Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells
Abstract
:1. Cancer, Chemotherapy, and Chemoresistance
2. Mechanisms of Chemoresistance in Breast Cancers
3. Counteracting Chemoresistance in Breast Cancers: Why Metformin?
3.1. Indirect and ‘Direct’ Anti-Cancer/Anti-Tumor Effects of Metformin in Breast Cancers
3.2. Chemo-Sensitizing/Re-Sensitizing Effects of Metformin in Breast Cancer
3.2.1. Endocrine Therapy Resistance and Metformin
3.2.2. Doxorubicin (DOX) and Metformin
3.2.3. Everolimus and Metformin
3.2.4. Trastuzumab and Metformin
3.2.5. Cisplatin and Metformin
3.2.6. Multidrug Resistance and Metformin
4. Role of BCSCs in Chemoresistance in Breast Cancers
4.1. BCSCs and Dormancy/Quiescence
4.2. Tumor Microenvironment
4.3. Altered Metabolism
4.4. Epithelial–Mesenchymal Transition (EMT)
4.5. Drug Efflux Transporters, Detoxification Mechanisms, and Drug Uptake-Related Chemoresistance, in BCSCs
4.6. Resistance to DNA Damage: The Role of Advanced DNA Repair Systems
4.7. Signaling Pathways and BCSC-Driven Resistance
5. Effect of Metformin on BCSCs in Counteracting Drug Resistance
5.1. Possible Mechanisms of Action of Metformin on BCSCs
5.2. Metformin and Counteracting Therapeutic Resistance in BCSCs
5.3. Advantages of Combinatory Metformin and BCSC Targeted Therapy over Conventional Cancer Cell-Targeted Therapy
- (1)
- The BCSCs, while resistant to several routinely used anti-cancer agents, are sensitive to metformin. The less abundant CSCs that survive the administration of the routinely used anti-cancer agents are ultimately responsible for differentiation into tumor progenitor cells that cause a relapse/recurrence of cancer. Besides, CSCs can self-renew and may also undergo mutation to give rise to advanced and aggressive forms of the tumor [127,244,245,246,275,276,281].
- (2)
- (3)
- Metformin treatment re-sensitizes the CSCs to multiple drugs and radiation, thus making the CSCs more susceptible to the routinely used standard anti-cancer agents and therefore improving the efficacy of the therapeutic intervention [127].
- (4)
- Combinatory metformin therapy to target CSCs increases the efficacy of the treatment and improves the overall outcome and prognosis. The inhibition of invasiveness and metastasis dramatically reduces the chances of cancer relapse/recurrence. Furthermore, the development of acquired resistance to drugs can be minimized when used in combinations [127,244,245,246,275,276,282].
6. Challenges and Future Directions
6.1. Intrinsic and Acquired Resistance to Metformin
6.2. Targeted Drug (Metformin) Delivery Systems for Improved Therapeutic Efficacy
6.3. Lack of Data from Clinical Trials
6.4. Future Directions
7. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
5-FU | 5-fluorouracil |
ABC | ATP binding cassette |
ABCB1 | ABC sub-family B1; MDR1; P-gp |
ABCB5 | ABC sub-family B5 |
ABCC1 | ABC sub-family C1 |
ABCC3 | ABC sub-family C3 |
ABCG2 | ABC sub-family G2; BCRP |
ACC | Acetyl–CoA carboxylase |
Akt | Protein kinase B |
ALDH | Aldehyde dehydrogenase |
AMPK | 5′ adenosine monophosphate-activated protein kinase |
BCRP | Breast cancer resistance protein; ABCG2 |
BCSCs | Breast cancer stem cells |
BL1 | Basal-like 1; TNBC subtype |
BL2 | Basal-like 2; TNBC subtype |
CAFs | Cancer-associated fibroblasts |
CCL2 | Chemotactic protein 1 |
CSCs | Cancer stem cells |
CTCs | Circulating tumor cells |
CXCL7 | Chemokine (C-X-C motif) ligand 7 |
CXCR1 | Chemokine (C-X-C motif) receptor 1 |
DDIT4 | DNA damage-inducible transcript 4 |
DICER | Double-stranded RNA specific endoribonuclease |
DNMT | DNA-methyl transferase |
DOX | Doxorubicin |
DTCs | Disseminated tumor cells |
DTX | Docetaxel |
DVL3 | Dishevelled 3 |
EGFR | Epidermal growth factor receptor |
EMT | Epithelial-mesenchymal transition |
Epi | Epirubicin |
ER | Estrogen receptor |
ERK | Extracellular signal-regulated kinase |
FAC | 5-FU, DOX and cyclophosphamide |
FACS | Fluorescence activated cell sorter |
FAK | Focal adhesion kinase |
FEC | 5-FU, epirubicin and cyclophosphamide |
Gli | Glioma-associated oncogene homolog |
GRP78 | Glucose regulated protein 78 |
GSK-3β | Glycogen synthase kinase 3 beta |
GST | Glutathione S-transferase |
HER2 | Human epidermal growth factor receptor 2 |
Hh | Hedgehog |
HIF-1α/β | Hypoxia-inducible factor 1 alpha/beta |
HMGB1 | High-mobility group box 1 |
IC50 | Half maximal inhibitory concentration |
IGF-1 | Insulin-like growth factor 1 |
IGF-1R | IGF-1 receptor |
IL6/8 | Interleukin 6/8 |
IM | Immunomodulatory; TNBC subtype |
JAK | Janus kinase |
KLF5 | Krüppel-like factor 5 |
LAR | Luminal androgen receptor; TNBC subtype |
M | Mesenchymal; TNBC subtype |
MAPK | Mitogen activated protein kinase |
MATE1/2 | Multidrug and toxin extrusion protein 1 and 2 |
MCL1 | Myeloid cell leukemia 1 |
MDR | Multi-drug resistance |
MDR1 | Multi-drug resistance protein 1; ABCB1; P-gp |
MET | Mesenchymal-epithelial transition |
miRNA | Micro-RNA |
Mito | Mitomycin |
MRP1 | Multidrug resistance-associated protein 1; ABCC1 |
MSFE | Mammosphere forming efficiency |
MSL | Mesenchymal stem-like; TNBC subtype |
mTOR | Mammalian target of rapamycin |
NF-κB | Nuclear factor-kappa B |
NOD | Non-obese diabetic |
NRF2 | Nuclear erythroid-related factor 2 |
O-CMC | O-carboxymethyl chitosan |
OCT1/2/3 | Organic cation transporters 1, 2 and 3 |
OS | Overall survival |
P-gp | P-glycoprotein; ABCB1; MDR1 |
PARP1 | Poly (ADP-ribose) polymerase 1 |
PDGF-B | Platelet-derived growth factor B |
PERK | Protein kinase RNA-like endoplasmic reticulum kinase |
PFS | Progression free survival |
PHGDH | Phosphoglycerate dehydrogenase |
PI3K | Phosphatidylinositol-3-kinase |
PKA | Protein kinase A |
PLGA-PEG | Poly (lactic acid-co-glycolic acid)-polyethylene glycol |
PMAT | Plasma membrane monoamine transporter |
PR | Progesterone receptor |
Ptch | Patched; Hh ligand receptor |
PTX | Paclitaxel |
RAD51 | RecA homolog DNA repair recombinase |
RANK-L | Receptor activator of nuclear factor-kappa B ligand |
REDD1 | Regulated DNA damage 1 |
RFS | Recurrence-free survival |
SCID | Severe combined immunodeficiency |
SHh | Sonic Hh |
SMAD | Sma-and mothers against decapentaplegic homolog |
Smo | Smoothened; Hh ligand co-receptor |
SOX2 | Sex determining region Y box transcription factor 2 |
STAT3 | Signal transducer and activator of transcription 3 |
TAMs | Tumor associated macrophages |
TFF1 | Trefoil factor 1/protein pS2 |
TGFβ | Transforming growth factor beta |
TNBCs | Triple negative breast cancers |
TNFα | Tumor necrosis factor alpha |
XIAP | X-linked inhibitor of apoptosis |
References
- WHO. Cancer-Fact Sheets. 2018. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 9 June 2020).
- Hawkes, N. A comprehensive history of cancer treatment. 2015. Available online: https://www.raconteur.net/healthcare/history-of-cancer-treatment (accessed on 9 June 2020).
- ACS. The History of Cancer-Cancer in the Sixteenth to Eighteenth Centuries. 2014. Available online: https://www.cancer.org/cancer/cancer-basics/history-of-cancer/sixteenth-to-eighteenth-centuries.html (accessed on 9 June 2020).
- Morrison, W.B. Cancer Chemotherapy: An Annotated History. J. Vet. Intern. Med. 2010, 24, 1249–1262. [Google Scholar] [CrossRef]
- DeVita, V.T.; Chu, E. A History of Cancer Chemotherapy. Cancer Res. 2008, 68, e8643. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, F.; Efferth, T. Tumor Heterogeneity, Single-Cell Sequencing, and Drug Resistance. Pharmaceuticals 2016, 9, 33. [Google Scholar] [CrossRef]
- Chatterjee, N.; Bivona, T.G. Polytherapy and Targeted Cancer Drug Resistance. Trends Cancer 2019, 5, 170–182. [Google Scholar] [CrossRef]
- McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef]
- Konieczkowski, D.J.; Johannessen, C.M.; Garraway, L.A. A Convergence-Based Framework for Cancer Drug Resistance. Cancer Cell 2018, 33, 801–815. [Google Scholar] [CrossRef] [Green Version]
- Gupta, P.B.; Pastushenko, I.; Skibinski, A.; Blanpain, C.; Kuperwasser, C. Phenotypic Plasticity: Driver of Cancer Initiation, Progression, and Therapy Resistance. Cell Stem Cell 2019, 24, 65–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muley, H.; Fadó, R.; Rodríguez-Rodríguez, R.; Casals, N. Drug uptake-based chemoresistance in breast cancer treatment. Biochem. Pharmacol. 2020, 177, e113959. [Google Scholar] [CrossRef] [PubMed]
- Varghese, E.; Samuel, S.M.; Abotaleb, M.; Cheema, S.; Mamtani, R.; Büsselberg, D. The “Yin and Yang” of Natural Compounds in Anticancer Therapy of Triple-Negative Breast Cancers. Cancers 2018, 10, e346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Varghese, E.; Samuel, S.M.; Líšková, A.; Samec, M.; Kubatka, P.; Büsselberg, D. Targeting Glucose Metabolism to Overcome Resistance to Anticancer Chemotherapy in Breast Cancer. Cancers 2020, 12, e2252. [Google Scholar] [CrossRef] [PubMed]
- Subbiah, S.; Gopu, G.; Senthilkumar, P.; Muniasamy, P. Molecular subtypes as a predictor of response to neoadjuvant chemotherapy in breast cancer patients. Indian J. Cancer 2017, 54, 652–657. [Google Scholar] [CrossRef] [PubMed]
- Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- ACS. Treatment of Breast Cancer by Stage. 2019. Available online: https://www.cancer.org/cancer/breast-cancer/treatment/treatment-of-breast-cancer-by-stage.html (accessed on 9 June 2020).
