Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
2-Deoxyglucose
2-Fludeoxyglucose
Three direct-acting antivirals
Adjuvant for breast cancer treatment using seven repurposed drugs
ATP-binding cassette superfamily G member 2
Alcohol dehydrogenase
Adrenoreceptor beta
Aldehyde dehydrogenase
Adenosine monophosphate
AMP-activated protein kinase
Artemisinin
Adenosine triphosphate
Breast cancer–resistance protein
Breast cancer stem cells
Basal-like breast cancer
Breast cancer-1
Cisplatin
Cyclin-dependent kinase-2
Cyclooxygenase-2
Cancer stem cells
Copper gluconate
Cytochrome p
Dichloroacetate
Ductal carcinoma in situ
Drug–drug interaction
Diethyldithiocarbamate
Drug Repurposing from Control System Theory
Dimethylbenz[a]anthracene
Deoxyribonucleic acid
DNA methyltransferase
Doxycycline
Disulfiram
Epidermal growth factor
Enhanced green fluorescent protein
Epidermal growth factor receptor
Epithelial–mesenchymal transition
Enolase-1
Estrogen receptor
Extracellular receptor kinase
Food and Drug Administration
Flunarizine
Glioblastoma
Glucagon like peptide-1
Glutathione S-transferase P1
Hepatitis C virus
Human epidermal growth factor receptor
Human immunodeficiency virus
Hexokinase-2
Hazard ratio
Human telomerase reverse transcriptase
Inflammatory breast cancer
Insulin-like growth factor-1
Insulin-like growth factor binding protein-3
Interleukin 6
Intravenous
c-Jun N-terminal kinase
Light chain-3
Lactate dehydrogenase A
Liposome-encapsulated disulfiram
Lily polysaccharide-1
Mitogen-activated protein kinase
Multidrug and toxin extrusion protein
Metastatic breast cancer
Mebendazole
Methylguanine DNA methyltransferase
Millimolar
Matrix metalloproteinases
Mitochondrial ribonucleic acid
Mesenchymal stem cells
Maximum tolerated dose
Metformin
Mammalian target of rapamycin
N-acetylcysteine
Nuclear factor kappa-light-chain-enhancer of activated B cells
National Institutes of Health
Nanomolar
Nonstructural protein 5B
Organic anion transporting polypeptide
Organic cation transporter
Oxidative stress response
P-glycoprotein
Paclitaxel
Physiologically based pharmacokinetics
Pathological complete response
Phosphoinositide-dependent kinase-1
Polyethylene glycol
Positron emission tomography
Phosphoinositide 3-kinase
Poly-lactic-c-glycolic acid
Polo like kinase-1
Plasma membrane monoamine transporter
Progesterone receptor
Phosphatase and tensin homolog
Quantitative polymerase chain reaction
Receptor activator of nuclear factor kappa-beta ligand
Reversion inducing cysteine rich protein with kazal motifs
Reactive oxygen species
Random periareolar fine-needle aspiration
Silibinin
Superoxide dismutase
The Cancer Genome Atlas
Transforming growth factor beta
T helper type 2
Toll-like receptor
Triple-negative breast cancer
Transformation related protein 53
Tetrathiomolybdate
Terminal deoxynucleotidyl transferase dUTP nick end labeling
Uridine diphosphate glucuronosyltransferase
Ubiquitin proteasome system
Vascular endothelial growth factor
Micromolar
Breast cancer is the most common type of cancer affecting women ( https://www.cancer.gov/types/common-cancers ; https://www.who.int/health-topics/cancer#tab=tab_1 ; https://www.wcrf.org/dietandcancer/cancer-trends/skin-cancer-statistics ). The tremendous impact of breast cancer calls for any and every method possible to be used in its therapy and treatment. Drug repurposing (also known as repositioning, reprofiling, redirecting, or rediscovering [ ]) means developing new uses for a drug beyond its original use or initially approved indication. Drug repurposing has attracted increasing attention in recent years as potentially inexpensive alternatives are needed urgently to compensate for the high costs and disappointing success rate associated with the drug discovery pipeline [ ]. Repurposing can help identify new therapies for diseases at a lower cost and in a shorter time, particularly in those cases where preclinical safety studies have already been completed [ ]. Repurposing an old drug for a new use, such as cancer therapy, is an attractive and exciting field [ , ]. During recent years, several reviews on drug repurposing have been published [ , ].
Drug repurposing is a possible alternative to the current therapies in breast cancer treatment. Multiple drugs have shown great promise in this aspect, from inhibition of breast cancer cell proliferation in general to blocking of pathways that specifically cause breast cancer. This chapter will focus on multiple old or currently used drugs that have shown potential promise in the therapy of breast cancer, by summarizing first their original uses or purposes and then their mechanisms of action responsible for the antibreast cancer effects. This chapter will then comprehensively review both preclinical and clinical (as available) studiesusing these repurposed drugs and their multitude of effects on various subtypes of breast cancer. It will end by discussing the future of drug repurposing—both as a field in general and specifically in the therapy of breast cancer.
Metformin (MTF; Fig. 5.1 ) is a well-acknowledged biguanide and a multiaction drug advertised under the name Glucophage as well as others. MTF was approved by the FDA in 1995 as an oral hypoglycemic drug in the management of diabetes mellitus and is the first-line medication for type 2 diabetes mellitus treatment [ ] ( Table 5.1 ). MTF could effectively reduce gluconeogenesis in the liver and improve insulin sensitivity by inducing peripheral glucose uptake and lowering the basal and postprandial plasma glucose [ ]. It was reported that MTF has antiinflammatory [ ], antiapoptotic, anticancer, hepatoprotective, cardioprotective [ ], renoprotective [ ], otoprotective [ ], radioprotective, radiosensitizing, and antioxidant activities [ ] ( Table 5.1 ). There has been a recent development in MTF repositioning for anticancer therapy. It exhibited a great potential to change the metabolic reprogramming and acts as a candidate for cancer management [ ]. In 2005, MTF was used for breast cancer treatment [ ].
