlled trials (RCTs) of metformin adjunctive therapy in nondiabetic patients with advanced NSCLC, including EMBASE, PubMed, the Cochrane Library, and Scopus database, were retrieved by computer, and the search time ranged from January 2017 to August 2022. The risk of bias assessment tool recommended by Cochrane Systematic Evaluator Manual 5.1.0 was used to evaluate the quality of the RCTs included. Rev Man 5.3 software and STATA15.0 were used for meta-analysis. Results: A total of 8 studies were included (925 patients). Meta-analysis results showed that there were no significant differences in progression-free survival (PFS) (hazard ratio [HR] = 0.95, 95% confidence interval [CI]: 0.66-1.36, P = 0.77), overall survival (OS) (HR = 0.89, 95% CI: 0.61-1.30, P = 0.55, n =7), objective response rate (ORR) (odds ratio [OR] = 1.37, 95% CI: 0.76-2.46, P = 0.30), and 1-year PFS rate (OR = 0.87, 95% CI: 0.39-1.94, P = 0.73, n = 3). Sensitivity analysis showed that PFS and OS indexes were stable. Conclusion: Metformin adjunctive therapy can improve the DCR of nondiabetic patients with advanced NSCLC. In addition, the patients cannot obtain a prolonged PFS, OS, 1-year PFS rate, and higher ORR rate.
... It is commonly believed that metformin exerts its anti-cancer activity via the LKB1-AMPK-mTOR pathway, and the overexpression of mTOR in this pathway is often associated with the development of numerous diseases such as tumors, among others [86][87][88][89][90]. Metformin activates AMPK after the inhibition of ETC I, inhibits the expression of mTOR and further inhibits the expression of more important factors mediating downstream procarcinogenic pathways and the process of tumor development, such as nuclear factor kappa B (NFkB), interleukin-6 (IL6), mitogen-activated protein kinase (MAPK), Ras and c-MYC [91][92][93]. ...
Article
Full-text available
Hematologic malignancies (HMs) mainly include acute and chronic leukemia, lymphoma, myeloma and other heterogeneous tumors that seriously threaten human life and health. The common effective treatments are radiotherapy, chemotherapy and hematopoietic stem cell transplantation (HSCT), which have limited options and are prone to tumor recurrence and (or) drug resistance. Metformin is the first-line drug for the treatment of type 2 diabetes (T2DM). Recently, studies identified the potential anti-cancer ability of metformin in both T2DM patients and patients that are non-diabetic. The latest epidemiological and preclinical studies suggested a potential benefit of metformin in the prevention and treatment of patients with HM. The mechanism may involve the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway by metformin as well as other AMPK-independent pathways to exert anti-cancer properties. In addition, combining current conventional anti-cancer drugs with metformin may improve the efficacy and reduce adverse drug reactions. Therefore, metformin can also be used as an adjuvant therapeutic agent for HM. This paper highlights the anti-hyperglycemic effects and potential anti-cancer effects of metformin, and also compiles the in vitro and clinical trials of metformin as an anti-cancer and chemosensitizing agent for the treatment of HM. The need for future research on the use of metformin in the treatment of HM is indicated.
... One study has found that metformin combined with cisplatin and radiation can further reduce cell proliferation by about 55-60%, significantly stronger than that of each drug alone [19], and one other study has uncovered that metformin combined with cisplatin can not only significantly inhibit cell activity but also induce cell apoptosis [20], which all imply that metformin combined with cisplatin has a strong effect on the biological mechanism of cancer cells. One laboratory study has revealed that metformin inhibits the proliferation of cancer cells and induces their apoptosis [21], and other studies have also concluded that metformin has direct antitumor effect and may inhibit tumor proliferation and induce apoptosis of tumor cells [22,23]. ...
Article
Full-text available
Background: Nasopharyngeal carcinoma (NPC) is an invasive squamous cell carcinoma located in the nasopharynx. NPC has a high recurrence risk after initial treatment due to its high metastatic and immune escape potential. One study has found that metformin can improve cancer outcomes and reduce cancer incidence. Objective: With antitumor activity, metformin can have low toxicity when used in combination with some common chemotherapy drugs. This study was designed to explore the effects of cisplatin combined with metformin on the proliferation and apoptosis of nasopharyngeal carcinoma (NPC) cells. Methods: An appropriate cisplatin concentration was selected for NPC cells, and the cells were treated with metformin at a gradient concentration, and then, some of them were treated with cisplatin. Subsequently, the biological effects (activity, migration, invasion, and apoptosis) of metformin alone and metformin combined with cisplatin on NPC cells were evaluated. Results: Metformin alone inhibited cell activity, migration, and invasion and promoted cell apoptosis in a concentration-dependent and time-dependent manner, while compared with cisplatin alone, cisplatin combined with metformin had stronger inhibition on cell activity, migration, and invasion and stronger induction to cell apoptosis, and a higher concentration of them demonstrated stronger effects. Conclusion: Cisplatin combined with metformin can strongly inhibit the activity of NPC cells and promote their apoptosis.