- Moiseenko, F.; Volkov, N.; Bogdanov, A.; Dubina, M.; Moiseyenko, V. Resistance mechanisms to drug therapy in breast cancer and other solid tumors: An opinion. F1000Research 2017, 6. [Google Scholar] [CrossRef] [Green Version]
- Tripathi, L.; Datta, S.S.; Agrawal, S.K.; Chatterjee, S.; Ahmed, R. Stigma Perceived by Women Following Surgery for Breast Cancer. Indian J. Med. Paediatr Oncol. 2017, 38, 146–152. [Google Scholar] [CrossRef] [PubMed]
- Vrinten, C.; Gallagher, A.; Waller, J.; Marlow, L.A.V. Cancer stigma and cancer screening attendance: A population based survey in England. BMC Cancer 2019, 19, e566. [Google Scholar] [CrossRef] [Green Version]
- Rivera-Franco, M.M.; Leon-Rodriguez, E. Delays in Breast Cancer Detection and Treatment in Developing Countries. Breast Cancer 2018, 12, e1178223417752677. [Google Scholar] [CrossRef] [Green Version]
- Gottesman, M.M. Mechanisms of Cancer Drug Resistance. Ann. Rev. Med. 2002, 53, 615–627. [Google Scholar] [CrossRef] [Green Version]
- Ozakinci, G.; Sobota, A.; Humphris, G. Fear of Cancer Recurrence Among Breast Cancer Survivors. Curr. Breast Cancer Rep. 2014, 6, 219–225. [Google Scholar] [CrossRef]
- Ziner, K.W.; Sledge, G.W.; Bell, C.J.; Johns, S.; Miller, K.D.; Champion, V.L. Predicting fear of breast cancer recurrence and self-efficacy in survivors by age at diagnosis. Oncol. Nurs. Forum 2012, 39, 287–295. [Google Scholar] [CrossRef] [Green Version]
- Van Maaren, M.C.; de Munck, L.; Strobbe, L.J.A.; Sonke, G.S.; Westenend, P.J.; Smidt, M.L.; Poortmans, P.M.P.; Siesling, S. Ten-year recurrence rates for breast cancer subtypes in the Netherlands: A large population-based study. Int. J. Cancer 2019, 144, 263–272. [Google Scholar] [CrossRef] [Green Version]
- Shim, H.J.; Kim, S.H.; Kang, B.J.; Choi, B.G.; Kim, H.S.; Cha, E.S.; Song, B.J. Breast cancer recurrence according to molecular subtype. Asian Pac. J. Cancer Prev. 2014, 15, 5539–5544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Montagna, E.; Bagnardi, V.; Rotmensz, N.; Viale, G.; Renne, G.; Cancello, G.; Balduzzi, A.; Scarano, E.; Veronesi, P.; Luini, A.; et al. Breast cancer subtypes and outcome after local and regional relapse. Ann. Oncol. 2012, 23, 324–331. [Google Scholar] [CrossRef]
- Lim, E.; Metzger-Filho, O.; Winer, E.P. The natural history of hormone receptor-positive breast cancer. Oncology 2012, 26, 688–694. [Google Scholar] [PubMed]
- Zimmerman, M.P.; Mehr, S.R. Recurrence of Breast Cancer Years After the Initial Tumor. Am. J. Manag. Care 2014, 20, SP409–SP413. [Google Scholar]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
- Seyfried, T.N.; Flores, R.E.; Poff, A.M.; D’Agostino, D.P. Cancer as a metabolic disease: Implications for novel therapeutics. Carcinogenesis 2014, 35, 515–527. [Google Scholar] [CrossRef]
- De Martel, C.; Ferlay, J.; Franceschi, S.; Vignat, J.; Bray, F.; Forman, D.; Plummer, M. Global burden of cancers attributable to infections in 2008: A review and synthetic analysis. Lancet Oncol. 2012, 13, 607–615. [Google Scholar] [CrossRef]
- Van Elsland, D.; Neefjes, J. Bacterial infections and cancer. EMBO Rep. 2018, 19, e46632. [Google Scholar] [CrossRef]
- Morales-Sánchez, A.; Fuentes-Pananá, E.M. Human viruses and cancer. Viruses 2014, 6, 4047–4079. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Zhang, H.; Chen, X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019, 2, 141–160. [Google Scholar] [CrossRef] [Green Version]
- Lovly, C.M.; Salama, A.K.S.; Salgia, R. Tumor Heterogeneity and Therapeutic Resistance. Am. Soc. Clin. Oncol. Educ. Book 2016, e585–e593. [Google Scholar] [CrossRef]
- Dzobo, K.; Senthebane, D.A.; Thomford, N.E.; Rowe, A.; Dandara, C.; Parker, M.I. Not Everyone Fits the Mold: Intratumor and Intertumor Heterogeneity and Innovative Cancer Drug Design and Development. Omics: A J. Integr. Biol. 2018, 22, 17–34. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Qin, Y.; Liu, S. Cytokines, breast cancer stem cells (BCSCs) and chemoresistance. Clin. Transl. Med. 2018, 7, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chuthapisith, S.; Eremin, J.; El-Sheemey, M.; Eremin, O. Breast cancer chemoresistance: Emerging importance of cancer stem cells. Surg. Oncol. 2010, 19, 27–32. [Google Scholar] [CrossRef] [PubMed]
- De Angelis, M.L.; Francescangeli, F.; Zeuner, A. Breast Cancer Stem Cells as Drivers of Tumor Chemoresistance, Dormancy and Relapse: New Challenges and Therapeutic Opportunities. Cancers 2019, 11, e1569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ji, X.; Lu, Y.; Tian, H.; Meng, X.; Wei, M.; Cho, W.C. Chemoresistance mechanisms of breast cancer and their countermeasures. Biomed. Pharmacother. 2019, 114, e108800. [Google Scholar] [CrossRef]
- Ma, L.; Zong, X. Metabolic Symbiosis in Chemoresistance: Refocusing the Role of Aerobic Glycolysis. Front. Oncol. 2020, 10, 5. [Google Scholar] [CrossRef] [Green Version]
- Nunes, T.; Hamdan, D.; Leboeuf, C.; El Bouchtaoui, M.; Gapihan, G.; Nguyen, T.T.; Meles, S.; Angeli, E.; Ratajczak, P.; Lu, H.; et al. Targeting Cancer Stem Cells to Overcome Chemoresistance. Int. J. Mol. Sci. 2018, 19, e4036. [Google Scholar] [CrossRef] [Green Version]
- Velaei, K.; Samadi, N.; Barazvan, B.; Soleimani Rad, J. Tumor microenvironment-mediated chemoresistance in breast cancer. Breast 2016, 30, 92–100. [Google Scholar] [CrossRef]
- Housman, G.; Byler, S.; Heerboth, S.; Lapinska, K.; Longacre, M.; Snyder, N.; Sarkar, S. Drug resistance in cancer: An overview. Cancers 2014, 6, 1769–1792. [Google Scholar] [CrossRef] [Green Version]
- Hu, W.; Tan, C.; He, Y.; Zhang, G.; Xu, Y.; Tang, J. Functional miRNAs in breast cancer drug resistance. Onco Targets Ther. 2018, 11, 1529–1541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nedeljković, M.; Damjanović, A. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer-How We Can Rise to the Challenge. Cells 2019, 8, e957. [Google Scholar] [CrossRef] [Green Version]
- Blendea, M.C.; Thompson, M.J.; Malkani, S. Diabetes and Chronic Liver Disease: Etiology and Pitfalls in Monitoring. Clin. Diabetes 2010, 28, e139. [Google Scholar] [CrossRef] [Green Version]
- Leon, B.M.; Maddox, T.M. Diabetes and cardiovascular disease: Epidemiology, biological mechanisms, treatment recommendations and future research. World J. Diabetes 2015, 6, 1246–1258. [Google Scholar] [CrossRef]
- Min, T.Z.; Stephens, M.W.; Kumar, P.; Chudleigh, R.A. Renal complications of diabetes. Br. Med. Bull. 2012, 104, 113–127. [Google Scholar] [CrossRef]
- Said, G. Diabetic neuropathy--a review. Nat. Clin. Pract. Neurol. 2007, 3, 331–340. [Google Scholar] [CrossRef]
- Samuel, S.M.; Varghese, E.; Kubatka, P.; Triggle, C.R.; Büsselberg, D. Metformin: The Answer to Cancer in a Flower? Current Knowledge and Future Prospects of Metformin as an Anti-Cancer Agent in Breast Cancer. Biomolecules 2019, 9, e846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samuel, S.M.; Varghese, E.; Varghese, S.; Busselberg, D. Challenges and perspectives in the treatment of diabetes associated breast cancer. Cancer Treat. Rev. 2018, 70, 98–111. [Google Scholar] [CrossRef] [Green Version]
- Giovannucci, E.; Harlan, D.M.; Archer, M.C.; Bergenstal, R.M.; Gapstur, S.M.; Habel, L.A.; Pollak, M.; Regensteiner, J.G.; Yee, D. Diabetes and cancer: A consensus report. Diabetes Care 2010, 33, 1674–1685. [Google Scholar] [CrossRef] [Green Version]
- Onitilo, A.A.; Engel, J.M.; Glurich, I.; Stankowski, R.V.; Williams, G.M.; Doi, S.A. Diabetes and cancer I: Risk, survival, and implications for screening. Cancer Causes Control 2012, 23, 967–981. [Google Scholar] [CrossRef] [Green Version]
- Chowdhury, T.A. Diabetes and cancer. QJM Mon. J. Assoc. Physicians 2010, 103, 905–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryu, T.Y.; Park, J.; Scherer, P.E. Hyperglycemia as a risk factor for cancer progression. Diabetes Metab. J. 2014, 38, 330–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, C.X.; Zhu, H.H.; Zhu, Y.M. Diabetes and cancer: Associations, mechanisms, and implications for medical practice. World J. Diabetes 2014, 5, 372–380. [Google Scholar] [CrossRef] [PubMed]