Drug | History and use | Original mechanisms of action | Proposed anticancer mechanisms |
---|---|---|---|
MTF | FDA approved in 1995 as an oral hypoglycemic drug in the management of diabetes mellitus | Inhibition of mitochondrial complex I and ATP production Inhibition of gluconeogenesis and lipogenesis Inhibition of fructose-1,6-bisphosphatase Reduction in CCL11 and proinflammatory cytokines Suppression of monocyte differentiation in macrophages Increased GLP-1 secretion in the gut and increased glucose utilization |
Reduction in the carcinoma cell proliferation via insulin/IGF-1 pathway, inhibition of NF-kB, AMPK activation, metabolic stress generation, inhibition of cell proliferation, DNA replication apoptosis through the BAX/BCL-2 apoptotic pathway and AMPK/mTOR/p70S6 growth pathway, cytotoxicity, and apoptosis via toll-like receptor (TLR) signaling |
DSF | FDA approved 1951 as Antabuse in alcohol deaddiction | Inhibition of alcohol dehydrogenase | Inhibition of proteasome (E3 ubiquitin ligase), generation of ROS, inhibition of SOD-1 activity, activation of MAPK pathway, inhibition of P-gp, inhibition of NF-kB, inactivation of Cu/Zn SOD, inactivation MMP, inhibition of DNA topoisomerase, inhibition of DNMT, inhibition of GSTP1, inhibition of MGMT, upregulation of RECK, reduction of CDK1 expression levels, causing G2 arrest, reduction in expression of PLK1 protein and mRNA |
Propranolol | FDA approved in 1967 for the management of hypertension. It is used for other cardiovascular conditions cardiac arrhythmias, postmyocardial infarction to reduce mortality. Other noncardiovascular uses are for prophylaxis of migraine, essential tremors, anxiety, portal hypertension, hyperthyroidism, and pheochromocytoma | Nonselective beta-adrenergic receptor blockage—blocks the action of catecholamines (adrenaline and noradrenaline) at both beta-1 and beta-2 adrenergic receptors | Reduced expression of hexokinase-2 Agonism at MT1 and MT2 melatonergic receptors |
Antivirals | Ombitasvir, dasabuvir, and paritaprevir were FDA approved in July 2016 for the treatment of hepatitis C Ritonavir was FDA approved in 1999 for the treatment of HIV AIDS |
Ombitasvir—HCV NS5A inhibition Dasabuvir—HCV NS5B inhibition Paritaprevir—HCV NS3/4A inhibition Ritonavir—cytochrome P3A4 inhibition causing protease inhibition |
Inhibition of P-gp, inhibition of breast cancer–resistance protein (BCRP), inhibition of transcriptional and translational factors, suppression of telomerase activity, and activation of human DNA polymerase, increase plasma exposure in drugs, binding to heat shock protein 90 (HSP90), and partial inhibition of its chaperone |
Antipsychotics | Risperidone, 9-hydroxy-risperidone (paliperidone), olanzapine, quetiapine, clozapine, haloperidol, and chlorpromazine | Exact mechanisms are not known, but drugs of this class are believed to inhibit mainly dopaminergic D2 and serotonergic 5HT2A receptors and sometimes alpha-1 adrenergic, H-1 histaminic, and muscarinic receptors | Inhibition of cellular uptake of mitoxantrone, inhibition of RANKL-mediated MAPK and NF-kB signaling pathways, inhibition of breast cancer–resistance protein (BCRP) |
Thalidomide | It was previously used in pregnant women to relieve morning sickness. However, its use was restricted because of its teratogenic effect on the developing fetus. Over the last decades, thalidomide showed a cytotoxic effect on different cancer cell lines, including breast cancer | The CD147 and MCT1 protein complex is stabilized and developed by the binding of the cereblon protein and this effect on the complex stimulates cell growth and facilitates the excretion of metabolic products like lactate. It is because of this that an increased abundance of this protein complex enables tumor cells to spread rapidly in diseases such as multiple myeloma. If such a cancer is treated with IMiDs, the complex is displaced from its binding to cereblon and therefore the protein complex of CD147 and MCT1 can no longer be activated, which in turn causes tumor cells to die. | The growth and progression of breast cancer cells including MCF-7 and MDA-MB-231 were inhibited by a small series of thalidomide-correlated compounds, which are very effective to induce cancer cell death via triggering TNFα-mediated apoptosis |
Artemisinin | Artemisinin (ART) is a chemical that was isolated from the sweet wormwood by Youyou Tu at the Chinese Academy of Traditional Chinese Medicine in 1972 . Originally used for the treatment of malaria, during the past two decades, studies revealed the anticancer activity of artemisinin and its derivatives, which indicates the effectiveness of these compounds as cancer therapeutic drugs | Artemisinin and its semisynthetic derivatives were originally formulated to be used against malaria. These drugs contain endoperoxide bridges, which are needed for antimalarial activity. The mechanism of action for this original use was believed to be a two-step mechanism. First, ART is activated by intraparasitic heme iron which catalyzes the cleavage of the endoperoxide bridges which then causes a free radical intermediate to be formed and kill the parasite by alkylating and poisoning one or more essential malarial proteins | ARS and DHA were reported to inhibit TGF-β signaling that inactivates cancer-associated fibroblasts (CAFs) which play an important role in tumor growth and metastasis, such as stimulating angiogenesis, cell proliferation, migration, and invasion |
Mebendazole | It was approved by the US FDA in 1974 for the treatment of nematode infections. Also, MBZ revealed efficacy against different types of solid tumors in vitro and in vivo, such as lung cancer, melanoma, colon cancer, glioblastoma multiforme, medulloblastoma, and head and neck squamous cell carcinoma | Mebendazole binds to the colchicine-sensitive site of tubulin and therefore causes degenerative alterations in the tegument and intestinal cells of the parasite. The parasite then has its glycogen stores depleted because this loss of cytoplasmic microtubules eventually leads to disruption in the glucose uptake by its larval and adult stages. This causes the parasites immobilization and eventual death | It was reported that MBZ was able to induce cell cycle arrest in the radiosensitive G2/M phase of the cell cycle in TNBC cells. This cell cycle arrest was followed by significant induction of apoptotic cell death in a dose- and time-dependent manner. Different studies revealed induction of apoptosis as the primary mode of cell death by MBZ in other tumor types, such as melanoma, lung cancer, and medulloblastoma. Also, tubulin depolymerization was reported to be the major target for benzimidazoles, including MBZ. MBZ exhibited induction of depolymerization of tubulin and inhibition of normal spindle formation in different cancer cell lines, resulting in mitotic arrest and apoptosis |
Flunarizine | Not FDA approved. Not prescribed in the United StatesIn other countries, include prophylaxis of migraine, peripheral vascular disease, and vertigo | Selective calcium entry block with calmodulin-binding properties and histamine H1–blocking activity | N-Ras degradation, inhibition of colony formation through TG101348 and FLN combination, inhibition of growth and transforming activity of BLBC cells, induction of autophagy-like activity |
MTF is an orally bioavailable drug metabolized in the liver. It is transported into hepatocytes by organic cation transporter-1 (OCT1). It then accumulates in the mitochondrial inner membrane, inhibits complex 1 of the electron transport chain, and reduces ATP production. The increase in AMP and ADP causes AMPK activation. Activated AMPK inhibits fat synthesis and promotes fat oxidation instead, thus reducing hepatic lipid stores and enhancing hepatic insulin sensitivity. The increase in AMP:ATP ratio also inhibits fructose-1,6-bisphosphatase, resulting in the acute inhibition of gluconeogenesis. MTF also acts via other pathways to inhibit gluconeogenesis, glycogenolysis, and release of glucose in the blood ( Table 5.1 ).
In vitro and in vivo data of MTF treatment revealed its ability to inhibit the growth of ovarian cancer stem cells (CSCs) [ ], glioma-initiating cells [ ], breast cancer cells [ ], endometrial cancer cells [ ], and non-small cell lung cancer cells [ ].
Anticancer properties of MTF have been shown either through its direct effect on the cancer cells by activating AMPK/inhibiting mammalian target of rapamycin (mTOR) pathway [ ] or via its indirect effect on the host by decreasing the blood glucose level in addition to its antiinflammatory effects [ , ] ( Table 5.1 ). The inhibition of mTOR in tumor cells was reported as one of the potential vital mechanisms of the anticancer properties of MTF [ ]. AMPK could inhibit mTORC1 via phosphorylation of mTOR-binding raptor [ ]. In addition, HER2 expression in human breast cancer cells treated with MTF was declined via direct inhibition of p70S6K1, which is a downstream effector of mTOR [ ]. Several studies also suggested that MTF causes many biological and molecular effects on breast cancer cells in addition to its ability to reduce cellular response to the tumorigenesis critical factors as insulin, interleukin 6 (IL-6), and epidermal growth factor through downregulation of IGF-1R, p-Stat-3, and EGFR [ ].
In MCF-7 breast cancer cells ( Table 5.2 ), MTF reduced phosphorylation of S6 kinase, ribosomal protein S6, and eIF4E-binding protein, inhibited mTOR, and reduced translation initiation via AMPK activation [ ].
Drug | Cancer cell line | ER | PR | HER2 | BRCA1 mutation | Subtype | Source |
---|---|---|---|---|---|---|---|
Metformin | BT474 | + | + | + | WT | LB | L |
MCF7, T47D | + | + | − | WT | LA | L | |
ZR7530 | + | − | + | WT | LB | L | |
MDAMB453, SKBR3 | − | − | + | WT | H | L | |
MDAMB231, MDAMB157, MDAMB468, HCC70, BT20, BT549 | − | − | − | WT | TNB | L | |
HCC38, HCC1143, HCC1187, HCC1806 | − | − | − | ND | TNB | L | |
HCC1937, MDAMB436 | − | − | − | MU | TNB | L | |
DSF | BT474 | + | + | + | WT | LB | L |
MCF7, T47D | + | + | − | WT | LA | L | |
SKBR3, SUM190 | − | − | + | WT | H | L | |
MDAMB157, MDAMB231, BT549 | − | − | − | WT | TNB | L | |
SUM149 | − | − | − | MU | TNB | L | |
HCC70, BT20 | − | − | − | WT | TNA | L | |
MDAMB436 | − | − | − | MU | TNA | L | |
Propranolol | MCF7 | + | + | − | WT | LA | L |
MDAMB231 | − | − | − | WT | TNB | L | |
Antiviral drugs | MCF7, T47D | + | + | − | WT | LA | L |
MDAMB231 | − | − | − | WT | TNB | L | |
MDAMB436 | − | − | − | MU | TNB | L | |
Antipsychotic drugs | MCF7 | + | + | − | WT | LA | L |
MDAMB231 | − | − | − | WT | TNB | L | |
Antimalarial drugs, Artemisinin (ART) |
MCF7 | + | + | − | WT | LA | L |
MDAMB231 | − | − | − | WT | TNB | L | |
Mebendazole | MCF7, T47D | + | + | − | WT | LA | L |
MDAMB231, SUM159PT | − | − | − | WT | TNB | L | |
Thalidomide | MCF7 | + | + | − | WT | LA | L |
MDAMB231 | − | − | − | WT | TNB | L | |
Flunarizine | SUM102PT | − | − | − | WT | TNB | L |
SUM149PT | − | − | − | MU | TNB | L |
Zordoky et al. reported that excess glucose amount in triple-negative MDA-MB-231 cells ( Table 5.2 ) blocked the MTF-induced cell death, which suggested that high glucose levels resulted in the production of enough energy for cell proliferation via aerobic glycolysis [ ]. In addition, Wahdan-Alaswad et al. reported that glucose increases the aggression of breast cancer cells and reduces the efficacy of MTF. Their preclinical studies on triple-negative breast cancer (TNBC) showed that MTF blocked several key enzymes necessary to glucose metabolism, but its effect was glucose-dependent. Thus, glucose monitoring for patients with breast cancer is suggested to be a crucial factor [ ]. In vivo studies also revealed the antiproliferative effect of MTF in breast cancer [ , ].
