Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 530
Filter
1.
Cells ; 12(24)2023 12 18.
Article in English | MEDLINE | ID: mdl-38132178

ABSTRACT

The effect of agonists on AMP-activated protein kinase (AMPK), mainly metformin and phenformin, has been appreciated in the treatment of multiple types of tumors. Specifically, the antitumor activity of phenformin has been demonstrated in melanomas containing the v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) activating mutation. In this report, we elucidated the synergistic antitumor effects of biguanides with metabolism inhibitors on colon tumors. Phenformin with 2-deoxy-D-glucose (2DG) inhibited tumor cell growth in cancer cell lines, including HT29 cells harboring BRAF- and p53-mutations. Biochemical analyses showed that two chemotherapeutics exerted cooperative effects to reduce tumor growth through cell cycle arrest, apoptosis, and autophagy. The drugs demonstrated activity against phosphorylated ERK and the gain-of-function p53 mutant protein. To demonstrate tumor regressive effects in vivo, we established patient-derived models, including xenograft (PDX) and organoids (PDO). Co-treatment of biguanides with chemotherapeutics efficiently reduced the growth of patient-derived colon models in comparison to treatment with a single agent. These results strongly suggest that significant therapeutic advantages would be achieved by combining AMPK activators such as phenformin and cancer metabolic inhibitors such as 2DG.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Metformin , Animals , Mice , Humans , Phenformin/pharmacology , Phenformin/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Tumor Suppressor Protein p53 , AMP-Activated Protein Kinases/metabolism , Drug Repositioning , Colonic Neoplasms/drug therapy , Metformin/pharmacology , Metformin/therapeutic use
2.
Adv Biol (Weinh) ; 7(7): e2300080, 2023 07.
Article in English | MEDLINE | ID: mdl-37303292

ABSTRACT

Lung cancer is one of the most fatal cancers worldwide. Resistance to conventional therapies remains a hindrance to patient treatment. Therefore, the development of more effective anti-cancer therapeutic strategies is imperative. Solid tumors exhibit a hyperglycolytic phenotype, leading to enhanced lactate production; and, consequently, its extrusion to the tumor microenvironment. Previous data reveals that inhibition of CD147, the chaperone of lactate transporters (MCTs), decreases lactate export in lung cancer cells and sensitizes them to phenformin, leading to a drastic decrease in cell growth. In this study, the development of anti-CD147 targeted liposomes (LUVs) carrying phenformin is envisioned, and their efficacy is evaluated to eliminate lung cancer cells. Herein, the therapeutic effect of free phenformin and anti-CD147 antibody, as well as the efficacy of anti-CD147 LUVs carrying phenformin on A549, H292, and PC-9 cell growth, metabolism, and invasion, are evaluated. Data reveals that phenformin decreases 2D and 3D-cancer cell growth and that the anti-CD147 antibody reduces cell invasion. Importantly, anti-CD147 LUVs carrying phenformin are internalized by cancer cells and impaired lung cancer cell growth in vitro and in vivo. Overall, these results provide evidence for the effectiveness of anti-CD147 LUVs carrying phenformin in compromising lung cancer cell aggressiveness.


Subject(s)
Lung Neoplasms , Phenformin , Humans , Phenformin/pharmacology , Phenformin/therapeutic use , Lung Neoplasms/drug therapy , Cell Proliferation , Lactates/pharmacology , Lactates/therapeutic use , Tumor Microenvironment
3.
Cells ; 11(15)2022 08 05.
Article in English | MEDLINE | ID: mdl-35954273

ABSTRACT

The treatment of many skin inflammation diseases, such as psoriasis and atopic dermatitis, is still a challenge and inflammation plays important roles in multiple stages of skin tumor development, including initiation, promotion and metastasis. Phenformin, a biguanide drug, has been shown to play a more efficient anti-tumor function than another well-known biguanide drug, metformin, which has been reported to control the expression of pro-inflammatory cytokines; however, little is known about the effects of phenformin on skin inflammation. This study used a mouse acute inflammation model, ex vivo skin organ cultures and in vitro human primary keratinocyte cultures to demonstrate that phenformin can suppress acute skin inflammatory responses induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) in vivo and significantly suppresses the pro-inflammatory cytokines IL-1ß, IL-6 and IL-8 in human primary keratinocytes in vitro. The suppression of pro-inflammatory cytokine expression by phenformin was not directly through regulation of the MAPK or NF-κB pathways, but by controlling the expression of c-Myc in human keratinocytes. We demonstrated that the overexpression of c-Myc can induce pro-inflammatory cytokine expression and counteract the suppressive effect of phenformin on cytokine expression in keratinocytes. In contrast, the down-regulation of c-Myc produces effects similar to phenformin, both in cytokine expression by keratinocytes in vitro and in skin inflammation in vivo. Finally, we showed that phenformin, as an AMPK activator, down-regulates the expression of c-Myc through regulation of the AMPK/mTOR pathways. In summary, phenformin inhibits the expression of pro-inflammatory cytokines in keratinocytes through the down-regulation of c-Myc expression to play an anti-inflammation function in the skin.