- Evans, J.M.; Donnelly, L.A.; Emslie-Smith, A.M.; Alessi, D.R.; Morris, A.D. Metformin and reduced risk of cancer in diabetic patients. BMJ 2005, 330, 1304–1305. [Google Scholar] [CrossRef] [Green Version]
- De, A.; Kuppusamy, G. Metformin in breast cancer: Preclinical and clinical evidence. Curr. Probl. Cancer 2019. [Google Scholar] [CrossRef]
- Chen, E.C.; Liang, X.; Yee, S.W.; Geier, E.G.; Stocker, S.L.; Chen, L.; Giacomini, K.M. Targeted disruption of organic cation transporter 3 attenuates the pharmacologic response to metformin. Mol. Pharmacol. 2015, 88, 75–83. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Teranishi, K.; Li, S.; Yee, S.W.; Hesselson, S.; Stryke, D.; Johns, S.J.; Ferrin, T.E.; Kwok, P.; Giacomini, K.M. Genetic variants in multidrug and toxic compound extrusion-1, hMATE1, alter transport function. Pharm. J. 2009, 9, 127–136. [Google Scholar] [CrossRef] [Green Version]
- Choi, M.K.; Jin, Q.R.; Jin, H.E.; Shim, C.K.; Cho, D.Y.; Shin, J.G.; Song, I.S. Effects of tetraalkylammonium compounds with different affinities for organic cation transporters on the pharmacokinetics of metformin. Biopharm. Drug Dispos. 2007, 28, 501–510. [Google Scholar] [CrossRef]
- Kimura, N.; Masuda, S.; Tanihara, Y.; Ueo, H.; Okuda, M.; Katsura, T.; Inui, K. Metformin is a superior substrate for renal organic cation transporter OCT2 rather than hepatic OCT1. Drug Metab. Pharmacokinet. 2005, 20, 379–386. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Chen, Y.; Zhang, S.; More, S.S.; Huang, X.; Giacomini, K.M. Role of organic cation transporter 1, OCT1 in the pharmacokinetics and toxicity of cis-diammine(pyridine)chloroplatinum(II) and oxaliplatin in mice. Pharm. Res. 2011, 28, 610–625. [Google Scholar] [CrossRef] [Green Version]
- Liang, X.; Giacomini, K.M. Transporters Involved in Metformin Pharmacokinetics and Treatment Response. J. Pharm. Sci. 2017, 106, 2245–2250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Masuda, S.; Terada, T.; Yonezawa, A.; Tanihara, Y.; Kishimoto, K.; Katsura, T.; Ogawa, O.; Inui, K. Identification and functional characterization of a new human kidney-specific H+/organic cation antiporter, kidney-specific multidrug and toxin extrusion 2. J. Am. Soc. Nephrol. JASN 2006, 17, 2127–2135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, M.; Xia, L.; Wang, J. Metformin transport by a newly cloned proton-stimulated organic cation transporter (plasma membrane monoamine transporter) expressed in human intestine. Drug Metab. Dispos. 2007, 35, 1956–1962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, X.; Chien, H.C.; Yee, S.W.; Giacomini, M.M.; Chen, E.C.; Piao, M.; Hao, J.; Twelves, J.; Lepist, E.I.; Ray, A.S.; et al. Metformin Is a Substrate and Inhibitor of the Human Thiamine Transporter, THTR-2 (SLC19A3). Mol. Pharm. 2015, 12, 4301–4310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shu, Y.; Brown, C.; Castro, R.A.; Shi, R.J.; Lin, E.T.; Owen, R.P.; Sheardown, S.A.; Yue, L.; Burchard, E.G.; Brett, C.M.; et al. Effect of genetic variation in the organic cation transporter 1, OCT1, on metformin pharmacokinetics. Clin. Pharm. Ther. 2008, 83, 273–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shu, Y.; Sheardown, S.A.; Brown, C.; Owen, R.P.; Zhang, S.; Castro, R.A.; Ianculescu, A.G.; Yue, L.; Lo, J.C.; Burchard, E.G.; et al. Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J. Clin. Investig. 2007, 117, 1422–1431. [Google Scholar] [CrossRef] [Green Version]
- Cai, H.; Zhang, Y.; Han, T.K.; Everett, R.S.; Thakker, D.R. Cation-selective transporters are critical to the AMPK-mediated antiproliferative effects of metformin in human breast cancer cells. Int. J. Cancer 2016, 138, 2281–2292. [Google Scholar] [CrossRef] [Green Version]
- Checkley, L.A.; Rudolph, M.C.; Wellberg, E.A.; Giles, E.D.; Wahdan-Alaswad, R.S.; Houck, J.A.; Edgerton, S.M.; Thor, A.D.; Schedin, P.; Anderson, S.M.; et al. Metformin Accumulation Correlates with Organic Cation Transporter 2 Protein Expression and Predicts Mammary Tumor Regression in Vivo. Cancer Prev. Res. 2017, 10, 198–207. [Google Scholar] [CrossRef] [Green Version]
- Cai, H.; Everett, R.S.; Thakker, D.R. Efficacious dose of metformin for breast cancer therapy is determined by cation transporter expression in tumours. Br. J. Pharmacol. 2019, 176, 2724–2735. [Google Scholar] [CrossRef]
- Chowdhury, S.; Yung, E.; Pintilie, M.; Muaddi, H.; Chaib, S.; Yeung, M.; Fusciello, M.; Sykes, J.; Pitcher, B.; Hagenkort, A.; et al. MATE2 Expression Is Associated with Cancer Cell Response to Metformin. PLoS ONE 2016, 11, e0165214. [Google Scholar] [CrossRef]
- Correia, S.; Carvalho, C.; Santos, M.S.; Seica, R.; Oliveira, C.R.; Moreira, P.I. Mechanisms of action of metformin in type 2 diabetes and associated complications: An overview. Mini Rev. Med. Chem. 2008, 8, 1343–1354. [Google Scholar] [CrossRef] [PubMed]
- Rahmani, J.; Manzari, N.; Thompson, J.; Gudi, S.K.; Chhabra, M.; Naik, G.; Mousavi, S.M.; Varkaneh, H.K.; Clark, C.; Zhang, Y. The effect of metformin on biomarkers associated with breast cancer outcomes: A systematic review, meta-analysis, and dose-response of randomized clinical trials. Clin. Transl. Oncol. 2019. [Google Scholar] [CrossRef]
- Ferroni, P.; Riondino, S.; Buonomo, O.; Palmirotta, R.; Guadagni, F.; Roselli, M. Type 2 Diabetes and Breast Cancer: The Interplay between Impaired Glucose Metabolism and Oxidant Stress. Oxid. Med. Cell Longev. 2015, 2015, e183928. [Google Scholar] [CrossRef] [PubMed]
- Rojas, L.B.; Gomes, M.B. Metformin: An old but still the best treatment for type 2 diabetes. Diabetol. Metab. Syndr. 2013, 5, e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Triggle, C.R.; Ding, H. Metformin is not just an antihyperglycaemic drug but also has protective effects on the vascular endothelium. Acta Physiol. 2017, 219, 138–151. [Google Scholar] [CrossRef]
- Sośnicki, S.; Kapral, M.; Węglarz, L. Molecular targets of metformin antitumor action. Pharmacol. Rep. 2016, 68, 918–925. [Google Scholar] [CrossRef]
- Warburg, O.; Wind, F.; Negelein, E. THE METABOLISM OF TUMORS IN THE BODY. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef] [Green Version]
- Le, A.; Lane, A.N.; Hamaker, M.; Bose, S.; Gouw, A.; Barbi, J.; Tsukamoto, T.; Rojas, C.J.; Slusher, B.S.; Zhang, H.; et al. Glucose-Independent Glutamine Metabolism via TCA Cycling for Proliferation and Survival in B Cells. Cell Metab. 2012, 15, 110–121. [Google Scholar] [CrossRef] [Green Version]
- Metallo, C.M.; Gameiro, P.A.; Bell, E.L.; Mattaini, K.R.; Yang, J.; Hiller, K.; Jewell, C.M.; Johnson, Z.R.; Irvine, D.J.; Guarente, L.; et al. Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia. Nature 2012, 481, 380–384. [Google Scholar] [CrossRef] [Green Version]
- Samuel, S.M.; Ghosh, S.; Majeed, Y.; Arunachalam, G.; Emara, M.M.; Ding, H.; Triggle, C.R. Metformin represses glucose starvation induced autophagic response in microvascular endothelial cells and promotes cell death. Biochem. Pharmacol. 2017, 132, 118–132. [Google Scholar] [CrossRef]
- Daugan, M.; Dufaÿ Wojcicki, A.; d’Hayer, B.; Boudy, V. Metformin: An anti-diabetic drug to fight cancer. Pharmacol. Res. 2016, 113, 675–685. [Google Scholar] [CrossRef]
- Hawley, S.A.; Gadalla, A.E.; Olsen, G.S.; Hardie, D.G. The Antidiabetic Drug Metformin Activates the AMP-Activated Protein Kinase Cascade via an Adenine Nucleotide-Independent Mechanism. Diabetes 2002, 51, e2420. [Google Scholar] [CrossRef] [Green Version]
- Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