In vitro and in vivo preclinical studies of MTF in combination with other drugs also revealed a synergistic effect to reduce cell proliferation in both breast cancer cell culture and animal models [ ].
Curcumin with MTF exhibited dose-dependent cytotoxicity and antiproliferative activity, as shown by a declined hTERT expression, reduced vascular endothelial growth factor (VEGF) expression, induced transformation related protein 53–independent apoptosis, and triggered Th2 immune response [ ]. In mice models, Falah et al. reported that the combination of MTF and curcumin in breast cancer caused angiogenesis inhibition, immune system modulation, and induction of p53-independent apoptosis [ ].
A combination of ursolic acid with MTF at low concentrations significantly inhibited invasion and migration of transforming growth factor-β (TGF-β)–induced breast cancer in MDA-MB-231 and MCF-7 cells. Combination of ursolic acid and MTF also downregulated expression of CXCR4, uPA, vimentin, E-cadherin, N-cadherin, and MMP-2/9 proteins in breast cancer cells [ ].
Vitamin D3 in combination with MTF increased levels of cleaved caspase-3, BAX, and AMPK and inhibited that of BCL-2, c-Myc, IGF-IR, mTOR, P70S6K, and S6, causing cell cycle arrest in breast cancer cells [ ].
Silibinin in combination with MTF synergistically downregulated expression levels of hTERT and cyclin D1 and enhanced inhibition of breast cancer cell growth [ ].
Chrysin with MTF synergistically caused breast cancer cell death by reducing cyclin D1 and hTERT gene [ ]. Flavone with MTF decreased MDMX protein expression, regulated p53 downstream target gene Bcl-2, and cleaved caspase-3 [ ].
Melatonin with MTF reduced tumor growth and size by increasing the expression of Bax and caspase-3 and inhibiting DMBA-induced breast cancer tumor growth [ ].
Lily polysaccharide-1 with MTF downregulated Bcl-2 expression and upregulated Bax, causing enhanced antiproliferation and apoptosis in breast cancer cells [ ].
Recent studies revealed the ability of MTF to enhance the efficacy of chemotherapeutics in combination therapy [ ]. It was reported that efficacies of several chemotherapeutics increased dramatically when given in combination with MTF [ ].
A combination of MTF with doxorubicin inhibited NF-κB activity in breast tumor cells, which further decreased TNF-α and IL-6 expressions in breast tumor cells, suppressed tumor cell proliferation, and enhanced apoptosis. Moreover, the therapy enhanced nuclear doxorubicin accumulation and overcame drug resistance by downregulating P-gp (P-glycoprotein) and intracellular ATP content in a reduced dose [ , ]. Combination therapy of doxorubicin + 2-deoxyglucose (2-DG) with MTF showed substantial metabolic stress at low dose via inhibition of glucose uptake and suppression of lactate, fatty acid, and ATP production. This combination therapy was associated with decreased cell viability, increased the intracellular oxidation, induced apoptosis and autophagy, and completely inhibited colony formation by activation of AMPK [ ]. Another combination experiment of MTF with doxorubicin and 2FDG increased phospho-AMPK but decreased phospho-Akt and phospho-ERK expressions [ ].
Sahra et al. reported that the combination of MTF with 2-DG revealed inhibition of mitochondrial respiration and glycolysis via p53-dependent apoptosis through AMP pathway. Furthermore, MTF showed a synergistic effect when combined with paclitaxel (PAC), carboplatin, or doxorubicin in xenograft mouse models of breast, lung, and prostate cancer [ ].
A combination of MTF with PAC exhibited significant inhibition of cell viability and induction of the G 2 /M phase arrest [ ]. In vitro studies revealed that MTF and PAC had a synergistic effect, and codelivery of the micelles induced higher cytotoxicity and apoptosis in 4T1 breast cancer cells than each free drug [ ].
Tamoxifen with MTF synergistically inhibited cell proliferation and DNA replication and triggered apoptosis at reduced doses, through regulating both Bax/Bcl-2 and AMPK/mTOR/p70S6 pathways [ ].
MTF in combination with 5-fluorouracil, epirubicin, and cyclophosphamide accelerated glucose consumption and lactate production in breast CSCs and significantly hampered intracellular ATP leading to a severe energy crisis and thus inducing DNA damage in breast cancer cells [ ].
Combination of everolimus with MTF inhibited cell survival, clonogenicity, mTOR signaling activity, mitochondrial respiration, and abrogated S6 and 4E-BP1 phosphorylation in breast cancer cells [ , ]. In addition, the combination of MTF with erlotinib in breast cancer cells enhanced the reduction of EGFR, AKT, S6, and 4EBP1 phosphorylation, associated with inhibition of mammosphere outgrowth [ ].
Aspirin and MTF combination enhanced breast cancer 4T1 cell apoptosis by inducing secretion of TGF-β1. The effect of this combination partly relied on cyclooxygenase-2 upregulation, without production of lipoxins [ ]. In both immune-deficient and immune-competent breast cancer preclinical models, atenolol increased MTF activity against angiogenesis, local and metastatic growth of HER2+, and triple-negative BC. Aspirin increased the activity of MTF only in immune-competent HER2+ BC models. Both aspirin and atenolol, when added to MTF, significantly reduced the endothelial cell component of tumor vessels, whereas pericytes were reduced by the addition of atenolol but not aspirin. These data indicate that the combination of aspirin or atenolol with MTF might be beneficial for BC treatment and that this anti-BC activity is likely due to the inhibitory effects on both BC and microenvironment cells [ ].
A combination of propranolol and MTF inhibited glucose metabolism via downregulation of ADRB2-dependent hexokinase-2, and posttranscription and activation of AMPK, along with the antioxidation activity [ ].
Dichloroacetate and MTF combination revealed synergistic induction of caspase-dependent apoptosis via oxidative damage through PDK1 inhibition and decreased lactate production in breast cancer cells. Expression of glycolytic enzymes, including HK2, LDHA, and ENO1, were downregulated, associated with induction of cell death [ , ].
Topotecan with MTF was shown to activate AMPK, downregulate excision repair cross-complementation group 1, and suppress DNA replication by inhibiting nuclear enzyme topoisomerase I. This combination also depolarized mitochondrial membrane and induced cell cycle arrest in breast cancer cells [ ]. MTF was also reported to prevent BRCA1 haploinsufficiency-driven RANKL gene overexpression and disrupt the autoregulatory feedback control of RANKL-addicted CSCs. The synergistic effect of MTF and denosumab decreased breast cancer–initiating cell (BCIC) population and self-renewal capacity [ ].
There are a total 45 registered clinical trials at different stages of development that have studied the efficacy of MTF as monotherapy or in combination with chemotherapy and/or radiotherapy exclusively for the management of breast cancer ( Table 5.3 ). The studies also focus on the process of establishing the effects of MTF on markers of cellular proliferation, pathological response rate, progression-free survival, tolerated safe dose, and recurrence-free survival for breast cancer.