Subject(s)
Cytokines , Dermatitis, Atopic , Proto-Oncogene Proteins c-myc/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cytokines/metabolism , Dermatitis, Atopic/metabolism , Humans , Inflammation/metabolism , Keratinocytes/metabolism , Mice , Phenformin/pharmacology , Phenformin/therapeutic use
4.
Invest New Drugs ; 40(3): 576-585, 2022 06.
Article in English | MEDLINE | ID: mdl-35015172

ABSTRACT

BACKGROUND: Myeloproliferative neoplasms (MPN) are disorders characterized by an alteration at the hematopoietic stem cell (HSC) level, where the JAK2 mutation is the most common genetic alteration found in classic MPN (polycythemia vera, essential thrombocythemia, and primary myelofibrosis). We and others previously demonstrated that metformin reduced splenomegaly and platelets counts in peripheral blood in JAK2V617F pre-clinical MPN models, which highlighted the antineoplastic potential of biguanides for MPN treatment. Phenformin is a biguanide that has been used to treat diabetes, but was withdrawn due to its potential to cause lactic acidosis in patients. AIMS: We herein aimed to investigate the effects of phenformin in MPN disease burden and stem cell function in Jak2V617F-knockin MPN mice. RESULTS: In vitro phenformin treatment reduced cell viability and increased apoptosis in SET2 JAK2V67F cells. Long-term treatment with 40 mg/kg phenformin in Jak2V617F knockin mice increased the frequency of LSK, myeloid progenitors (MP), and multipotent progenitors (MPP) in the bone marrow. Phenformin treatment did not affect peripheral blood counts, spleen weight, megakaryocyte count, erythroid precursors frequency, or ex vivo clonogenic capacity. Ex vivo treatment of bone marrow cells from Jak2V617F knockin mice with phenformin did not affect hematologic parameters or engraftment in recipient mice. CONCLUSIONS: Phenformin increased the percentages of LSK, MP, and MPP populations, but did not reduce disease burden in Jak2V617F-knockin mice. Additional studies are necessary to further understand the effects of phenformin on early hematopoietic progenitors.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Animals , Bone Marrow , Disease Models, Animal , Humans , Janus Kinase 2 , Mice , Mutation , Myeloproliferative Disorders/drug therapy , Phenformin/pharmacology , Phenformin/therapeutic use , Polycythemia Vera/genetics
5.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33762304

ABSTRACT

MYCN-amplified neuroblastoma is a lethal subset of pediatric cancer. MYCN drives numerous effects in the cell, including metabolic changes that are critical for oncogenesis. The understanding that both compensatory pathways and intrinsic redundancy in cell systems exists implies that the use of combination therapies for effective and durable responses is necessary. Additionally, the most effective targeted therapies exploit an "Achilles' heel" and are tailored to the genetics of the cancer under study. We performed an unbiased screen on select metabolic targeted therapy combinations and correlated sensitivity with over 20 subsets of cancer. We found that MYCN-amplified neuroblastoma is hypersensitive to the combination of an inhibitor of the lactate transporter MCT1, AZD3965, and complex I of the mitochondrion, phenformin. Our data demonstrate that MCT4 is highly correlated with resistance to the combination in the screen and lowly expressed in MYCN-amplified neuroblastoma. Low MCT4 combines with high expression of the MCT2 and MCT1 chaperone CD147 in MYCN-amplified neuroblastoma, altogether conferring sensitivity to the AZD3965 and phenformin combination. The result is simultaneous disruption of glycolysis and oxidative phosphorylation, resulting in dramatic disruption of adenosine triphosphate (ATP) production, endoplasmic reticulum stress, and cell death. In mouse models of MYCN-amplified neuroblastoma, the combination was tolerable at concentrations where it shrank tumors and did not increase white-blood-cell toxicity compared to single drugs. Therefore, we demonstrate that a metabolic combination screen can identify vulnerabilities in subsets of cancer and put forth a metabolic combination therapy tailored for MYCN-amplified neuroblastoma that demonstrates efficacy and tolerability in vivo.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Monocarboxylic Acid Transporters/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/drug therapy , Symporters/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Basigin/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Electron Transport Complex I/metabolism , Gene Amplification , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Monocarboxylic Acid Transporters/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Phenformin/pharmacology , Phenformin/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Symporters/metabolism , Thiophenes/pharmacology , Thiophenes/therapeutic use , Xenograft Model Antitumor Assays
6.
Laryngoscope ; 131(7): 1570-1577, 2021 07.
Article in English | MEDLINE | ID: mdl-32857885