- Meng, S.; Cao, J.; He, Q.; Xiong, L.; Chang, E.; Radovick, S.; Wondisford, F.E.; He, L. Metformin activates AMP-activated protein kinase by promoting formation of the αβγ heterotrimeric complex. J. Biol. Chem. 2015, 290, 3793–3802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Howell, J.J.; Hellberg, K.; Turner, M.; Talbott, G.; Kolar, M.J.; Ross, D.S.; Hoxhaj, G.; Saghatelian, A.; Shaw, R.J.; Manning, B.D. Metformin Inhibits Hepatic mTORC1 Signaling via Dose-Dependent Mechanisms Involving AMPK and the TSC Complex. Cell Metab. 2017, 25, 463–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sabnis, H.S.; Somasagara, R.R.; Bunting, K.D. Targeting MYC Dependence by Metabolic Inhibitors in Cancer. Genes 2017, 8, e114. [Google Scholar] [CrossRef]
- Shen, P.; Reineke, L.C.; Knutsen, E.; Chen, M.; Pichler, M.; Ling, H.; Calin, G.A. Metformin blocks MYC protein synthesis in colorectal cancer via mTOR-4EBP-eIF4E and MNK1-eIF4G-eIF4E signaling. Mol. Oncol. 2018, 12, 1856–1870. [Google Scholar] [CrossRef] [Green Version]
- Hattori, Y.; Suzuki, K.; Hattori, S.; Kasai, K. Metformin Inhibits Cytokine-Induced Nuclear Factor κB Activation Via AMP-Activated Protein Kinase Activation in Vascular Endothelial Cells. Hypertension 2006, 47, 1183–1188. [Google Scholar] [CrossRef] [Green Version]
- Sekino, N.; Kano, M.; Matsumoto, Y.; Sakata, H.; Akutsu, Y.; Hanari, N.; Murakami, K.; Toyozumi, T.; Takahashi, M.; Otsuka, R.; et al. Antitumor effects of metformin are a result of inhibiting nuclear factor kappa B nuclear translocation in esophageal squamous cell carcinoma. Cancer Sci. 2018, 109, 1066–1074. [Google Scholar] [CrossRef]
- Xu, S.; Yang, Z.; Jin, P.; Yang, X.; Li, X.; Wei, X.; Wang, Y.; Long, S.; Zhang, T.; Chen, G.; et al. Metformin Suppresses Tumor Progression by Inactivating Stromal Fibroblasts in Ovarian Cancer. Mol. Cancer Ther. 2018, 17, e1291. [Google Scholar] [CrossRef] [Green Version]
- Li, P.; Zhao, M.; Parris, A.; Feng, X.; Yang, X. P53 is required for metformin-induced growth inhibition, senescence and apoptosis in breast cancer cells. Biochem. Biophys. Res. Commun. 2015, 464. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Ahmad, N.; Liu, X. Combining p53 stabilizers with metformin induces synergistic apoptosis through regulation of energy metabolism in castration-resistant prostate cancer. Cell Cycle 2016, 15, 840–849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yi, Y.; Zhang, W.; Yi, J.; Xiao, Z.-X. Role of p53 Family Proteins in Metformin Anti-Cancer Activities. J. Cancer 2019, 10, 2434–2442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yi, G.; He, Z.; Zhou, X.; Xian, L.; Yuan, T.; Jia, X.; Hong, J.; He, L.; Liu, J. Low concentration of metformin induces a p53-dependent senescence in hepatoma cells via activation of the AMPK pathway. Int. J. Oncol. 2013, 43, 1503–1510. [Google Scholar] [CrossRef] [PubMed]
- Noren Hooten, N.; Martin-Montalvo, A.; Dluzen, D.F.; Zhang, Y.; Bernier, M.; Zonderman, A.B.; Becker, K.G.; Gorospe, M.; de Cabo, R.; Evans, M.K. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging Cell 2016, 15, 572–581. [Google Scholar] [CrossRef] [PubMed]
- Pulito, C.; Donzelli, S.; Muti, P.; Puzzo, L.; Strano, S.; Blandino, G. microRNAs and cancer metabolism reprogramming: The paradigm of metformin. Ann. Transl. Med. 2014, 2, 58. [Google Scholar] [CrossRef] [PubMed]
- Blandino, G.; Valerio, M.; Cioce, M.; Mori, F.; Casadei, L.; Pulito, C.; Sacconi, A.; Biagioni, F.; Cortese, G.; Galanti, S.; et al. Metformin elicits anticancer effects through the sequential modulation of DICER and c-MYC. Nat. Commun. 2012, 3, e865. [Google Scholar] [CrossRef] [PubMed]
- Lumachi, F.; Brunello, A.; Maruzzo, M.; Basso, U.; Basso, S.M. Treatment of estrogen receptor-positive breast cancer. Curr. Med. Chem. 2013, 20, 596–604. [Google Scholar] [CrossRef] [PubMed]
- Ma, J.; Guo, Y.; Chen, S.; Zhong, C.; Xue, Y.; Zhang, Y.; Lai, X.; Wei, Y.; Yu, S.; Zhang, J.; et al. Metformin enhances tamoxifen-mediated tumor growth inhibition in ER-positive breast carcinoma. BMC Cancer 2014, 14, e172. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Lee, J.; Jang, S.Y.; Kim, C.; Choi, Y.; Kim, A. Anticancer effect of metformin on estrogen receptor-positive and tamoxifen-resistant breast cancer cell lines. Oncol. Rep. 2016, 35, 2553–2560. [Google Scholar] [CrossRef] [Green Version]
- Giles, E.D.; Jindal, S.; Wellberg, E.A.; Schedin, T.; Anderson, S.M.; Thor, A.D.; Edwards, D.P.; MacLean, P.S.; Schedin, P. Metformin inhibits stromal aromatase expression and tumor progression in a rodent model of postmenopausal breast cancer. Breast Cancer Res. BCR 2018, 20, 50. [Google Scholar] [CrossRef]
- Zhao, Y.; Gong, C.; Wang, Z.; Zhang, J.; Wang, L.; Zhang, S.; Cao, J.; Tao, Z.; Li, T.; Wang, B.; et al. A randomized phase II study of aromatase inhibitors plus metformin in pre-treated postmenopausal patients with hormone receptor positive metastatic breast cancer. Oncotarget 2017, 8, 84224–84236. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.-C.; Li, G.-Y.; Wang, B.; Han, S.-X.; Sun, X.; Jiang, Y.-N.; Shen, Y.-W.; Zhou, C.; Feng, J.; Lu, S.-Y.; et al. Metformin inhibits metastatic breast cancer progression and improves chemosensitivity by inducing vessel normalization via PDGF-B downregulation. J. Exp. Clin. Cancer Res. 2019, 38, e235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shafiei-Irannejad, V.; Samadi, N.; Yousefi, B.; Salehi, R.; Velaei, K.; Zarghami, N. Metformin enhances doxorubicin sensitivity via inhibition of doxorubicin efflux in P-gp-overexpressing MCF-7 cells. Chem. Biol. Drug Des. 2018, 91, 269–276. [Google Scholar] [CrossRef] [PubMed]
- Shafiei-Irannejad, V.; Samadi, N.; Salehi, R.; Yousefi, B.; Rahimi, M.; Akbarzadeh, A.; Zarghami, N. Reversion of Multidrug Resistance by Co-Encapsulation of Doxorubicin and Metformin in Poly(lactide-co-glycolide)-d-α-tocopheryl Polyethylene Glycol 1000 Succinate Nanoparticles. Pharm. Res. 2018, 35, e119. [Google Scholar] [CrossRef] [PubMed]
- Rao, M.; Gao, C.; Guo, M.; Law, B.Y.K.; Xu, Y. Effects of metformin treatment on radiotherapy efficacy in patients with cancer and diabetes: A systematic review and meta-analysis. Cancer Manag. Res. 2018, 10, 4881–4890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davies, G.; Lobanova, L.; Dawicki, W.; Groot, G.; Gordon, J.R.; Bowen, M.; Harkness, T.; Arnason, T. Metformin inhibits the development, and promotes the resensitization, of treatment-resistant breast cancer. PLoS ONE 2017, 12, e0187191. [Google Scholar] [CrossRef] [Green Version]
- Ariaans, G.; Jalving, M.; Vries, E.G.E.d.; Jong, S.d. Anti-tumor effects of everolimus and metformin are complementary and glucose-dependent in breast cancer cells. BMC Cancer 2017, 17, e232. [Google Scholar] [CrossRef] [Green Version]
- Deng, J.; Peng, M.; Wang, Z.; Zhou, S.; Xiao, D.; Deng, J.; Yang, X.; Peng, J.; Yang, X. Novel application of metformin combined with targeted drugs on anticancer treatment. Cancer Sci. 2019, 110, 23–30. [Google Scholar] [CrossRef] [Green Version]
- Liu, B.; Fan, Z.; Edgerton, S.M.; Yang, X.; Lind, S.E.; Thor, A.D. Potent anti-proliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions. Cell Cycle 2011, 10, 2959–2966. [Google Scholar] [CrossRef] [Green Version]
- Zeglinski, M.; Ludke, A.; Jassal, D.S.; Singal, P.K. Trastuzumab-induced cardiac dysfunction: A ‘dual-hit’. Exp. Clin. Cardiol. 2011, 16, 70–74. [Google Scholar] [PubMed]
- Smith, T.A.; Phyu, S.M.; Akabuogu, E.U. Effects of Administered Cardioprotective Drugs on Treatment Response of Breast Cancer Cells. Anticancer Res. 2016, 36, 87–93. [Google Scholar] [PubMed]
- Vázquez-Martín, A.; Oliveras-Ferraros, C.; del Barco, S.; Martín-Castillo, B.; Menéndez, J.A. mTOR inhibitors and the anti-diabetic biguanide metformin: New insights into the molecular management of breast cancer resistance to the HER2 tyrosine kinase inhibitor lapatinib (Tykerb). Clin. Transl. Oncol. 2009, 11, 455–459. [Google Scholar] [CrossRef] [PubMed]
- Chung, Y.-C.; Chang, C.-M.; Wei, W.-C.; Chang, T.-W.; Chang, K.-J.; Chao, W.-T. Metformin-induced caveolin-1 expression promotes T-DM1 drug efficacy in breast cancer cells. Sci. Rep. 2018, 8, 3930. [Google Scholar] [CrossRef] [PubMed]
- Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.O.; Kang, M.J.; Byun, W.S.; Kim, S.A.; Seo, I.H.; Han, J.A.; Moon, J.W.; Kim, J.H.; Kim, S.J.; Lee, E.J.; et al. Metformin overcomes resistance to cisplatin in triple-negative breast cancer (TNBC) cells by targeting RAD51. Breast Cancer Res. 2019, 21, e115. [Google Scholar] [CrossRef] [Green Version]
- Tsai, M.S.; Kuo, Y.H.; Chiu, Y.F.; Su, Y.C.; Lin, Y.W. Down-regulation of Rad51 expression overcomes drug resistance to gemcitabine in human non-small-cell lung cancer cells. J. Pharmacol. Exp. Ther. 2010, 335, 830–840. [Google Scholar] [CrossRef]
- Quiros, S.; Roos, W.P.; Kaina, B. Rad51 and BRCA2 - New Molecular Targets for Sensitizing Glioma Cells to Alkylating Anticancer Drugs. PLoS ONE 2011, 6, e27183. [Google Scholar] [CrossRef] [Green Version]
- Qu, C.; Zhang, W.; Zheng, G.; Zhang, Z.; Yin, J.; He, Z. Metformin reverses multidrug resistance and epithelial-mesenchymal transition (EMT) via activating AMP-activated protein kinase (AMPK) in human breast cancer cells. Mol. Cell Biochem. 2014, 386, 63–71. [Google Scholar] [CrossRef]
- Zheng, G.; Peng, F.; Ding, R.; Yu, Y.; Ouyang, Y.; Chen, Z.; Xiao, Z.; He, Z. Identification of proteins responsible for the multiple drug resistance in 5-fluorouracil-induced breast cancer cell using proteomics analysis. J. Cancer Res. Clin. Oncol. 2010, 136, 1477–1488. [Google Scholar] [CrossRef]
- Zheng, G.; Xiong, Y.; Yi, S.; Zhang, W.; Peng, B.; Zhang, Q.; He, Z. 14-3-3σ regulation by p53 mediates a chemotherapy response to 5-fluorouracil in MCF-7 breast cancer cells via Akt inactivation. FEBS Lett. 2012, 586, 163–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, W.; Feng, M.; Zheng, G.; Chen, Y.; Wang, X.; Pen, B.; Yin, J.; Yu, Y.; He, Z. Chemoresistance to 5-fluorouracil induces epithelial-mesenchymal transition via up-regulation of Snail in MCF7 human breast cancer cells. Biochem. Biophys. Res. Commun. 2012, 417, 679–685. [Google Scholar] [CrossRef] [PubMed]
- Soo, J.S.; Ng, C.H.; Tan, S.H.; Malik, R.A.; Teh, Y.C.; Tan, B.S.; Ho, G.F.; See, M.H.; Taib, N.A.; Yip, C.H.; et al. Metformin synergizes 5-fluorouracil, epirubicin, and cyclophosphamide (FEC) combination therapy through impairing intracellular ATP production and DNA repair in breast cancer stem cells. Apoptosis 2015, 20, 1373–1387. [Google Scholar] [CrossRef] [PubMed]
- Lobo, N.A.; Shimono, Y.; Qian, D.; Clarke, M.F. The Biology of Cancer Stem Cells. Ann. Rev. Cell Dev. Biol. 2007, 23, 675–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pierce, G.B.; Wallace, C. Differentiation of Malignant to Benign Cells. Cancer Res. 1971, 31, e127. [Google Scholar]
- Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef]
- Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [Green Version]
- Hurt, E.M.; Kawasaki, B.T.; Klarmann, G.J.; Thomas, S.B.; Farrar, W.L. CD44+ CD24(-) prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis. Br. J. Cancer 2008, 98, 756–765. [Google Scholar] [CrossRef] [Green Version]
- Patrawala, L.; Calhoun, T.; Schneider-Broussard, R.; Li, H.; Bhatia, B.; Tang, S.; Reilly, J.G.; Chandra, D.; Zhou, J.; Claypool, K.; et al. Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene 2006, 25, 1696–1708. [Google Scholar] [CrossRef] [Green Version]
- Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of human brain tumour initiating cells. Nature 2004, 432, 396–401. [Google Scholar] [CrossRef]
- Ricci-Vitiani, L.; Lombardi, D.G.; Pilozzi, E.; Biffoni, M.; Todaro, M.; Peschle, C.; De Maria, R. Identification and expansion of human colon-cancer-initiating cells. Nature 2007, 445, 111–115. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Heidt, D.G.; Dalerba, P.; Burant, C.F.; Zhang, L.; Adsay, V.; Wicha, M.; Clarke, M.F.; Simeone, D.M. Identification of Pancreatic Cancer Stem Cells. Cancer Res. 2007, 67, e1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herschkowitz, J.I. Breast cancer stem cells: Initiating a new sort of thinking. Dis. Models Mech. 2010, 3, 257–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Velasco-Velázquez, M.A.; Homsi, N.; De La Fuente, M.; Pestell, R.G. Breast cancer stem cells. Int. J. Biochem. Cell Biol. 2012, 44, 573–577. [Google Scholar] [CrossRef] [Green Version]
- Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef] [Green Version]
- Morel, A.-P.; Lièvre, M.; Thomas, C.; Hinkal, G.; Ansieau, S.; Puisieux, A. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS ONE 2008, 3, e2888. [Google Scholar] [CrossRef]
- Mani, S.A.; Guo, W.; Liao, M.-J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [Green Version]
- Ponti, D.; Costa, A.; Zaffaroni, N.; Pratesi, G.; Petrangolini, G.; Coradini, D.; Pilotti, S.; Pierotti, M.A.; Daidone, M.G. Isolation and In vitro Propagation of Tumorigenic Breast Cancer Cells with Stem/Progenitor Cell Properties. Cancer Res. 2005, 65, e5506. [Google Scholar] [CrossRef] [Green Version]
- Visvader, J.E.; Lindeman, G.J. Cancer stem cells in solid tumours: Accumulating evidence and unresolved questions. Nat. Rev. Cancer 2008, 8, 755–768. [Google Scholar] [CrossRef]
- Alison, M.R.; Lim, S.M.L.; Nicholson, L.J. Cancer stem cells: Problems for therapy? J. Pathol. 2011, 223, 148–162. [Google Scholar] [CrossRef]
- Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef] [PubMed]
- Auffinger, B.; Tobias, A.L.; Han, Y.; Lee, G.; Guo, D.; Dey, M.; Lesniak, M.S.; Ahmed, A.U. Conversion of differentiated cancer cells into cancer stem-like cells in a glioblastoma model after primary chemotherapy. Cell Death Differ. 2014, 21, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
- Hamerlik, P.; Lathia, J.D.; Rasmussen, R.; Wu, Q.; Bartkova, J.; Lee, M.; Moudry, P.; Bartek, J., Jr.; Fischer, W.; Lukas, J.; et al. Autocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J. Exp. Med. 2012, 209, 507–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shien, K.; Toyooka, S.; Yamamoto, H.; Soh, J.; Jida, M.; Thu, K.L.; Hashida, S.; Maki, Y.; Ichihara, E.; Asano, H.; et al. Acquired resistance to EGFR inhibitors is associated with a manifestation of stem cell-like properties in cancer cells. Cancer Res. 2013, 73, 3051–3061. [Google Scholar] [CrossRef] [Green Version]
- Martins-Neves, S.R.; Cleton-Jansen, A.-M.; Gomes, C.M.F. Therapy-induced enrichment of cancer stem-like cells in solid human tumors: Where do we stand? Pharmacol. Res. 2018, 137, 193–204. [Google Scholar] [CrossRef]
- Gao, X.; Sishc, B.J.; Nelson, C.B.; Hahnfeldt, P.; Bailey, S.M.; Hlatky, L. Radiation-Induced Reprogramming of Pre-Senescent Mammary Epithelial Cells Enriches Putative CD44+/CD24−/low Stem Cell Phenotype. Front. Oncol. 2016, 6, e138. [Google Scholar] [CrossRef] [Green Version]
- Debeb, B.G.; Xu, W.; Woodward, W.A. Radiation Resistance of Breast Cancer Stem Cells: Understanding the Clinical Framework. J. Mammary Gland Biol. Neoplasia 2009, 14, 11–17. [Google Scholar] [CrossRef]
- Li, F.; Zhou, K.; Gao, L.; Zhang, B.; Li, W.; Yan, W.; Song, X.; Yu, H.; Wang, S.; Yu, N.; et al. Radiation induces the generation of cancer stem cells: A novel mechanism for cancer radioresistance. Oncol. Lett. 2016, 12, 3059–3065. [Google Scholar] [CrossRef] [Green Version]
- Zielske, S.P.; Spalding, A.C.; Wicha, M.S.; Lawrence, T.S. Ablation of breast cancer stem cells with radiation. Transl. Oncol. 2011, 4, 227–233. [Google Scholar] [CrossRef] [Green Version]
- Vidal, S.J.; Rodriguez-Bravo, V.; Galsky, M.; Cordon-Cardo, C.; Domingo-Domenech, J. Targeting cancer stem cells to suppress acquired chemotherapy resistance. Oncogene 2014, 33, 4451–4463. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Liao, R.; Li, D.; Sun, J. Induced cancer stem cells generated by radiochemotherapy and their therapeutic implications. Oncotarget 2017, 8, 17301–17312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mukherjee, P.; Gupta, A.; Chattopadhyay, D.; Chatterji, U. Modulation of SOX2 expression delineates an end-point for paclitaxel-effectiveness in breast cancer stem cells. Sci. Rep. 2017, 7, 9170. [Google Scholar] [CrossRef]