Clinical trial | Condition | N1 | N2 | Design | Primary outcome | Status | |
---|---|---|---|---|---|---|---|
1 | NCT00897884 | Breast cancer | 40 | 39 | Metformin 500 mg tablet, TID X 2–3 weeks prior to surgery | Reduction in cell proliferation rates in tumor tissue | Completed |
2 | NCT01266486 | Breast cancer | 40 | 41 | Extended release metformin 1500 mg OD X 14–21 days | Measure metformin induced effects in phosphorylation of S6K, 4e-BP-1 and AMPK | Completed |
3 | NCT02882581 | Breast cancer | 10 | 7 | 400MBq of 11C-metformin is injected in the cubital vein, followed by PET scan | Metformin uptake in breast cancer | Completed |
4 | NCT01310231 | Metastatic breast cancer | 78 | 40 | Metformin 850 mg BID + standard chemotherapy (anthracyclines, platinum, taxanes, or capecitabine; first or second line) Placebo + standard chemotherapy |
Progression-free survival | Completed |
5 | NCT01650506 | Breast cancer | 20 | 8 | Standard 3 + 3 dose escalation Metformin 850 mg BID to TID Erlotinib 150 mg daily |
Maximum tolerated dose of metformin in combination with a fixed dose of 150 mg erlotinib daily | Completed |
6 | NCT01340300 | Colorectal cancer Breast cancer |
200 | 139 | 1. Exercise training 2. Exercise training + oral metformin QD X 2 weeks, then BID 3. Oral metformin QD X 2 weeks, then BID Control—educational information |
Change in fasting insulin level | Completed |
7 | NCT01589367 | Hormone receptor–positive malignant neoplasm of breast | 208 | 208 | Arm 1: Metformin experimental Arm 2: Letrozole alone/placebo |
Clinical response rate comparing with RECIST 1.1 from baseline | Completed |
8 | NCT01793948 | Breast cancer Obesity |
24 | 24 | Experimental: Arm I: Metformin hydrochloride PO OD on days 1–30 in course 1 and BID on days 1–30 thereafter X 12 courses Arm II: Placebo |
Changes in the phosphorylation of proteins after metformin exposure | Completed |
9 | NCT01885013 | Human epidermal growth factor 2 negative carcinoma of breast | 112 | 126 | Arm A: Metformin (day 1–day 3, 1000 mg OD; day 4 to day 13, 1000 mg BID) + myocet 60 mg/m 2 , IV on day 1/21 days + cyclophosphamide 600 mg/m 2 IV on day 1/21 days Arm B: Myocet + cyclophosphamide |
Progression-free survival (PFS) | Completed |
10 | NCT00909506 | Breast cancer | 105 | 105 | Placebo comparator: Placebo Active comparator: Metformin 500 mg/day OD X 1–2 weeks Active comparator: Metformin 1000 mg/day (dose-eascalate) |
Weight loss | Completed |
11 | NCT00933309 | Breast cancer | 24 | 25 | Group 1: Exemestane 25 mg PO OD Group 1: Exemestane 25 mg PO OD + avandamet (rosiglitazone 2 mg + metformin 500 mg PO OD) |
Dose-limiting toxicity (DLT) | Completed |
12 | NCT00659568 | Breast cancer Endometrial cancer Kidney cancer Lung cancer Lymphoma Unspecified adult solid tumor, protocol specific |
28 | 28 | Metformin hydrochloride OD/BID/TID on d1–d28 + temsirolimus IV d1, 8, 15, 22/28 days | Maximum tolerated dose and recommended Phase II dose of metformin hydrochloride when administered with temsirolimus | Completed |
13 | NCT02145559 | Breast neoplasms Lung neoplasms Cancer of liver Lymphoma Cancer of kidney |
64 | 24 | Experimental: Sirolimus (3 mg daily) d1–7 + metformin XR (500 mg PO OD d8–d28; from d15 1000 mg PO OD) Experimental: Sirolimus (3 mg daily) d1–7, + delayed metformin XR (500 mg PO OD from d22; after 1 week 1000 mg PO OD) |
Pharmacodynamic biomarker p70S6K | Completed |
14 | NCT02431676 | Breast cancer Prostate cancer Lung cancer Colon cancer Melanoma of skin Endometrial cancer Liver cancer Pancreatic cancer Rectal cancer Kidney cancer Other solid malignant tumors |
120 | 121 | Active comparator: Self-directed behavioral self-control weight loss Experimental: Coach-directed behavioral weight loss Experimental: Metformin up to 2000 mg daily X 12 months |
IGF-1 levels IGF-1 levels: IGFBP3 levels (ratio) |
Completed |
15 | NCT02028221 | Breast cancer | 150 | 151 | Metformin 850 mg PO OD X 4 weeks followed by 850 mg PO BID. Placebo |
Change from baseline in breast density at 6 and 12 months | Active, not recruiting |
16 | NCT02488564 | HER2-positive breast cancer | 46 | 49 | Day 1: Liposome-encapsulated doxorubicin, 50 mg/m 2 IV 1 h Day 2 and 9: Docetaxel, 30 mg/m 2 IV 1 h Day 2, 9, and 16: Trastuzumab 4 mg/kg loading dose, 2 mg/kg/week for subsequent injections Day 13–11: Metformin 1000 mg OD; Day 10–0: Metformin 1000 mg BID until end of the study treatment. |
Pathologic complete response rate (pCR) | Active, not recruiting |
17 | NCT01101438 | Breast cancer | 3582 | 3649 | Experimental Arm I: Oral metformin HCL BID (OD in weeks 1–4) continue up to 5 years in ER+, PR+ Placebo Arm II: Placebo BID (OD in weeks 1–4) continue up to 5 years in ER+, PR+ |
Invasive disease-free survival in hormone receptor negative and positive subgroups | Active, not recruiting |
18 | NCT02278965 | Stage 0—III breast carcinoma Breast neoplasms |
20 | 19 | Metformin 850 mg, oral, twice a day for 12 months Omega-3 fatty acids 2 capsules (560 mg each) oral, twice a day for 12 months |
Assess the safety and feasibility of a 1-year intervention of metformin and omega-3 fatty acids in early stage breast cancer patients who completed adjuvant treatment | Active, not recruiting |
19 | NCT04143282 | Metastatic breast cancer (nondiabetic) | 50 | 50 | Metformin 850 mg to 1 gm, BID | Disease progression through tumor size assessed by CT scan (chest–abdomen–pelvis), bone scan, and MRI | Recruiting |
20 | NCT04170465 | Breast cancer female | 60 | 60 | 1. Oral metformin HCl 850 mg BID X 6 months + AC-T chemotherapy regimen ([doxorubicin 60 mg/m 2 IV + cyclophosphamide 600 mg/m 2 IV[ X 4 cycles, every 3 weeks + [paclitaxel 80 mg/m 2 IV] every week X 12 weeks) 2. AC-T chemotherapy regimen alone |
Evaluation of the effect on tumor proliferation as measured by Ki-67 immunohistochemical (IHC) assessment (%) Tissue level of Ki-67 expression in the excised tumor Evaluation of the effect on tumor apoptosis as measured by caspase-3 Chemotherapy toxicities |
Recruiting |
21 | NCT03238495 | HER2-positive breast cancer | 100 | 100 | 1. Active comparator: Chemotherapy only—taxotere, carboplatin, herceptin + pertuzumab (TCH + P) 1. Experimental: Metformin 850 mg OD during the first cycle, then 850 mg twice daily for the remaining 5 cycles + (TCH + P) |
Pathologic complete response | Recruiting |
22 | NCT01980823 | Breast cancer Breast tumors |
40 | 40 | Metformin 1500 mg per day: Divided 500 mg in the morning and 1000 mg in the evening + atorvastatin 80 mg OD | Change in tissue levels of the proliferation marker Ki-67 | Recruiting |
23 | NCT02506777 | Breast cancer | 96 | 96 | 1. Experimental: Metformin 850 mg BID + FDC (fluoruracil 500 mg/m 2 , doxorubicin 50 mg/m 2 , cyclophosphamide 500 mg/m 2 ) once every 21 days 2. Experimental: melatonin 3 mg before sleeping daily + FDC 3. Active comparator: FDC |
Response rate evaluated by RECIST criteria Pathomorphological response assessed after surgery by miller and payne sscale |
Recruiting |
24 | NCT02506790 | Breast cancer | 96 | 96 | Experimental: Toremifene 60 mg daily + metformin 850 mg BID Experimental: Toremifene 60 mg daily with melatonin 3 mg before sleep daily Active comparator: Toremifene |
Response rate evaluated by RECIST criteria Pathomorphological response assessed after surgery by miller and payne scale |
Recruiting |