ABSTRACT

OBJECTIVES: Iatrogenic laryngotracheal stenosis (iLTS) is the pathological narrowing of the glottis, subglottis, and/or trachea due to scar tissue. Patients with type 2 diabetes mellitus (T2DM) are over 8 times more likely to develop iLTS and represent 26% to 53% of all iLTS patients. In this investigation, we compared iLTS scar-derived fibroblasts in patients with and without T2DM. STUDY DESIGN: Controlled ex vivo study. METHODS: iLTS scar fibroblasts were isolated and cultured from subglottic scar biopsies in iLTS patients diagnosed with or without type 2 diabetes (non-T2DM). Fibroblast proliferation, fibrosis-related gene expression, and metabolic utilization of oxidative phosphorylation (OXPHOS) and glycolysis were assessed. Contractility was measured using a collagen-based assay. Metabolically targeted drugs (metformin, phenformin, amobarbital) were tested, and changes in fibrosis-related gene expression, collagen protein, and contractility were evaluated. RESULTS: Compared to non-T2DM, T2DM iLTS scar fibroblasts had increased α-smooth muscle actin (αSMA) expression (8.2× increased, P = .020), increased contractility (mean 71.4 ± 4.3% vs. 51.7 ± 16% Δ area × 90 minute-1 , P = .016), and reduced proliferation (1.9× reduction at 5 days, P < .01). Collagen 1 (COL1) protein was significantly higher in the T2DM group (mean 2.06 ± 0.19 vs. 0.74 ±.44 COL1/total protein [pg/µg], P = .036). T2DM iLTS scar fibroblasts had increased measures of OXPHOS, including basal respiration (mean 86.7 vs. 31.5 pmol/minute/10 µg protein, P = .016) and adenosine triphosphate (ATP) generation (mean 97.5 vs. 25.7 pmol/minute/10 µg protein, P = .047) compared to non-T2DM fibroblasts. Amobarbital reduced cellular contractility; decreased collagen protein; and decreased expression of αSMA, COL1, and fibronectin. Metformin and phenformin did not significantly affect fibrosis-related gene expression. CONCLUSION: T2DM iLTS scar fibroblasts demonstrate a myofibroblast phenotype and greater contractility compared to non-T2DM. Their bioenergetic preference for OXPHOS drives their increased contractility, which is selectively targeted by amobarbital. LEVEL OF EVIDENCE: NA Laryngoscope, 131:1570-1577, 2021.


Subject(s)
Cicatrix/pathology , Diabetes Mellitus, Type 2/complications , Laryngostenosis/pathology , Myofibroblasts/pathology , Tracheal Stenosis/pathology , Adult , Aged , Amobarbital/pharmacology , Biopsy , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Cicatrix/etiology , Constriction, Pathologic/etiology , Constriction, Pathologic/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Female , Glottis/cytology , Glottis/injuries , Glottis/pathology , Glycolysis/drug effects , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Iatrogenic Disease , Intubation, Intratracheal/adverse effects , Laryngostenosis/etiology , Male , Metformin/pharmacology , Metformin/therapeutic use , Middle Aged , Muscle Contraction/drug effects , Myofibroblasts/metabolism , Oxidative Phosphorylation/drug effects , Phenformin/pharmacology , Phenformin/therapeutic use , Primary Cell Culture , Trachea/cytology , Trachea/injuries , Trachea/pathology , Tracheal Stenosis/etiology , Tracheostomy/adverse effects , Young Adult
7.
Cells ; 9(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32883024