... In addition, metformin could induce apoptosis by the AMPK-dependent PKA (protein kinase A)/GSK-3β (glycogen synthase kinase-3beta) pathway in NSCLC [40]. In an in vitro study of a lung cancer cell line provided by Zhang et al. [41], the combination of metformin with pemetrexed showed a synergistic anti-tumorigenesis effect by altering the cell cycle and enhancing apoptosis. Other AMPK-independent pathways, such as protein phosphatase 2 (PP2A), interferon regulatory factor-1 (IRF-1), hepatocyte growth factor (HGF), and autophagy, have been investigated [42]. ...
Article
Full-text available
Lung cancer remains a challenge in daily practice. Chemotherapy is first considered for advanced lung adenocarcinoma bearing no active driver mutations. Maintaining drug efficacy and overcoming drug resistance are essential. This study aimed to explore the real-world use of anti-diabetic agent metformin in combination with pemetrexed-based platinum doublets in a first-line setting. We retrospectively collected data during 2004~2013 from TaiwaN′s National Health Insurance Research Database to access the survival benefit of metformin combined with pemetrexed-based platinum doublets as a first-line therapy for diabetic patients with advanced lung adenocarcinoma. Demographic data and information regarding platinum reagents, diabetes medications, and metformin doses were gathered, and overall survival status regarding metformin use was analyzed. Overall survival status based on the daily dose and the calculated cumulative defined daily dose (DDD) of metformin prescribed during the first 3 months after lung cancer was diagnosed was also assessed. A total of 495 patients were enrolled with a mean age of 67 years old, and the majority of the patients were male. After adjusting for age, sex, diabetes medication, and platinum reagents used, the adjusted hazard ratio (HR) for the metformin-user group was 0.61 (95% confidence interval (CI); 0.46~0.79; p < 0.001). The metformin-user group had a survival benefit (log-rank p < 0.001). We analyzed metformin dosing during the first 3 months after lung cancer diagnosis, and for a daily dose ≥ 1,500 mg, the adjusted hazard ratio (aHR) was 0.42 (95% CI; 0.27~0.65; p < 0.001). Regarding the cumulative DDD of metformin, a DDD equal to or exceeding 21 resulted in aHR of 0.48 (95% CI; 0.34~0.69; p < 0.001). In this study, we found that the combination of metformin and pemetrexed-based platinum doublets provides a robust survival benefit as a first-line therapy for diabetic patients with advanced lung adenocarcinoma. It is worth conducting a large and randomized clinical trial to further investigate the antitumor effects of metformin on advanced lung adenocarcinoma when used as a first-ling therapy, including in non-diabetic patients.
... Few studies have focused on the antitumor mechanism induced by metformin combined with pemetrexed. Although a lung cancer cell line in vitro study performed by Zhang et al. (2017) proposed antiproliferative and apoptosis processes, there is still a lack of in vivo studies and real-time imaging monitoring to expand and elucidate the possible mechanism besides the AMPK-dependent pathway. Therefore, we set up serial experiments and platforms in in vitro, ex vivo and in vivo models to identify potential additive and novel therapeutic targets, such as angiogenesis, in combination therapy with metformin and pemetrexed in non-small-cell lung cancer. ...
... Mounting evidence have shown the half maximal inhibitory concentration (IC50) of metformin and pemetrexed in NSCLC cell lines, the IC50 values at 48 h of metformin was 1-10 mM and the pemetrexed was 1.5-3.5 µM (Zhang et al., 2017). To investigate the potential mechanism by which lower dosage of metformin and pemetrexed influence NSCLC cell growth, we analyzed the cell cycle and apoptosis. ...
... Metformin can also enhance p53-mediated cell cycle arrest via the AMPK-related apoptosis pathway (Buzzai et al., 2007;Emami Riedmaier et al., 2013). In an in vitro study of A549, H1975, and HCC827 lung cancer cell lines, Zhang et al. (2017) found that metformin combined with pemetrexed not only inhibited cell proliferation but also induced cell apoptosis, as validated by western blot analysis of reduced levels of an antiapoptotic molecule (Bcl-2) and elevated levels of a proapoptotic molecule (Bax). In our study, we showed a similar result as Gong et al. described (Steiner et al., 2007) that the single uses of low dosage of metformin show no antiproliferative or apoptotic effect on A549 cells. ...