25 | NCT01929811 | Breast cancer | 200 | Experimental: Metformin 500 mg PO TID (500 mg PO OD for 1st cycle) + docetaxel + epirubicin + cyclophsophomide TEC: Docetaxel 75mg/m2 IV d1 q3w∗6 + epirubicin 75mg/m2 IV d1 q3w∗6 + cyclophsophomide 500mg/m2 IV d1 q3w∗6 |
Pathologic complete response rate (pCR) | Recruiting | |
26 | NCT01905046 | Atypical ductal breast hyperplasia BRCA1, BRCA2 DCIS, LCIS |
400 | 128 | Arm I: Metformin hydrochloride PO (OD/BID X 24months; 850 mg BID 13–24months) Arm II: Placebo OD/BID X 12 months; patients may cross over to Arm I for months 13–24 |
Cytological atypia in unilateral or bilateral RPFNA aspirates | Recruiting |
27 | NCT03006172 | Breast cancer Solid tumor |
156 | 104 | Experimental Stage I Arm A: GDC-0077 6 mg (escalating doses) d1, OD from d8/28 days cycle (35 days cycle 1) Experimental Stage I Arm B: GDC-0077 3 mg (escalating doses) OD, d1–d28 + palbociclib d1–d21 + letrozole d1–d28/28 days cycle Experimental Stage I Arm C: GDC-0077 (escalating doses) OD, d1–d28 + letrozole d1–d28/28 days cycle Experimental Stage II Arm B: GDC-0077 OD, d1-–28 + palbociclib d1–d21 + letrozole d1–d28/28 days cycle Experimental Stage II Arm C: GDC-0077 OD, d1–d28 + letrozole d1–d28/28days cycle Experimental Stage II Arm D: GDC-0077 d1–d28 + fulvestrant d1, d15/cycle 1, d1 from cycle 2/28 days Experimental Stage II Arm E: GDC-0077, d1–d28 + palbociclib d1–d21 + fulvestrant d1, d15/cycle 1, d1 from cycle 2/28 days Experimental Stage II Arm F: GDC-0077, d1–d28 + palbociclib d1–d21 + fulvestrant d1, d15/Cycle 1, d1 from cycle 2+ metformin d1–d28/28 days |
Dose-limiting toxicities Recommended Phase II dose of GDC-0077 Adverse events and serious adverse events |
Recruiting |
28 | NCT03168880 | Triple-negative breast cancer | A: Paclitaxel 100 mg/m2/week X 8 weeks + AC/EC (60/600 or 90/600)/3 weekly | Disease-free survival (DFS) Overall survival (OS) |
Recruiting | ||
29 | NCT04001725 | Brain metastases Melanoma Lung cancer Breast cancer |
110 | A: Dexamethasone 8 mg(minimum) PO/IM/IV OD/BID B: Dexamethasone + metformin (850 mg/day, progressively increased to 1700 mg/day on d4 and 2550 mg/day on d7, if well tolerated) |
Prevention of precocious (14 days) dexamethasone-induced diabetes | Recruiting | |
30 | NCT02874430 | Breast carcinoma Endometrial clear cell adenocarcinoma Endometrial serous adenocarcinoma Uterine corpus cancer Uterine corpus carcinosarcoma |
74 | 46 | Metformin hydrochloride PO OD d1–3, BID from d4 + doxycycline PO BID/7 days | Change in the percent of stromal cells expressing caveolin-1 (CAV1) at an intensity of 1+ or greater assessed by immunohistochemistry analyzed using the wilcoxon signed-rank test | Recruiting |
31 | NCT01042379 | Breast neoplasms Breast cancer Breast tumors |
800 | 1920 | 1. Active comparator: Paclitaxel, herceptin followed by doxorubicin, cyclophosphomide 2. AMG 386 with or without trastuzumab 3. AMG 479 (ganitumab) plus metformin 4. MK-2206 with or without trastuzumab 5. T-DM1 and pertuzumab 6. Pertuzumab and trastuzumab 7. Ganetespib 8. ABT-888 9. Neratinib 10. PLX3397 11. Pembrolizumab—4 cycles 12. Talazoparib plus irinotecan 13. Patritumab and trastuzumab 14. Pembrolizumab—8 cycles 15. SGN-LIV1A 16. Durvalumab plus olaparib 17. SD-101 + pembrolizumab 18. Tucatinib |
Pathologic complete response (pCR) for experimental + standard neoadjuvant chemotherapy versus standard neoadjuvant chemotherapy | Recruiting |
32 | NCT02695121 | Breast cancer Bladder cancer |
NA | Dapagliflozin, insulin, metformin, sulfonylureas | Incidence of breast cancer and bladder cancer | Recruiting | |
33 | NCT02201381 | Cancer | 2000 | 207 | Experimental: Metabolic treatment Atorvastatin PO up to 80 mg OD + metformin up to 1000 mg PO OD, increased to BID after 2 weeks + Doxycycline 100 mg PO OD +mg PO OD. |
Overall survival (OS) | Recruiting |
34 | NCT01302002 | Breast cancer | 30 | 0 | Metformin 500 mg tablet, BID X 3 weeks | Determine the in situ effects of metformin on proliferation (Ki67) and apoptosis (TUNEL), phosphorylate AKT, CD1a CD83, CD68, F40/80, arginase iNOS and T cells CD4(+), CD45RA(+), CD 45RO, CD4, CD8, and FOXP3(+) | Withdrawn |
35 | NCT00984490 | Breast cancer | 30 | 5 | Metformin 850 mg PO BID X 7–21 days | Change in Ki67 levels before and after treatment | Terminated (poor accrual) |
36 | NCT01627067 | Breast cancer | 40 | 23 | Exemestane 25 mg OD + everolimus 10 mg OD + metformin 500 mg OD every 3 days—increase to 1000 mg BID if no toxicity | Progression-free survival (PFS) | Terminated (komen foundation funding terminated) |
37 | NCT01477060 | Metastatic breast cancer | 168 | 32 | ARM A: Hormonal therapy + lapatinib (1250mg/die) ARM B: Hormonal therapy + metformin (1500 mg/die) ARM C: Hormonal therapy + lapatinib + metformin |
Rate of patients free from disease progression | Terminated (poor accrual) |
38 | NCT02472353 | Breast cancer Breast tumors |
44 | 30 | 1. Active comparator—doxorubicin Experimental: Metformin + doxorubicin |
Decrease in the incidence of change in left ventricle ejection fraction (LVEF) | Terminated (poor accrual) |
39 | NCT02360059 | Breast cancer | 42 | 1 | 12 days prior to start of paclitaxel 1. Experimental arm: Metformin 500 mg OD X 5 days, followed by 500 mg BID X 5 days, followed by 1000 mg BID X 2 days Questionnaires; sensory and fine -motor tests 2. Placebo |
Mean change in neuropathy | Terminated (poor accrual) |
40 | NCT00930579 | Breast cancer | 15 | 35 | Metformin 1500 mg: divided 500 mg in the morning and 1000 mg in the evening, for at least 2 weeks prior to surgery | Effects of metformin on AMPK/mTOR signaling pathway | Unknown |
41 | NCT03192293 | Breast cancer | 28 | 28 | Experimental: 7 days lead-in period: Metformin 850 mg OD + simvastatin 20 mg OD If well-tolerated, fulvestrant at standard doses: Cycle 1: 500 mg at day 1 and day 15/28 days cycle Cycle 2 and beyond: 500 mg at day 1/28 days cycle |
Clinical benefit rate (CBR) | Unknown |
42 | NCT01666171 | Breast cancer | 458 | Metformin hydrochloride clinical observation, diagnostic laboratory biomarker analysis, imaging biomarker analysis, medical chart review; procedure: Digital mammography | Change in percent mammographic breast density in contralateral (unaffected) breast from baseline to 1 year using two-sample t-test or wilcoxon rank-sum test | Unknown | |
43 | NCT01566799 | Locally advanced malignant neoplasm | 60 | Paclitaxel X 12 weeks followed by 4 cycles of FAC combined with metformin 500 mg PO OD X 24 weeks | Pathologic complete response (pCR) | Unknown | |
44 | NCT01286233 | Breast cancer Depression Fatigue Sleep disorders |
454 | 394 | Group 1: Metformin 850 mg PO BID X 5 years Group 2: Placebo PO BID X 5 years |
Questionnaire scores about fatigue, stress, sleep, depression, general quality of life, and behavioral risks; biological correlates of fatigue DNA polymorphisms Changes in RNA gene expression |
Unknown |
45 | NCT01302379 | Breast neoplasms | 340 | 333 | 1. Metformin: (week 1 - 500 mg PO OD, week 2–4–1000 mg PO OD, weeks 5+–500 mg PO daily) + lifestyle intervention 2. Placebo + lifestyle intervention 3. Metformin + standard dietary guidelines 4. Placebo + standard dietary guidelines |
Biological markers associated with breast cancer survival | Unknown |
46 | NCT03323346 | Breast neoplasm Metastatic breast cancer |
150 | 150 | Disulfiram 400 mg PO daily + copper (2 mg elemental cu) PO daily | Clinical response rate (RR) Clinical benefit rate (CBR) |