ABSTRACT

The greatest challenge in cancer therapy is posed by drug-resistant recurrence following treatment. Anticancer chemotherapy is largely focused on targeting the rapid proliferation and biosynthesis of cancer cells. This strategy has the potential to trigger autophagy, enabling cancer cell survival through the recycling of molecules and energy essential for biosynthesis, leading to drug resistance. Autophagy recycling contributes amino acids and ATP to restore mTOR complex 1 (mTORC1) activity, which leads to cell survival. However, autophagy with mTORC1 activation can be stalled by reducing the ATP level. We have previously shown that cytosolic NADH production supported by aldehyde dehydrogenase (ALDH) is critical for supplying ATP through oxidative phosphorylation (OxPhos) in cancer cell mitochondria. Inhibitors of the mitochondrial complex I of the OxPhos electron transfer chain and ALDH significantly reduce the ATP level selectively in cancer cells, terminating autophagy triggered by anticancer drug treatment. With the aim of overcoming drug resistance, we investigated combining the inhibition of mitochondrial complex I, using phenformin, and ALDH, using gossypol, with anticancer drug treatment. Here, we show that OxPhos targeting combined with anticancer drugs acts synergistically to enhance the anticancer effect in mouse xenograft models of various cancers, which suggests a potential therapeutic approach for drug-resistant cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Gossypol/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidative Phosphorylation/drug effects , Phenformin/therapeutic use , Aldehyde Dehydrogenase/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Drug Synergism , Electron Transport Complex I/antagonists & inhibitors , Gossypol/pharmacology , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/pathology , Phenformin/pharmacology , Xenograft Model Antitumor Assays
8.
Cell Rep ; 30(6): 1735-1752.e7, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049007

ABSTRACT

The antidiabetic drug phenformin displays potent anticancer activity in different tumors, but its mechanism of action remains elusive. Using Shh medulloblastoma as model, we show here that at clinically relevant concentrations, phenformin elicits a significant therapeutic effect through a redox-dependent but complex I-independent mechanism. Phenformin inhibits mitochondrial glycerophosphate dehydrogenase (mGPD), a component of the glycerophosphate shuttle, and causes elevations of intracellular NADH content. Inhibition of mGPD mimics phenformin action and promotes an association between corepressor CtBP2 and Gli1, thereby inhibiting Hh transcriptional output and tumor growth. Because ablation of CtBP2 abrogates the therapeutic effect of phenformin in mice, these data illustrate a biguanide-mediated redox/corepressor interplay, which may represent a relevant target for tumor therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Co-Repressor Proteins/drug effects , Hedgehog Proteins/drug effects , Hypoglycemic Agents/therapeutic use , Neoplasms/drug therapy , Phenformin/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Mice , Phenformin/pharmacology
9.
Melanoma Res ; 30(1): 1-13, 2020 02.
Article in English | MEDLINE | ID: mdl-31116160

ABSTRACT

Therapeutic failures in cancer therapy are often associated with metabolic plasticity. The use of metabolic modulators as anti-cancer agents has been effective in correcting metabolic alterations; however, molecular events behind metabolic switch are still largely unexplored. Herein, we characterize the molecular and functional events that follow prolonged oxidative phosphorylation inhibition by phenformin in order to study how melanoma cells adapt to this specific metabolic pressure. We show that melanoma cells cultured up to 3 months with high doses of phenformin (R-cells) are less viable and migrate and invade less than parental (S-) cells. Microarray analysis of R-melanoma cells reveals a switch in the energy production strategy accompanied by the modulation of several immunological-associated genes. R-cells display low oxygen consumption rate and high basal extracellular acidification rate. When treated with vemurafenib, R-cell viability, growth and extracellular signal-regulated kinase activation decrease. Finally, phenformin withdrawal reverts R-cells phenotype. In summary, our study provides an in vitro model of on-off metabolic switch in melanoma and reveals interesting molecular signatures controlling metabolic reprogramming in this tumour.