- Zhou, J.; Chen, Q.; Zou, Y.; Chen, H.; Qi, L.; Chen, Y. Stem Cells and Cellular Origins of Breast Cancer: Updates in the Rationale, Controversies, and Therapeutic Implications. Front. Oncol. 2019, 9, 820. [Google Scholar] [CrossRef] [PubMed]
- De Angelis, M.L.; Francescangeli, F.; La Torre, F.; Zeuner, A. Stem Cell Plasticity and Dormancy in the Development of Cancer Therapy Resistance. Front. Oncol. 2019, 9, 626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phi, L.T.H.; Sari, I.N.; Yang, Y.-G.; Lee, S.-H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer Stem Cells (CSCs) in Drug Resistance and their Therapeutic Implications in Cancer Treatment. Stem Cells Int. 2018, 2018, 5416923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fluegen, G.; Avivar-Valderas, A.; Wang, Y.; Padgen, M.R.; Williams, J.K.; Nobre, A.R.; Calvo, V.; Cheung, J.F.; Bravo-Cordero, J.J.; Entenberg, D.; et al. Phenotypic heterogeneity of disseminated tumour cells is preset by primary tumour hypoxic microenvironments. Nat. Cell Biol. 2017, 19, 120–132. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.; Lin, Q.; Glazer, P.M.; Yun, Z. The hypoxic tumor microenvironment in vivo selects the cancer stem cell fate of breast cancer cells. Breast Cancer Res. BCR 2018, 20, 16. [Google Scholar] [CrossRef] [Green Version]
- Harrison, H.; Rogerson, L.; Gregson, H.J.; Brennan, K.R.; Clarke, R.B.; Landberg, G. Contrasting Hypoxic Effects on Breast Cancer Stem Cell Hierarchy Is Dependent on ER-α Status. Cancer Res. 2013, 73, e1420. [Google Scholar] [CrossRef] [Green Version]
- Peppicelli, S.; Andreucci, E.; Ruzzolini, J.; Laurenzana, A.; Margheri, F.; Fibbi, G.; Del Rosso, M.; Bianchini, F.; Calorini, L. The acidic microenvironment as a possible niche of dormant tumor cells. Cell. Mol. Life Sci. 2017, 74, 2761–2771. [Google Scholar] [CrossRef]
- Linde, N.; Fluegen, G.; Aguirre-Ghiso, J.A. The Relationship Between Dormant Cancer Cells and Their Microenvironment. Adv. Cancer Res. 2016, 132, 45–71. [Google Scholar] [CrossRef] [Green Version]
- Ghiabi, P.; Jiang, J.; Pasquier, J.; Maleki, M.; Abu-Kaoud, N.; Rafii, S.; Rafii, A. Endothelial cells provide a notch-dependent pro-tumoral niche for enhancing breast cancer survival, stemness and pro-metastatic properties. PLoS ONE 2014, 9, e112424. [Google Scholar] [CrossRef] [PubMed]
- Comerford, K.M.; Wallace, T.J.; Karhausen, J.; Louis, N.A.; Montalto, M.C.; Colgan, S.P. Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res. 2002, 62, 3387–3394. [Google Scholar] [PubMed]
- Sansone, P.; Ceccarelli, C.; Berishaj, M.; Chang, Q.; Rajasekhar, V.K.; Perna, F.; Bowman, R.L.; Vidone, M.; Daly, L.; Nnoli, J.; et al. Self-renewal of CD133(hi) cells by IL6/Notch3 signalling regulates endocrine resistance in metastatic breast cancer. Nat. Commun. 2016, 7, 10442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, M.; Wicha, M.S. Metabolic plasticity of cancer stem cells. Oncotarget 2015, 6, 35141–35142. [Google Scholar] [CrossRef]
- Fiorillo, M.; Sotgia, F.; Lisanti, M.P. “Energetic” Cancer Stem Cells (e-CSCs): A New Hyper-Metabolic and Proliferative Tumor Cell Phenotype, Driven by Mitochondrial Energy. Front. Oncol. 2019, 8, e677. [Google Scholar] [CrossRef] [Green Version]
- Jiang, B. Aerobic glycolysis and high level of lactate in cancer metabolism and microenvironment. Genes Dis. 2017, 4, 25–27. [Google Scholar] [CrossRef]
- Thiery, J.P.; Acloque, H.; Huang, R.Y.J.; Nieto, M.A. Epithelial-Mesenchymal Transitions in Development and Disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
- Hong, D.; Fritz, A.J.; Zaidi, S.K.; van Wijnen, A.J.; Nickerson, J.A.; Imbalzano, A.N.; Lian, J.B.; Stein, J.L.; Stein, G.S. Epithelial-to-mesenchymal transition and cancer stem cells contribute to breast cancer heterogeneity. J. Cell Physiol. 2018, 233, 9136–9144. [Google Scholar] [CrossRef]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
- Zeisberg, M.; Neilson, E.G. Biomarkers for epithelial-mesenchymal transitions. J. Clin. Investig. 2009, 119, 1429–1437. [Google Scholar] [CrossRef] [Green Version]
- Loh, C.-Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, e1118. [Google Scholar] [CrossRef] [Green Version]
- Krawczyk, N.; Meier-Stiegen, F.; Banys, M.; Neubauer, H.; Ruckhaeberle, E.; Fehm, T. Expression of Stem Cell and Epithelial-Mesenchymal Transition Markers in Circulating Tumor Cells of Breast Cancer Patients. BioMed Res. Int. 2014, 2014, e415721. [Google Scholar] [CrossRef] [PubMed]
- Christowitz, C.; Davis, T.; Isaacs, A.; van Niekerk, G.; Hattingh, S.; Engelbrecht, A.-M. Mechanisms of doxorubicin-induced drug resistance and drug resistant tumour growth in a murine breast tumour model. BMC Cancer 2019, 19, e757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, W.-C.; Lai, Y.-A.; Lin, Y.-C.; Ma, J.-W.; Huang, L.-F.; Yang, N.-S.; Ho, C.-T.; Kuo, S.-C.; Way, T.-D. Curcumin Suppresses Doxorubicin-Induced Epithelial–Mesenchymal Transition via the Inhibition of TGF-β and PI3K/AKT Signaling Pathways in Triple-Negative Breast Cancer Cells. J. Agric. Food Chem. 2013, 61, 11817–11824. [Google Scholar] [CrossRef]
- Kim, R.-K.; Kaushik, N.; Suh, Y.; Yoo, K.-C.; Cui, Y.-H.; Kim, M.-J.; Lee, H.-J.; Kim, I.-G.; Lee, S.-J. Radiation driven epithelial-mesenchymal transition is mediated by Notch signaling in breast cancer. Oncotarget 2016, 7, 53430–53442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hugo, H.; Ackland, M.L.; Blick, T.; Lawrence, M.G.; Clements, J.A.; Williams, E.D.; Thompson, E.W. Epithelial—mesenchymal and mesenchymal—epithelial transitions in carcinoma progression. J. Cell Physiol. 2007, 213, 374–383. [Google Scholar] [CrossRef] [PubMed]
- Yao, D.; Dai, C.; Peng, S. Mechanism of the Mesenchymal–Epithelial Transition and Its Relationship with Metastatic Tumor Formation. Mol. Cancer Res. 2011, 9, e1608. [Google Scholar] [CrossRef] [Green Version]
- Luo, M.; Brooks, M.; Wicha, M.S. Epithelial-mesenchymal plasticity of breast cancer stem cells: Implications for metastasis and therapeutic resistance. Curr. Pharm. Des. 2015, 21, 1301–1310. [Google Scholar] [CrossRef] [Green Version]
- Thiery, J.P. Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2002, 2, 442–454. [Google Scholar] [CrossRef]
- Prieto-Vila, M.; Takahashi, R.-U.; Usuba, W.; Kohama, I.; Ochiya, T. Drug Resistance Driven by Cancer Stem Cells and Their Niche. Int. J. Mol. Sci. 2017, 18, e2574. [Google Scholar] [CrossRef] [Green Version]
- Sridharan, S.; Howard, C.M.; Tilley, A.M.C.; Subramaniyan, B.; Tiwari, A.K.; Ruch, R.J.; Raman, D. Novel and Alternative Targets Against Breast Cancer Stemness to Combat Chemoresistance. Front. Oncol. 2019, 9, e1003. [Google Scholar] [CrossRef] [PubMed]
- Leccia, F.; Del Vecchio, L.; Mariotti, E.; Di Noto, R.; Morel, A.-P.; Puisieux, A.; Salvatore, F.; Ansieau, S. ABCG2, a novel antigen to sort luminal progenitors of BRCA1- breast cancer cells. Mol. Cancer 2014, 13, 213. [Google Scholar] [CrossRef] [PubMed]
- Tiezzi, D.G.; Sicchieri, R.D.; Mouro, L.R.; Oliveira, T.M.G.; Silveira, W.A.; Antonio, H.M.R.; Muglia, V.F.; de Andrade, J.M. ABCG2 as a potential cancer stem cell marker in breast cancer. J. Clin. Oncol. 2013, 31, e12007. [Google Scholar] [CrossRef]
- Britton, K.M.; Eyre, R.; Harvey, I.J.; Stemke-Hale, K.; Browell, D.; Lennard, T.W.J.; Meeson, A.P. Breast cancer, side population cells and ABCG2 expression. Cancer Lett. 2012, 323, 97–105. [Google Scholar] [CrossRef] [Green Version]
- Arumugam, A.; Subramani, R.; Nandy, S.B.; Terreros, D.; Dwivedi, A.K.; Saltzstein, E.; Lakshmanaswamy, R. Silencing growth hormone receptor inhibits estrogen receptor negative breast cancer through ATP-binding cassette sub-family G member 2. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Balaji, S.A.; Udupa, N.; Chamallamudi, M.R.; Gupta, V.; Rangarajan, A. Role of the Drug Transporter ABCC3 in Breast Cancer Chemoresistance. PLoS ONE 2016, 11, e0155013. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Z.-S.; Sun, Y.-Z.; Wang, S.-M.; Ruan, J.-S. Epithelial-mesenchymal transition: Potential regulator of ABC transporters in tumor progression. J. Cancer 2017, 8, 2319–2327. [Google Scholar] [CrossRef] [Green Version]