Recruiting |
47 | NCT01847001 | Locally advanced malignant neoplasm Breast cancer |
20 | 10 | Propranolol (20 mg BID up to 40 mg BID) + neoadjuvant chemotherapy (regimen I: paclitaxel; Regimen II: doxorubicin and cyclophosphamide; if tumor is HER2 positive add trastuzumab and pertuzumab) | Percentage of patients compliant with taking > 80% take the drug while on chemotherapy | Active, not recruiting |
48 | NCT02596867 | Breast cancer | 30 | 2 | Propranolol 1.5 mg/kg/day, BID X 3 weeks | Reduction the tumor proliferative index using Ki-67 | Terminated (poor accrual) |
49 | NCT00502684 | Primary operable breast cancer | 460 | 32 | Experimental: propranolol 10 mg QID, starting on d-3 pre-op, X 6 days, till POD 2 + etodolac 400 mg BID, starting on d-3 pre-op, for 6 days, till POD 2 Placebo |
Number and cytotoxic activity of NK cells, levels of NKT cells, lymphocytes, monocytes, and granulocytes; cytokine levels; in vitro cytokine secretion; levels of cortisol and VEGF. Cancer recurrence in 5 years | Unknown |
50 | NCT02013492 | Male breast cancer, recurrent melanoma, Stage IV breast cancer, Stage IV melanoma, Stage IV ovarian epithelial cancer, Stage IV ovarian Germ cell tumor, unspecified adult solid tumor, protocol specific hepatocellular carcinoma | 35 | 35 | Propranolol PO BID X 4 months | Incidence of toxicity graded according to common criteria for adverse events (CTCAEs) V. 4.0 Change in vascular endothelial growth factor (VEGF) Effect of beta-adrenergic blockade on the tumor microenvironment and on the host immune system |
Recruiting |
51 | NCT02649101 | Metastatic breast cancer | 60 | 60 | Thalidomide tablet 100 mg QN, PO | Progression-free survival (PFS) | Unknown |
52 | NCT00193102 | Breast cancer | 40 | 40 | Thalidomide | Overall response rate Time to disease progression |
Terminated |
53 | NCT00049296 | Cancer | 26 | 26 | Thalidomide PO, BID Docetaxel IV over 30 min once weekly |
Determine maximum tolerated dose of docetaxel when administered with thalidomide | Completed |
54 | NCT00764036 | Metastatic breast cancer Locally advanced breast cancer |
18 | 23 | Artesunate PO QD add-on therapy of 100, 150 or 200 mg | Dose-limiting adverse events with possible, probable, or definite relation with the respective dose level of the add-on therapy |
In vitro studies showed that MTF at 5–10 μM was able to activate AMPK [ ]. However, clinically such low concentrations of MTF were not sufficient to cause AMPK activation, although metabolic alteration could be evoked [ ]. Therefore, scientists have focused on using pharmacological combinations that can aid in producing anticancer effects at an effective and safe dose profile of MTF [ ].
Other pharmacological agents may be used together with MTF for enhancing the therapeutic efficacy of the drug. Still, the effective and safe dose for breast cancer is a great challenge. A Phase I study was conducted using radioactive 400MBq 11C-MTF, injected in cubital veins of 10 males and females age >50 years with breast cancer, and PET images were taken after 120 min. Quantitative polymerase chain reaction confirmed the drug transportation pathway through OCT, specifically OCT1-3, MATE 1 and 2, and PMAT. PET imaging also confirmed the drug uptake by breast tissue [ ]. In postmenopausal women with breast cancer, MTF therapy changed the levels of protein phosphorylation. In obese patients with the ordinal level of breast density, MTF decreased the risk of breast cancer [ ].
MTF therapy prior to surgery was shown to reduce cancer cell proliferation and inhibit tumor growth in early stage breast cancer patients [ ]. MTF was also shown to improve general body condition including weight loss in overweight or prediabetes mellitus–operable breast cancer patients [ ]. In Stage I to III colorectal or breast cancer patients, MTF was able to decrease fasting insulin levels and change other insulin-related biomarkers like C-peptide, IGFR, IGFBP-3, adiponectin, and lectin [ ].
In another trial, MTF was found to be able to inhibit cell proliferation and tumor growth in patients with early stage breast cancer before surgery. MTF before surgery in operable breast cancer patients improved general body condition. In a subset of breast cancer patients with tumor size >3 cm, 1500 mg extended-release MTF was shown to cause an alteration of S6K, 4E-BP-1, and AMPK via histochemical analysis, thus effectively altering breast cancer metabolism [ ].
Phase I trial of MTF with exemestane and rosiglitazone was well tolerated in obese postmenopausal ER+ and PR+ breast cancer patients [ ]. Another Phase I trial was conducted using MTF and temsirolimus to determine the maximum tolerated dose (MTD) for the combination in breast cancer patients [ ]. A combination study of MTF with standard first-, second-, third- or fourth-line chemotherapy like anthracycline, taxane, platinum, capecitabine, or vinorelbine-based regimens showed improved survival and tumor response in women with metastatic breast cancer [ ]. Another combination therapy of MTF and erlotinib showed good tolerance in TNBC patients [ ]. MTF acted as a biomarker for tracing the mechanism of cancer cell resistance. MTF, along with myocet and cyclophosphamide in HER2-negative breast cancer patients, led to an increase in the progression-free survival as well as characterization of sensitivity in insulin levels [ ].
In invasive breast cancer patients, when MTF was used before PAC therapy, there was a reduction in the occurrence of some adverse effects, specifically peripheral neuropathy [ ]. In another subset of breast cancer patients, combination therapy of MTF and doxorubicin reduced cardiotoxicity by decreasing the tendency of significant change in left ventricle ejection fraction [ ].
An ongoing trial of MTF in postmenopausal obese women is studying the effect of MTF in the obesity-related breast cancer and breast density along with serum IGF-1 to IGFBP-3 ratio [ ]. Another trial of MTF in women who have had a prior breast biopsy demonstrating atypical hyperplasia is studying the efficacy of MTF for prevention and alteration of RPFNA or blood biomarkers of atypical hyperplasia in unilateral or bilateral RPFNA aspirates of breast cancer [ ].
The combined effect of MTF with 5-fluorouracil, doxorubicin, and cyclophosphamide is being studied in women with locally advanced breast cancer [ ]. The pathomorphological and toxicological effect of MTF and toremifene is being studied in women above the age of 18 [ ]. A presurgical trial of combination therapy of MTF and atorvastatin in women with operable DCIS breast cancer is studying the benefits of the combination as well as a change in the proliferation marker Ki-67 levels in the breast tissue of these patients [ ]. In patients with ER+ breast cancer, the efficacy of neoadjuvant therapy of a combination of MTF and letrozole QD is currently being studied in a clinical trial. This trial also investigates the response rate of breast-conserving surgery and changes in Ki67 level after treatment with the combination [ ].
An ongoing clinical trial with repurposed MTF along with taxotere, carboplatin, herceptin, and pertuzumab in patients with HER2+ breast cancer with cT1c-cT4a-d node without metastasis is studying the effect of the combination as adjuvant therapy [ ].