Subject(s)
Cellular Reprogramming/immunology , Hypoglycemic Agents/therapeutic use , Melanoma/genetics , Phenformin/therapeutic use , Humans , Hypoglycemic Agents/pharmacology , Phenformin/pharmacology
10.
Cancer Sci ; 110(9): 2834-2845, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31278880

ABSTRACT

Recurrence and chemoresistance in colorectal cancer remain important issues for patients treated with conventional therapeutics. Metformin and phenformin, previously used in the treatment of diabetes, have been shown to have anticancer effects in various cancers, including breast, lung and prostate cancers. However, their molecular mechanisms are still unclear. In this study, we examined the effects of these drugs in chemoresistant rectal cancer cell lines. We found that SW837 and SW1463 rectal cancer cells were more resistant to ionizing radiation and 5-fluorouracil than HCT116 and LS513 colon cancer cells. In addition, metformin and phenformin increased the sensitivity of these cell lines by inhibiting cell proliferation, suppressing clonogenic ability and increasing apoptotic cell death in rectal cancer cells. Signal transducer and activator of transcription 3 and transforming growth factor-ß/Smad signaling pathways were more activated in rectal cancer cells, and inhibition of signal transducer and activator of transcription 3 expression using an inhibitor or siRNA sensitized rectal cancer cells to chemoresistant by inhibition of the expression of antiapoptotic proteins, such as X-linked inhibitor of apoptosis, survivin and cellular inhibitor of apoptosis protein 1. Moreover, metformin and phenformin inhibited cell migration and invasion by suppression of transforming growth factor ß receptor 2-mediated Snail and Twist expression in rectal cancer cells. Therefore, metformin and phenformin may represent a novel strategy for the treatment of chemoresistant rectal cancer by targeting signal transducer and activator of transcription 3 and transforming growth factor-ß/Smad signaling.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Metformin/pharmacology , Phenformin/pharmacology , Rectal Neoplasms/therapy , Signal Transduction/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chemoradiotherapy/methods , Colon/pathology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Male , Metformin/therapeutic use , Mice , Mice, Nude , Neoplasm Recurrence, Local , Phenformin/therapeutic use , Rectal Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/radiation effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
11.
Cell Rep ; 27(3): 690-698.e4, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995468

ABSTRACT

AMPK acts downstream of the tumor suppressor LKB1, yet its role in cancer has been controversial. AMPK is activated by biguanides, such as metformin and phenformin, and metformin use in diabetics has been associated with reduced cancer risk. However, whether this is mediated by cell-autonomous AMPK activation within tumor progenitor cells has been unclear. We report that T-cell-specific loss of AMPK-α1 caused accelerated growth of T cell acute lymphoblastic leukemia/lymphoma (T-ALL) induced by PTEN loss in thymic T cell progenitors. Oral administration of phenformin, but not metformin, delayed onset and growth of lymphomas, but only when T cells expressed AMPK-α1. This differential effect of biguanides correlated with detection of phenformin, but not metformin, in thymus. Phenformin also enhanced apoptosis in T-ALL cells both in vivo and in vitro. Thus, AMPK-α1 can be a cell-autonomous tumor suppressor in the context of T-ALL, and phenformin may have potential for the prevention of some cancers.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Proliferation/drug effects , Hypoglycemic Agents/pharmacology , Phenformin/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , Administration, Oral , Animals , Disease Models, Animal , Disease-Free Survival , Female , Glycolysis/drug effects , Hypoglycemic Agents/therapeutic use , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phenformin/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Sirolimus/pharmacology
13.
Oncotarget ; 8(30): 50187-50192, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28418852

ABSTRACT

Phenformin's recently demonstrated efficacy in melanoma and Gleevec's demonstrated anti-proliferative action in chronic myeloid leukemia may lie within these drugs' significant pharmacokinetics, pharmacodynamics and structural homologies, which are reviewed herein. Gleevec's success in turning a fatal leukemia into a manageable chronic disease has been trumpeted in medical, economic, political and social circles because it is considered the first successful targeted therapy. Investments have been immense in omics analyses and while in some cases they greatly helped the management of patients, in others targeted therapies failed to achieve clinically stable recurrence-free disease course or to substantially extend survival. Nevertheless protein kinase controlling approaches have persisted despite early warnings that the targeted genomics narrative is overblown. Experimental and clinical observations with Phenformin suggest an alternative explanation for Gleevec's mode of action. Using 13C-guided precise flux measurements, a comparative multiple cell line study demonstrated the drug's downstream impact on submolecular fatty acid processing metabolic events that occurred independent of Gleevec's molecular target. Clinical observations that hyperlipidemia and diabetes are both reversed in mice and in patients taking Gleevec support the drugs' primary metabolic targets by biguanides and statins. This is evident by structural data demonstrating that Gleevec shows pyridine- and phenyl-guanidine homology with Phenformin and identical phenylcarbamoyl structural and ligand binding homology with Lipitor. The misunderstood mechanism of action of Gleevec is emblematic of the pervasive flawed reasoning that genomic analysis will lead to targeted, personalized diagnosis and therapy. The alternative perspective for Gleevec's mode of action may turn oncotargets towards metabolic channel reaction architectures in leukemia and melanoma, as well as in other cancers.