- Dittmer, J. Breast cancer stem cells: Features, key drivers and treatment options. Semin. Cancer Biol. 2018, 53, 59–74. [Google Scholar] [CrossRef]
- Saxena, M.; Stephens, M.A.; Pathak, H.; Rangarajan, A. Transcription factors that mediate epithelial-mesenchymal transition lead to multidrug resistance by upregulating ABC transporters. Cell Death Dis. 2011, 2, e179. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Yu, F.; Jiao, Y.; Feng, J.; Tang, W.; Yao, H.; Gong, C.; Chen, J.; Su, F.; Zhang, Y.; et al. Reduced miR-128 in breast tumor-initiating cells induces chemotherapeutic resistance via Bmi-1 and ABCC5. Clin. Cancer Res. 2011, 17, 7105–7115. [Google Scholar] [CrossRef] [Green Version]
- Gao, M.; Miao, L.; Liu, M.; Li, C.; Yu, C.; Yan, H.; Yin, Y.; Wang, Y.; Qi, X.; Ren, J. miR-145 sensitizes breast cancer to doxorubicin by targeting multidrug resistance-associated protein-1. Oncotarget 2016, 7, 59714–59726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, Z.; Wu, H.; Xia, J.; Li, Y.; Zhang, Y.; Huang, K.; Wagar, N.; Yoon, Y.; Cho, H.T.; Scala, S.; et al. Involvement of miR-326 in chemotherapy resistance of breast cancer through modulating expression of multidrug resistance-associated protein 1. Biochem. Pharmacol. 2010, 79, 817–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turdo, A.; Veschi, V.; Gaggianesi, M.; Chinnici, A.; Bianca, P.; Todaro, M.; Stassi, G. Meeting the Challenge of Targeting Cancer Stem Cells. Front. Cell Dev. Biol. 2019, 7, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shafee, N.; Smith, C.R.; Wei, S.; Kim, Y.; Mills, G.B.; Hortobagyi, G.N.; Stanbridge, E.J.; Lee, E.Y.H.P. Cancer stem cells contribute to cisplatin resistance in Brca1/p53-mediated mouse mammary tumors. Cancer Res. 2008, 68, 3243–3250. [Google Scholar] [CrossRef] [Green Version]
- Del Vecchio, C.A.; Feng, Y.; Sokol, E.S.; Tillman, E.J.; Sanduja, S.; Reinhardt, F.; Gupta, P.B. De-differentiation confers multidrug resistance via noncanonical PERK-Nrf2 signaling. Plos. Biol. 2014, 12, e1001945. [Google Scholar] [CrossRef] [Green Version]
- Phillips, T.M.; McBride, W.H.; Pajonk, F. The response of CD24(-/low)/CD44+ breast cancer-initiating cells to radiation. J. Natl. Cancer Inst. 2006, 98, 1777–1785. [Google Scholar] [CrossRef] [Green Version]
- Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Dorie, M.J.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M.; et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458, 780–783. [Google Scholar] [CrossRef]
- Saini, N.; Yang, X. Metformin as an anti-cancer agent: Actions and mechanisms targeting cancer stem cells. Acta Biochim. Biophys. Sin. 2018, 50, 133–143. [Google Scholar] [CrossRef] [Green Version]
- Wang, N.; Wang, Z.; Peng, C.; You, J.; Shen, J.; Han, S.; Chen, J. Dietary compound isoliquiritigenin targets GRP78 to chemosensitize breast cancer stem cells via β-catenin/ABCG2 signaling. Carcinogenesis 2014, 35, 2544–2554. [Google Scholar] [CrossRef] [Green Version]
- Mao, J.; Song, B.; Shi, Y.; Wang, B.; Fan, S.; Yu, X.; Tang, J.; Li, L. ShRNA targeting Notch1 sensitizes breast cancer stem cell to paclitaxel. Int. J. Biochem. Cell Biol. 2013, 45, 1064–1073. [Google Scholar] [CrossRef]
- Jia, D.; Tan, Y.; Liu, H.; Ooi, S.; Li, L.; Wright, K.; Bennett, S.; Addison, C.L.; Wang, L. Cardamonin reduces chemotherapy-enriched breast cancer stem-like cells in vitro and in vivo. Oncotarget 2016, 7, 771–785. [Google Scholar] [CrossRef] [Green Version]
- Saha, S.; Mukherjee, S.; Khan, P.; Kajal, K.; Mazumdar, M.; Manna, A.; Mukherjee, S.; De, S.; Jana, D.; Sarkar, D.K.; et al. Aspirin Suppresses the Acquisition of Chemoresistance in Breast Cancer by Disrupting an NFκB–IL6 Signaling Axis Responsible for the Generation of Cancer Stem Cells. Cancer Res. 2016, 76, e2000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Woo, Y.; Oh, J.; Kim, J.-S. Suppression of Nrf2 Activity by Chestnut Leaf Extract Increases Chemosensitivity of Breast Cancer Stem Cells to Paclitaxel. Nutrients 2017, 9, e760. [Google Scholar] [CrossRef] [PubMed]
- Pandrangi, S.L.; Chikati, R.; Chauhan, P.S.; Kumar, C.S.; Banarji, A.; Saxena, S. Effects of ellipticine on ALDH1A1-expressing breast cancer stem cells—an in vitro and in silico study. Tumor Biol. 2014, 35, 723–737. [Google Scholar] [CrossRef] [PubMed]
- Samanta, D.; Park, Y.; Andrabi, S.A.; Shelton, L.M.; Gilkes, D.M.; Semenza, G.L. PHGDH Expression Is Required for Mitochondrial Redox Homeostasis, Breast Cancer Stem Cell Maintenance, and Lung Metastasis. Cancer Res. 2016, 76, e4430. [Google Scholar] [CrossRef] [Green Version]
- Sims-Mourtada, J.; Opdenaker, L.M.; Davis, J.; Arnold, K.M.; Flynn, D. Taxane-induced hedgehog signaling is linked to expansion of breast cancer stem-like populations after chemotherapy. Mol. Carcinog. 2015, 54, 1480–1493. [Google Scholar] [CrossRef]
- Wang, X.; Wang, X.; Gu, J.; Zhou, M.; He, Z.; Wang, X.; Ferrone, S. Overexpression of miR-489 enhances efficacy of 5-fluorouracil-based treatment in breast cancer stem cells by targeting XIAP. Oncotarget 2017, 8, 113837–113846. [Google Scholar] [CrossRef]
- Xie, Q.; Wang, S.; Zhao, Y.; Zhang, Z.; Qin, C.; Yang, X. MiR-519d impedes cisplatin-resistance in breast cancer stem cells by down-regulating the expression of MCL-1. Oncotarget 2017, 8, 22003–22013. [Google Scholar] [CrossRef] [Green Version]
- Korkaya, H.; Kim, G.-i.; Davis, A.; Malik, F.; Henry, N.L.; Ithimakin, S.; Quraishi, A.A.; Tawakkol, N.; D’Angelo, R.; Paulson, A.K.; et al. Activation of an IL6 Inflammatory Loop Mediates Trastuzumab Resistance in HER2+ Breast Cancer by Expanding the Cancer Stem Cell Population. Mol. Cell 2012, 47, 570–584. [Google Scholar] [CrossRef] [Green Version]
- Achuthan, S.; Santhoshkumar, T.R.; Prabhakar, J.; Nair, S.A.; Pillai, M.R. Drug-induced Senescence Generates Chemoresistant Stemlike Cells with Low Reactive Oxygen Species. J. Biol. Chem. 2011, 286, 37813–37829. [Google Scholar] [CrossRef] [Green Version]
- Sun, R.; Liu, Y.; Li, S.-Y.; Shen, S.; Du, X.-J.; Xu, C.-F.; Cao, Z.-T.; Bao, Y.; Zhu, Y.-H.; Li, Y.-P.; et al. Co-delivery of all-trans-retinoic acid and doxorubicin for cancer therapy with synergistic inhibition of cancer stem cells. Biomaterials 2015, 37, 405–414. [Google Scholar] [CrossRef]
- Sun, R.; Shen, S.; Zhang, Y.-J.; Xu, C.-F.; Cao, Z.-T.; Wen, L.-P.; Wang, J. Nanoparticle-facilitated autophagy inhibition promotes the efficacy of chemotherapeutics against breast cancer stem cells. Biomaterials 2016, 103, 44–55. [Google Scholar] [CrossRef] [PubMed]
- Li, S.-Y.; Sun, R.; Wang, H.-X.; Shen, S.; Liu, Y.; Du, X.-J.; Zhu, Y.-H.; Jun, W. Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells. J. Control. Release 2015, 205, 7–14. [Google Scholar] [CrossRef] [PubMed]
- Kolev, V.N.; Tam, W.F.; Wright, Q.G.; McDermott, S.P.; Vidal, C.M.; Shapiro, I.M.; Xu, Q.; Wicha, M.S.; Pachter, J.A.; Weaver, D.T. Inhibition of FAK kinase activity preferentially targets cancer stem cells. Oncotarget 2017, 8, 51733–51747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samanta, D.; Gilkes, D.M.; Chaturvedi, P.; Xiang, L.; Semenza, G.L. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, e5429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burnett, J.P.; Lim, G.; Li, Y.; Shah, R.B.; Lim, R.; Paholak, H.J.; McDermott, S.P.; Sun, L.; Tsume, Y.; Bai, S.; et al. Sulforaphane enhances the anticancer activity of taxanes against triple negative breast cancer by killing cancer stem cells. Cancer Lett. 2017, 394, 52–64. [Google Scholar] [CrossRef]
- Ginestier, C.; Liu, S.; Diebel, M.E.; Korkaya, H.; Luo, M.; Brown, M.; Wicinski, J.; Cabaud, O.; Charafe-Jauffret, E.; Birnbaum, D.; et al. CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts. J. Clin. Investig. 2010, 120, 485–497. [Google Scholar] [CrossRef]