An ongoing trial of MTF, doxorubicin, docetaxel, and trastuzumab focuses on the efficacy of the combination for operable and locally advanced HER2+ breast cancer. This trial evaluates pCR and systemic tolerance, with precise consideration to cardiac toxicity [ ]. The clinical benefit rate and toxicity profile of combination therapy of MTF and simvastatin are currently being studied in a trial for ER+ breast cancer patients [ ]. The pCR after combination therapy of MTF with docetaxel, epirubicin, and cyclophosphamide is also being studied in breast cancer patients with a life expectancy of >12 months [ ].
An ongoing trial of a combination of MTF and doxycycline (DOX) is being studied in women with localized breast or uterine cancer. The main purpose of the study is to analyze the change in stromal cells expressing caveolin-1 at an intensity of 1+ by immunohistochemistry [ ].
MTF has been off-patent protection since 2002 and can be easily synthesized with dimethylamine hydrochloride and 2-cyanoguanidine. It is known to have well-tolerated side effects making its compliance better than other anticancer medications. By reducing insulin, MTF reduces the anabolic capability of the body, which in turn reduces tumor growth. In the tumor cells, MTF alters the high energy metabolism, reducing their ability to survive and proliferate. The hydrophilic characteristics of the drug facilitate its direct transport through the cell membrane and its action via OCTs, plasma monoamine transporter, and multidrug and toxin extrusion protein.
Disulfiram (DSF, tetraethylthiuram disulfide, or Antabuse; Fig. 5.1 ) is one of the most popular repurposed drugs. In the beginning, it was used in the rubber industry as an industrial catalyst [ ], then it was introduced into medicine as scabicide and vermicide [ ]. It was accidentally found to be a bad combination with alcohol when it was used in a study to treat stomach ailments. The US FDA-approved DSF in 1951 as Antabuse in alcohol deaddiction ( Table 5.1 ). For about 50 years, DSF was used as an antialcoholism drug and now is being repurposed in cancer treatment [ , ] ( Table 5.1 ). DSF repurposition as a potential anticancer drug is based on its unique properties in the human body, e.g., low cost, fewer side effects, high selectivity against different cancers, and synergistic activity with other drugs [ ]. Moreover, DSF showed proteasome inhibition and suppressed different cancer-associated pathways [ , , ].
DSF is the best-known aldehyde dehydrogenase (ALDH) irreversible inhibitor [ ]. Inhibition of ALDH by DSF is through the formation of an intramolecular disulfide bond by two mechanisms: (i) between DSF and an active site thiol in the enzyme or (ii) between the active site thiol and thiol of another cysteine residue via unstable mixed disulfide adduct [ ]. When alcohol enters the body, it is converted into acetaldehyde by alcohol dehydrogenase and then rapidly metabolized into acetic acid by the action of ALDH in the liver. DSF was reported to block the action of ALDH that causes an increased serum acetaldehyde concentration, which is toxic and results in an unpleasant DSF-ethanol reaction to the individual ( Table 5.1 ).
Since the 1970s, scientists have noticed the tumor-suppressive effect of DSF [ ]. Many anticancer properties of DSF have been demonstrated in different preclinical models of breast, prostate, myeloma, leukemia, lung cancer, cervical adenocarcinoma, melanoma, neuroblastoma, and colorectal cancer [ ] ( Table 5.1 ).
DSF, as a thiuram disulfide of dithiocarbamate, could form complexes with metals. The interaction of DSF with copper (II) chloride in solution reveals a rapid formation of the bis (N, N-diethyldithiocarbamate) copper (II) complex in situ [ ]. The anticancer activity of DSF is copper-dependent [ ]. Up to 10 μM, DSF alone did not exhibit cytotoxic effect in cancer cells [ ], while DSF/Cu at lower concentrations exerted anticancer activity [ ] and suppressed cell proliferation, tumor growth, invasion, and migration [ , ], in addition to eliminating tumor-initiating cells and inhibiting colony formation and tumor formation in different animal models [ ]. The antiangiogenic effect was also seen following DSF/Cu treatment via reducing microvessel density and VEGF expression [ ]. DSF with exogenous copper had better antitumor effect when compared to DSF alone in a breast cancer xenograft model [ ].
CSCs are a subpopulation of cells that have the ability of self-renewal and differentiation. Levels of CSCs correlate with tumorigenesis, metastasis, radio-/chemoresistance, and cancer recurrence clinically [ , ]. DSF exhibited a highly cytotoxic effect on breast cancer stem cells (BCSCs), which are considered a significant cause of chemoresistance that leads to the failure of breast cancer chemotherapy.
DSF–Cu complex exhibited proteasome inhibition and induction of apoptosis in breast cancer cell cultures. It suppressed the growth of breast cancer xenografts, which is independent of the status of PIK3CA, the gene encoding α form of class IA PI3Ks, that heightened and overexpressed in a variety of human cancers, including breast cancer. Treatment of a DSF–Cu mixture decreased the expression of PTEN protein in a time- and dose-dependent manner and activated AKT in cell lines regardless of the presence of PIK3CA mutations, which are thought to lead to the activation of the AKT signaling pathway. Therefore, DSF appears to trigger two conflicting signaling pathways in breast cancer cells. First, DSF–Cu inhibits the proteasome, thus bringing about the death pathway and inducing apoptosis. Second, DSF–Cu activates the PI3K/PTEN/AKT survival signaling pathway [ ].
HER2 is an important determinant of survival for BCSCs that are associated with a high risk of tumor recurrence [ , ]. HER2 overexpression [ ] is able to alter the apoptotic and molecular signaling pathways [ ]. DSF/Cu combination was reported to inhibit HER2/Akt signaling and eliminated BCSCs, suggesting the potential effectiveness of DSF for HER2-positive breast cancer treatment [ ].
TNBC represents an aggressive subtype, for which radiation and chemotherapy are the only options [ ]. Acquired chemoresistance remains the primary cause of therapeutic failure of TNBC [ ]. In the clinic, the relapsed TNBC is commonly pan-resistant to various drugs with entirely different resistance mechanisms. Investigation of the resistance mechanisms and development of new drugs to target pan-chemoresistance will potentially improve the therapeutic outcomes of TNBC patients [ ].
The MDA-MB-231 PAC10 cell line is made up of a high population of cells expressing stem cell markers that may play a vital role in the pan-resistance. These cells express high ALDH activity and a panel of embryonic stem cell–related proteins (e.g., Oct4, Sox2, Nanog) and nuclearization of HIF2a and NF-kBp65. These cells are highly cross-resistant to PAC, cisplatin (CDDP), docetaxel, and doxorubicin. DSF was reported to abolish CSC characters and completely reverse PAC and CDDP resistance in MDA-MB-231 PAC10 cells ( Table 5.2 ). In addition, DSF/Cu exposure for 4 h leads to inhibition of both ALDH activity and expression of Sox2 and Nanog in the resistant cells. In combination with DSF/Cu, the cytotoxicity of PAC and CDDP in MDA-MB-231 PAC10PAC10 cells was significantly higher than PAC, CDDP, or DSF/Cu single-drug exposure. The cytotoxicity of DSF/Cu plus PAC was synergistic in a wide range of concentrations [ ].
DSF/Cu combination induced the expression of Bax while inhibited the expression of Bcl2 in MDA-MB-231 PAC10 cells, which lead to a significant increase in Bax/Bcl2 ratio in the resistant cell line. Expression of p21 and p53 proteins was also enhanced by DSF/Cu combination, which did not effect on that of CDK2 and cyclin D1 and E [ ].
Analysis of the impact of various treatments on CSCs population in MDA-MB-231 and T47D cell lines revealed that formation of mammosphere from both cell lines was blocked entirely by exposure to the combination of DSF (1 μM)/Cu (1 μM) plus PAC (40 nM) for 48 h but not affected by PAC, DSF, or Cu alone. In addition, DSF/Cu, but not DSF or Cu, treatment significantly inhibited the ALDH-positive population in mammospheres [ ].
Although there have been advances in multimodality treatment, inflammatory breast cancer (IBC) is a discrete, advanced BC subtype characterized by high rates of residual disease and recurrence [ , ]. Metagene analysis of patient samples revealed significantly higher oxidative stress response (OSR) scores in IBC tumor samples when compared to normal or non-IBC tissues. These higher OSR scores were responsible for the inadequate response of IBC tumors to standard treatment strategies. DSF–Cu antagonized NF-kB signaling [ ], ALDH activity, and antioxidant levels [ ], causing the induction of oxidative stress–mediated apoptosis in multiple IBC cellular models [ ].