Subject(s)
Atorvastatin/therapeutic use , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Melanoma/drug therapy , Metformin/therapeutic use , Phenformin/therapeutic use , Atorvastatin/pharmacology , Humans , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Melanoma/pathology , Metformin/pharmacology , Phenformin/pharmacology
14.
Nanomedicine (Lond) ; 10(18): 2819-32, 2015.
Article in English | MEDLINE | ID: mdl-26377155

ABSTRACT

AIM: Phenformin-loaded micelles (Phen M) were used in combination with gemcitabine-loaded micelles (Gem M) to study their combined effect against H460 human lung cancer cells and cancer stem cells (CSCs) in vitro and in vivo. MATERIALS & METHODS: Gem M and Phen M were prepared via self-assembly of a mixture of a diblock copolymer of PEG and urea-functionalized polycarbonate (PEG-PUC) and a diblock copolymer of PEG and acid-functionalized polycarbonate (PEG-PAC) through hydrogen bonding and ionic interactions. Gem M and Phen M were characterized and tested for efficacy both in vitro and in vivo against cancer cells and CSCs. RESULTS: The combination of Gem M/Phen M exhibited higher cytotoxicity against CSCs and non-CSCs than Gem M and Phen M alone, and showed significant cell cycle growth arrest in vitro. The combination therapy had superior tumor suppression and apoptosis in vivo without inducing toxicity to liver and kidney. CONCLUSION: The combination of Gem M and Phen M may be potentially used in lung cancer therapy.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Carriers/chemistry , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Phenformin/administration & dosage , Animals , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Delivery Systems , Drug Synergism , Female , Humans , Lung/drug effects , Lung/pathology , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Micelles , Neoplastic Stem Cells/pathology , Phenformin/pharmacology , Phenformin/therapeutic use , Polycarboxylate Cement/chemistry , Polyethylene Glycols/chemistry , Urea/analogs & derivatives , Gemcitabine
15.
PLoS One ; 10(6): e0131207, 2015.
Article in English | MEDLINE | ID: mdl-26114294

ABSTRACT

Breast cancer remains a world-wide challenge, and additional anti-cancer therapies are still urgently needed. Emerging evidence has demonstrated the potent anti-tumor effect of biguanides, among which phenformin was reported to potentially be a more active anti-cancer agent than metformin. However, little attention has been given to the role of phenformin in breast cancer. In this study, we reveal the role of phenformin in cell death of the MCF7, ZR-75-1, MDA-MB-231 and SUM1315 breast cancer cell lines. The respective IC50 values of phenformin in MCF7, ZR-75-1, MDA-MB-231 and SUM1315 cells were 1.184±0.045 mM, 0.665±0.007 mM, 2.347±0.010 mM and 1.885±0.015 mM (mean± standard error). Phenformin induced cell cycle change and apoptosis in breast cancer cells via the AMPK/mTOR/p70s6k and MAPK/ERK pathways. Interestingly, phenformin induced MET (mesenchymal-epithelial transition) and decreased the migration rate in breast cancer cell lines. Furthermore, our results suggest that phenformin inhibits breast cancer cell metastasis after intracardiac injection into nude mice. Taken together, our study further confirms the potential benefit of phenformin in breast cancer treatment and provides novel mechanistic insight into its anti-cancer activity in breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Cycle/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hypoglycemic Agents/pharmacology , Phenformin/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Female , Humans , Hypoglycemic Agents/therapeutic use , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Phenformin/therapeutic use , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Cell Biochem Biophys ; 71(2): 513-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25312480