Article
Full-text available
Lung cancer is heterogeneous and challenging to cope with once it has progressed. Chemotherapy is the first step once no active driver mutation has been discovered. Non-antitumor drugs have been found to be beneficial when used as adjuvants to chemotherapy. In this study, the additive effect and mechanism of metformin combined with pemetrexed in non-small-cell lung cancer (NSCLC) cells were elucidated. Three NSCLC cell lines, A549, H1975, and HCC827, were used to analyze tumor cell proliferation, colony formation and the cell cycle in vitro when exposed to metformin alone, pemetrexed alone or their combination. We found that combination treatment in three cell lines exerted antiproliferative effects through cell cycle arrest in the S phase. An ex vivo chicken chorioallantoic membrane (CAM) assay was used to examine the antiangiogenic effect of metformin combined with pemetrexed on vascular structure formation. We further created an A549 orthotopic xenograft model with an in vivo imaging system (IVIS) and explored the associated indicators involved in the tumorigenic process. The in vitro results showed that the combination of metformin and pemetrexed exhibited an antiproliferative effect in reducing cell viability and colony formation, the downregulation of cyclin D1 and A2 and the upregulation of CDKN1B, which are involved in the G1/S phase. For antiangiogenic effects, the combination therapy inhibited the vascular structure, as proven by the CAM assay. We elucidated that combination therapy could target VEGFA and Endoglin by RT-qPCR, ELISA and histopathological findings in an A549 orthotopic NSCLC xenograft model. Our research demonstrated the additive antiproliferative and antiangiogenic effects of the combination of metformin with pemetrexed in NSCLC and could be applied to clinical lung cancer therapy.
... Even though several anticarcinogenic effects of metformin are observed, the clinical data are still contentious depending on disease-related (type of tumor, clinical stage, form of treatment) and on patient-related factors (insulin resistance, age, sex) [21]. For in vitro experiments, the combination of metformin with several chemotherapeutic agents shows controversial results ranging from synergistic effects [22][23][24] to even adverse effects [25,26]. So far, these investigations underline the need for a more detailed understanding of the molecular mechanisms that occur when combining metformin with chemotherapeutics before applying it as a potential adjuvant in chemotherapy. ...
Article
Full-text available
Background Despite considerable medical proceedings, cancer is still a leading cause of death. Major problems for tumor therapy are chemoresistance as well as toxic side effects. In recent years, the additional treatment with the antidiabetic drug metformin during chemotherapy showed promising results in some cases. The aim of this study was to develop an in vitro tumor therapy model in order to further investigate the potential of a combined chemotherapy with metformin. Methods Cytotoxic effects of a combined treatment on BALB/c fibroblasts were proven by the resazurin assay. Based on the BALB/c cell transformation assay, the BALB/c tumor therapy model was established successfully with four different and widely used chemotherapeutics from different categories. Namely, Doxorubicin as a type-II isomerase inhibitor, Docetaxel as a spindle toxin, Mitomycin C as an alkylating agent and 5-Fluorouracil as an antimetabolite. Moreover, glucose consumption in the medium supernatant was measured and protein expressions were determined by Western Blotting. Results Initial tests for the combined treatment with metformin indicated unexpected results as metformin could partly mitigate the cytotoxic effects of the chemotherapeutic agents. These results were further confirmed as metformin induced resistance to some of the drugs when applied simultaneously in the tumor therapy model. Mechanistically, an increased glucose consumption was observed in non-transformed cells as well as in the mixed population of malignant transformed cell foci and non-transformed monolayer cells, suggesting that metformin could also increase glucose consumption in transformed cells. Conclusion In conclusion, this study suggests a cautious use of metformin during chemotherapy. Moreover, the BALB/c tumor therapy model offers a potent tool for further mechanistic studies of drug-drug interactions during cancer therapy. Graphical abstract
... Notably, synergisms with metformin has been reported with numerous anticancer agents and modalities including chemotherapy [35], targeted drugs [36], and radiotherapy [37]. In the past ten years alone, metformin synergism with chemotherapies pemetrexed [38], temozolomide [39], cisplatin [40], gemcitabine [41], paclitaxel [42], 5FU [43], vincristine [44] with targeted agents erlotinib against non-small cell lung cancer [45], imatinib against colon cancer [46], gefitinib against bladder cancer [47], trastuzumab against human epidermal growth factor receptor 2 (HER2) positive breast cancer [48], celecoxib against NSCLC [49], regorafenib against liver cancer [50], with everolimus as neuroendocrine cancers [51]; and other anticancer agents such as with nelfinavir against cervical cancer [52], propranolol against breast cancer [53], 2-deoxyglucose against ovarian cancer [54], arsenic trioxide against cholangiocarcinoma [55], and with natural compounds epigallocatechin-3-gallate [56], curcumin [57], berberine [58], resveratrol [59]. ...