Under in vivo condition, DSF–Cu caused apoptosis and inhibited growth exclusively in tumor cells without significant toxicity. It has been suggested that strong redox adaption of IBC tumors may contribute to their resistance to ROS-inducing therapies. DSF, through redox modulation, may be advantageous in the manner of enhancing chemo- and/or radiosensitivity for extreme BC subtypes that showed therapeutic contention. DSF's potency was significantly enhanced by the addition of 10 μM Cu (DSF–Cu), with a decrease of roughly 100-fold in IC 50 values in SUM149 and rSUM149 ( Table 5.2 ). Remarkably, DSF-Cu caused cell death in redox-adapted rSUM149 cells at levels comparable to parental redox-sensitive SUM149 cells [ ].
DSF–Cu complex is nontoxic, at up to 20 µM , to normal, immortalized breast cells (MCF10A). Similar to SUM149 cells, DSF-induced, Cu-dependent cell death was observed in other IBC cell lines tested, which included MDA-IBC-3 (HER2-overexpressing), SUM190 (HERer2-overexpressing, ROS sensitive), and rSUM190 (an isogenic derivative of SUM190 with therapeutic resistance, redox adaptation) ( Table 5.2 ). SUM190 cells, even at extremely reduced concentrations of DSF, were exceedingly susceptible to DSF–Cu treatment. In the presence of DSF–Cu, the lessening of IBC cell viability corresponded with a drop off in levels of XIAP, the most potent mammalian caspase inhibitor and antiapoptotic protein. This is coherent with the induction of oxidative stress triggering the intrinsic apoptotic pathway. XIAP overexpression in IBC cells was identified to be correlated with resistance to therapeutic apoptosis. DSF–Cu-mediated cell death was also related to the decreased expression of eIF4G1 (a disease pathogenesis factor identified in IBC tumors) and increased PARP cleavage. These findings suggested the induction of apoptotic cell death being a fundamental mode of action of DSF–Cu [ ].
Addition of bathocuproine disulfonate or tetrathiomolybdate (TTM), two high affinity chelators to sequester free Cu, hindered DSF–Cu-induced cell death altogether in SUM149 and SUM149 cells, thus showing the importance of Cu binding in the intensification of DSF's cytotoxic effects. Although TTM is able to induce cancer cell death through sequestering Cu, the TTM–Cu mixture showed no effect in these models of IBC. These data confirmed that the activity of DSF is not related to Cu sequestration, but rather a gain of function that results in increased ROS production [ ].
Han et al. reported that without copper supplementation, exogenous SOD potentiated subtoxic DSF toxicity antagonized by subtoxic TTM or by the antioxidant N-acetylcysteine. Exogenous glucose oxidase, another H 2 O 2 generator, paralleled exogenous SOD in potentiating subtoxic DSF. It was confirmed that potentiation of sublethal DSF toxicity by extracellular H 2 O 2 against the human tumor cell lines only needed basal Cu and heightened ROS production. This is critical as these findings accentuate the importance of extracellular H 2 O 2 as a novel mechanism to improve the anticancer effects of DSF while minimizing copper toxicity [ ]. The authors also documented that DSF could significantly suppress the TGF-β–induced upregulation of vimentin and N-cadherin as well as downregulation of E-cadherin in a dose-dependent manner. These findings suggested that DSF can inhibit TGF-β–induced EMT by modifying the expression of EMT-related proteins.
DSF was able to affect 20S proteasome activity at low micromolar concentrations, but through a mechanism different from that of the authentic proteasome inhibitors as bortezomib or MG132 that block the active site of the proteasome. DSF acts at some other sites on the proteasome as a slow-binding partial noncompetitive inhibitor [ ]. However, DSF and its combination with copper exhibited a strong inhibitory effect on 26S proteasomes [ ], and the inhibition was copper-dependent [ ].
MDA-MB-231 BC cells with high levels of cellular copper showed proteasome inhibition in the presence of DSF alone, which suggested that copper acts as an endogenous element in the cell, which can form a complex with DSF and induce cell death. Similar findings were observed in mice bearing MDA-MB-231 tumor xenografts treated with DSF that inhibited tumor growth and proteasome activity and induced apoptosis in vivo [ ] ( Table 5.2 ).
A study reported that breast cancer MCF-10 DCIS cells were more sensitive to DSF–Cd-induced 20S proteasome inhibition and apoptosis induction than nontumorigenic human breast MCF-10A cells [ , ]. Thus, the DSF–Cd complex could selectively induce proteasome inhibition and apoptosis in human breast tumor cells [ ].
The availability of extracellular zinc was suggested to influence DSF efficacy significantly. Live cell confocal microscopy using fluorescent endocytic probes and the zinc dye FluoZin-3 showed that DSF elevated zinc levels selectively and speedily in endolysosomes. DSF was reported to cause the spatial disorganization of late endosomes and lysosomes, suggesting that they could be used as novel targets for DSF. Moreover, DSF was shown to increase the intracellular zinc levels in breast cancer cells specifically. It was reported that 10–100 μM DSF greatly heightened intracellular zinc levels in both MCF-7 and MDA-MB-231 cell lines ( Table 5.2 ), while zinc levels in the noncancerous MCF-10A cells remained unaffected by the same treatment. In serum-free media containing low zinc and copper, neither sodium pyrithione nor DSF induced a statistically significant increase in intracellular zinc in MCF-7 cells. Supplementation of serum-free media with 20 μM zinc was enough to restore completely, and exaggerate, the ionophore ability of both DSF and sodium pyrithione, demonstrating that this ionophore activity is dependent on extracellular zinc levels [ ].
Robinson et al. reported that DSF, as well as the related compound thiram, were identified as the most potent growth inhibitors against multiple TNBC cell lines. Combination treatment with four different drugs commonly used to treat TNBC revealed that DSF synergizes most effectively with doxorubicin to inhibit cell growth of TNBC cells. DSF and doxorubicin cooperated to induce cell death as well as cellular senescence and targeted the ESA+/CD24−/low/CD44+ CSC population. The results suggested that DSF may be repurposed to treat TNBC in combination with doxorubicin [ ].
Studies performed on HCC70, MDA-MB-231, MDA-MB-436, and Bt549 cell lines revealed that DSF was more effective against each cell line than doxorubicin, daunorubicin, mitoxantrone, colchicine, or PAC. Notably, MDA-MB-436 cells were resistant to the mitotic inhibitors, colchicine, and PAC but highly susceptible to DSF. In a panel of 13 human-derived TNBC lines, both DSF and thiram effectively suppressed the growth of TNBC cells, with an average IC 50 across all lines of 300 and 360 nM, respectively ( Table 5.2 ). Similar effect was seen when these drugs were used in basal-A or basal-B TNBC cell lines. These and other results all indicate that DSF/Cu is cytotoxic and nonspecific to breast cancer cells and can kill all subtypes of breast cancer.
There was a strong synergistic effect between DSF/Cu and PAC over a wide range of concentrations (IC 50 – IC 90 ). DSF/Cu significantly enhanced (3.7- to 15.5-fold) the cytotoxicity of PAC in BC cell lines. Contrary to the slight induction of apoptosis at low concentration of PAC alone (1 nM), the proportion of apoptotic cells was greatly elevated by DSF/Cu (DSF 100–150 nM/Cu 1 μM) and PAC in combination. DSF/Cu-induced cytotoxicity in BC cell lines was caused by ROS activation and was reversed by the addition of NAC, a ROS inhibitor, in the culture [ ].
DSF/Cu combination was also seen to inhibit NF-kB activity in BC cell lines. The NF-kB is a ROS-induced transcription factor with substantial antiapoptotic activity, which in turn dampens the proapoptotic effect of ROS [ ]. Therefore, the blocking of NF-kB activation enhances ROS-induced cytotoxicity. Both PAC and DSF/Cu inhibited NF-kB DNA-binding activity in BC cell lines. The most potent inhibition was observed in the BC cells treated with PAC/DSF/Cu in combination.
The clonogenicity of BC cell lines was significantly inhibited by DSF/Cu and eradicated by exposure to PAC plus DSF/Cu [ ]. Furthermore, inhibition of ALDH activity by DSF has been suggested to contribute to reduce CSCs and overcome drug resistance [ ].
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here