ABSTRACT

Biguanides, used for anti-diabetic drugs, bring more attention in cancer research for their beneficial effects. Phenformin is more potent than metformin. However its potential application as a anti-cancer regent is far behind metformin. In order to investigate any beneficial effect of combination of Phenformin and radiotherapy, non-small cell lung cancer cell lines A549 and H1299 were exposure under different dose of ionizing radiation with or without Phenformin. Results indicated Phenformin showed synergistic effect and could induce more cancer cell apoptosis and inhibition of tumor growth compared with ionizing radiation alone. Furthermore, this synergistic effect may be through different pathway according to cancer cell genotype background. Our results showed Phenformin induced AMPK activation in A549 but not H1299. However, Phenformin activated eIF2α in both cell lines. Our findings implicated Phenformin may be used as radiosensitizer for non-small cell lung cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Phenformin/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Female , Humans , Lung Neoplasms/radiotherapy , Mice , Mice, Inbred BALB C , Mice, Nude , Phenformin/pharmacology , Radiation-Sensitizing Agents/pharmacology , Transcription Factors/metabolism
18.
Cancer Cell ; 23(2): 143-58, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23352126

ABSTRACT

The LKB1 (also called STK11) tumor suppressor is mutationally inactivated in ∼20% of non-small cell lung cancers (NSCLC). LKB1 is the major upstream kinase activating the energy-sensing kinase AMPK, making LKB1-deficient cells unable to appropriately sense metabolic stress. We tested the therapeutic potential of metabolic drugs in NSCLC and identified phenformin, a mitochondrial inhibitor and analog of the diabetes therapeutic metformin, as selectively inducing apoptosis in LKB1-deficient NSCLC cells. Therapeutic trials in Kras-dependent mouse models of NSCLC revealed that tumors with Kras and Lkb1 mutations, but not those with Kras and p53 mutations, showed selective response to phenformin as a single agent, resulting in prolonged survival. This study suggests phenformin as a cancer metabolism-based therapeutic to selectively target LKB1-deficient tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Hypoglycemic Agents/therapeutic use , Lung Neoplasms/drug therapy , Phenformin/therapeutic use , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/physiology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cells, Cultured , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mutation/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/physiology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
Br J Cancer ; 106(6): 1117-22, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22361631

ABSTRACT

BACKGROUND: Observations that diabetics treated with biguanide drugs have a reduced risk of developing cancer have prompted an enthusiasm for these agents as anti-cancer therapies. We sought to determine the efficacy of the biguanide phenformin in the chemoprophylaxis and in the treatment of oestrogen receptor (ER)-positive MCF7 and receptor triple-negative MDAMB231 xenografts in immunocompromised mice. We also compared the efficacy of phenformin and metformin in the treatment of MDAMB231. METHODS: Immunocompromised mice were divided into groups: (1) phenformin administered for 2 weeks prior to cell injection; (2) established tumours treated with phenformin; (3) established tumours treated with metformin (only for MDAMB231 tumours); (4) untreated controls. Post-treatment tumours, liver and spleen were harvested for further analysis. RESULTS: Phenformin significantly inhibited both the development and growth of MCF7 and MDAMB231 tumours, and for MDAMB231 at greater efficacy than metformin without murine toxicity. The number of mitotic figures was significantly fewer in xenografts treated with phenformin compared with controls. Results suggested that the mechanism of action of phenformin in vivo is consistent with AMPK activation. CONCLUSION: Phenformin has clinical potential as an antineoplastic agent and should be considered for clinical trials both in ER-positive and triple-negative breast cancer.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/prevention & control , Metformin/therapeutic use , Phenformin/therapeutic use , Adenylate Kinase/metabolism , Animals , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Enzyme Activation , Female , Histones/metabolism , Humans , Ki-67 Antigen/metabolism , Liver/enzymology , Metformin/pharmacology , Mice , Mice, Nude , Phenformin/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Spleen/enzymology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Future Oncol ; 6(8): 1313-23, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20799876

ABSTRACT

Comparing the experience accumulated for more than 40 years in the Laboratory of Endocrinology of Petrov Institute of Oncology (St Petersburg, Russia) with similar approaches practiced elsewhere, evidence supports the reasonability of metabolic rehabilitation of patients suffering from breast cancer or other hormone-dependent malignancies. The primary objective of such approaches is to improve treatment results by ameliorating hormonal-metabolic disturbances, including excess body fat, glucose intolerance, insulin resistance and manifestations of endocrine-genotoxic switchings, and modify tissue and cellular targets or mechanisms related or nondirectly related to the aforementioned disturbances. The relevant measures may be categorized as pharmacological (antidiabetic biguanides exemplified with metformin being most popular but not exclusive) and nonpharmacological (rational nutrition, moderate physical activity and so forth) and used separately or in different combinations.


Subject(s)
Diet Therapy , Exercise Therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Neoplasms/rehabilitation , Phenformin/therapeutic use , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...