Chapter
Full-text available
Metformin is a cornerstone treatment of diabetes mellitus. Since 2005 when it has been first reported to reduce the risk of cancer in diabetics, a large number of preclinical and clinical studies have implicated its potential role as a preventative and adjunct therapy for a broad range of cancers. Whereas preclinical studies demonstrate its actions on a multitude of molecular pathways involving nearly all aspects of cancer development including metabolism, angiogenesis, apoptosis, autophagy, immunity, epigenetics, inflammation and crosstalk with the microbi-ome, other studies demonstrate its synergism with a range of anticancer modalities including chemotherapy, radiotherapy, immunotherapy, and targeted therapies. Furthermore, an increasing number of clinical studies not only confirm its preventative properties against cancers but have extended its potential for a possible adjunctive role in the neoadjuvant, adjuvant, maintenance and salvage therapies of cancer. This article intends to summarize the basic science that allows us to understand the complex multiple mechanisms of action of this remarkable multitasking molecule as well as review the recent meta-analyses that have summarized the clinical studies assessing the therapeutic efficacy of metformin for various cancers.
... Even though several anticarcinogenic effects of metformin are observed, the clinical data are still contentious depending on disease-related (type of tumor, clinical stage, form of treatment) and on patient-related factors (insulin resistance, age, sex) (21). For in vitro experiments, the combination of metformin with several chemotherapeutic agents shows controversial results ranging from synergistic effects (22)(23)(24) to even adverse effects (25,26). So far, these investigations underline the need for a more detailed understanding of the molecular mechanisms that occur when combining metformin with chemotherapeutics before applying it as a potential adjuvant in chemotherapy. ...
Preprint
Full-text available
Background Despite considerable medical proceedings, cancer is still a leading cause of death. Major problems for tumor therapy are chemoresistance as well as toxic side effects. In recent years, the additional treatment with the antidiabetic drug metformin during chemotherapy showed promising results in some cases. The aim of this study was to develop an in vitro tumor therapy model in order to further investigate the potential of a combined chemotherapy with metformin. Methods Cytotoxic effects of a combined treatment on BALB/c fibroblasts were proven by the resazurin assay. Based on the BALB/c cell transformation assay, the BALB/c tumor therapy model was established successfully with four different chemotherapeutics (Doxorubicin, Docetaxel, Mitomycin C and 5-Fluorouracil). Moreover, glucose consumption in the medium supernatant was measured and protein expressions were determined by Western Blotting. Results Initial tests for the combined treatment with metformin indicated unexpected results as metformin could partly mitigate the cytotoxic effects of the chemotherapeutic agents. These results were further confirmed as metformin induced resistance to some of the drugs when applied simultaneously in the tumor therapy model. Mechanistically, an increased glucose consumption was observed in non-transformed cells as well as in the mixed population of malignant transformed cell foci and non-transformed monolayer cells, suggesting that metformin could also increase glucose consumption in transformed cells. Conclusion In conclusion, this study suggests a cautious use of metformin during chemotherapy. Moreover, the BALB/c tumor therapy model offers a potent tool for further mechanistic studies of drug-drug interactions during cancer therapy.
... Different genotypes may also have different impacts on the response to combination therapy. It was reported that metformin in combination with pemetrexed significantly altered the cell cycle distribution of a certain adenocarcinoma cell line [104]. Tumor genetic profiling is required to identify patients most likely to benefit from metformin treatment. ...
Article
Full-text available
Metformin is one of the most commonly used first-line oral medications for type 2 diabetes mellitus. Multiple observational studies, reviewed in numerous systematic reviews, have shown that metformin treatment may not only reduce the risk of cancer but may also improve the efficacy of cancer treatment in diabetic patients. Recent studies have been conducted to determine whether a similar protective effect can be demonstrated in nondiabetic cancer patients. However, the results are controversial. The potential optimal dose, schedule, and duration of metformin treatment and the heterogeneity of histological subtypes and genotypes among cancer patients might contribute to the different clinical benefits. In addition, as the immune property of metformin was investigated, further studies of the immunomodulatory effect of metformin on cancer cells should also be taken into account to optimize its clinical use. In this review, we present and discuss the latest findings regarding the anticancer potential of metformin in nondiabetic patients with cancer.