- Kolev, V.N.; Wright, Q.G.; Vidal, C.M.; Ring, J.E.; Shapiro, I.M.; Ricono, J.; Weaver, D.T.; Padval, M.V.; Pachter, J.A.; Xu, Q. PI3K/mTOR Dual Inhibitor VS-5584 Preferentially Targets Cancer Stem Cells. Cancer Res. 2015, 75, e446. [Google Scholar] [CrossRef] [Green Version]
- Yu, Q.C.; Verheyen, E.M.; Zeng, Y.A. Mammary Development and Breast Cancer: A Wnt Perspective. Cancers 2016, 8, e65. [Google Scholar] [CrossRef] [Green Version]
- Yook, J.I.; Li, X.-Y.; Ota, I.; Hu, C.; Kim, H.S.; Kim, N.H.; Cha, S.Y.; Ryu, J.K.; Choi, Y.J.; Kim, J.; et al. A Wnt–Axin2–GSK3β cascade regulates Snail1 activity in breast cancer cells. Nat. Cell Biol. 2006, 8, 1398–1406. [Google Scholar] [CrossRef]
- Xu, J.; Prosperi, J.R.; Choudhury, N.; Olopade, O.I.; Goss, K.H. β-Catenin Is Required for the Tumorigenic Behavior of Triple-Negative Breast Cancer Cells. PLoS ONE 2015, 10, e0117097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harrison, H.; Farnie, G.; Howell, S.J.; Rock, R.E.; Stylianou, S.; Brennan, K.R.; Bundred, N.J.; Clarke, R.B. Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor. Cancer Res. 2010, 70, 709–718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, B.; Stephen, S.L.; Hanby, A.M.; Horgan, K.; Perry, S.L.; Richardson, J.; Roundhill, E.A.; Valleley, E.M.A.; Verghese, E.T.; Williams, B.J.; et al. Chemotherapy induces Notch1-dependent MRP1 up-regulation, inhibition of which sensitizes breast cancer cells to chemotherapy. BMC Cancer 2015, 15, e634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schott, A.F.; Landis, M.D.; Dontu, G.; Griffith, K.A.; Layman, R.M.; Krop, I.; Paskett, L.A.; Wong, H.; Dobrolecki, L.E.; Lewis, M.T.; et al. Preclinical and Clinical Studies of Gamma Secretase Inhibitors with Docetaxel on Human Breast Tumors. Clin. Cancer Res. 2013, 19, e1512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mamaeva, V.; Niemi, R.; Beck, M.; Özliseli, E.; Desai, D.; Landor, S.; Gronroos, T.; Kronqvist, P.; Pettersen, I.K.N.; McCormack, E.; et al. Inhibiting Notch Activity in Breast Cancer Stem Cells by Glucose Functionalized Nanoparticles Carrying γ-secretase Inhibitors. Mol. Ther. 2016, 24, 926–936. [Google Scholar] [CrossRef] [PubMed]
- Park, E.Y.; Chang, E.; Lee, E.J.; Lee, H.-W.; Kang, H.-G.; Chun, K.-H.; Woo, Y.M.; Kong, H.K.; Ko, J.Y.; Suzuki, H.; et al. Targeting of miR34a–NOTCH1 Axis Reduced Breast Cancer Stemness and Chemoresistance. Cancer Res. 2014, 74, e7573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Habib, J.G.; O’Shaughnessy, J.A. The hedgehog pathway in triple-negative breast cancer. Cancer Med. 2016, 5, 2989–3006. [Google Scholar] [CrossRef]
- Li, X.; Deng, W.; Nail, C.D.; Bailey, S.K.; Kraus, M.H.; Ruppert, J.M.; Lobo-Ruppert, S.M. Snail induction is an early response to Gli1 that determines the efficiency of epithelial transformation. Oncogene 2006, 25, 609–621. [Google Scholar] [CrossRef] [Green Version]
- Tao, Y.; Mao, J.; Zhang, Q.; Li, L. Overexpression of Hedgehog signaling molecules and its involvement in triple-negative breast cancer. Oncol. Lett. 2011, 2, 995–1001. [Google Scholar] [CrossRef] [Green Version]
- Das, S.; Samant, R.S.; Shevde, L.A. Nonclassical Activation of Hedgehog Signaling Enhances Multidrug Resistance and Makes Cancer Cells Refractory to Smoothened-targeting Hedgehog Inhibition. J. Biol. Chem. 2013, 288, 11824–11833. [Google Scholar] [CrossRef] [Green Version]
- Arnold, K.M.; Flynn, N.J.; Sims-Mourtada, J. Activation of Inflammatory Responses Correlate With Hedgehog Activation and Precede Expansion of Cancer Stem-Like Cells in an Animal Model of Residual Triple Negative Breast Cancer after Neoadjuvant Chemotherapy. Cancer Stud. Mol. Med. 2015, 2, 80–86. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Wulfkuhle, J.; Zhang, H.; Gu, P.; Yang, Y.; Deng, J.; Margolick, J.B.; Liotta, L.A.; Petricoin, E., 3rd; Zhang, Y. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc. Natl. Acad. Sci. USA 2007, 104, 16158–16163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karthik, G.-M.; Ma, R.; Lövrot, J.; Kis, L.L.; Lindh, C.; Blomquist, L.; Fredriksson, I.; Bergh, J.; Hartman, J. mTOR inhibitors counteract tamoxifen-induced activation of breast cancer stem cells. Cancer Lett. 2015, 367, 76–87. [Google Scholar] [CrossRef] [Green Version]
- Daverey, A.; Drain, A.P.; Kidambi, S. Physical Intimacy of Breast Cancer Cells with Mesenchymal Stem Cells Elicits Trastuzumab Resistance through Src Activation. Sci. Rep. 2015, 5, e13744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, M.; Sakamaki, T.; Casimiro, M.C.; Willmarth, N.E.; Quong, A.A.; Ju, X.; Ojeifo, J.; Jiao, X.; Yeow, W.-S.; Katiyar, S.; et al. The canonical NF-kappaB pathway governs mammary tumorigenesis in transgenic mice and tumor stem cell expansion. Cancer Res. 2010, 70, 10464–10473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vazquez-Santillan, K.; Melendez-Zajgla, J.; Jimenez-Hernandez, L.E.; Gaytan-Cervantes, J.; Muñoz-Galindo, L.; Piña-Sanchez, P.; Martinez-Ruiz, G.; Torres, J.; Garcia-Lopez, P.; Gonzalez-Torres, C.; et al. NF-kappaΒ-inducing kinase regulates stem cell phenotype in breast cancer. Sci. Rep. 2016, 6, 37340. [Google Scholar] [CrossRef] [Green Version]
- Smith, S.M.; Lyu, Y.L.; Cai, L. NF-kappaB affects proliferation and invasiveness of breast cancer cells by regulating CD44 expression. PLoS ONE 2014, 9, e106966. [Google Scholar] [CrossRef] [Green Version]
- Neuzillet, C.; Tijeras-Raballand, A.; Cohen, R.; Cros, J.; Faivre, S.; Raymond, E.; de Gramont, A. Targeting the TGFβ pathway for cancer therapy. Pharmacol. Ther. 2015, 147, 22–31. [Google Scholar] [CrossRef] [Green Version]
- Wahdan-Alaswad, R.S.; Thor, A.D. Metformin Activity against Breast Cancer: Mechanistic Differences by Molecular Subtype and Metabolic Conditions. In Metformin; Stoian, A.P., Rizzo, M., Eds.; IntechOpen: London, UK, 2020. [Google Scholar] [CrossRef] [Green Version]
- Cufi, S.; Corominas-Faja, B.; Vazquez-Martin, A.; Oliveras-Ferraros, C.; Dorca, J.; Bosch-Barrera, J.; Martin-Castillo, B.; Menendez, J.A. Metformin-induced preferential killing of breast cancer initiating CD44+CD24-/low cells is sufficient to overcome primary resistance to trastuzumab in HER2+ human breast cancer xenografts. Oncotarget 2012, 3, 395–398. [Google Scholar] [CrossRef] [Green Version]
- Hirsch, H.A.; Iliopoulos, D.; Tsichlis, P.N.; Struhl, K. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res. 2009, 69, 7507–7511. [Google Scholar] [CrossRef] [Green Version]
- Vazquez-Martin, A.; Oliveras-Ferraros, C.; Del Barco, S.; Martin-Castillo, B.; Menendez, J.A. The anti-diabetic drug metformin suppresses self-renewal and proliferation of trastuzumab-resistant tumor-initiating breast cancer stem cells. Breast Cancer Res. Treat. 2011, 126, 355–364. [Google Scholar] [CrossRef] [PubMed]
- Bao, B.; Azmi, A.S.; Ali, S.; Zaiem, F.; Sarkar, F.H. Metformin may function as anti-cancer agent via targeting cancer stem cells: The potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers. Ann. Transl. Med. 2014, 2, 59. [Google Scholar] [CrossRef] [PubMed]
- Zou, Y.F.; Xie, C.W.; Yang, S.X.; Xiong, J.P. AMPK activators suppress breast cancer cell growth by inhibiting DVL3-facilitated Wnt/β-catenin signaling pathway activity. Mol. Med. Rep. 2017, 15, 899–907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, X.; Chhipa, R.R.; Nakano, I.; Dasgupta, B. The AMPK Inhibitor Compound C Is a Potent AMPK-Independent Antiglioma Agent. Mol. Cancer Ther. 2014, 13, e596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samuel, S.M.; Satheesh, N.J.; Ghosh, S.; Büsselberg, D.; Majeed, Y.; Ding, H.; Triggle, C.R. Treatment with a Combination of Metformin and 2-Deoxyglucose Upregulates Thrombospondin-1 in Microvascular Endothelial Cells: Implications in Anti-Angiogenic Cancer Therapy. Cancers 2019, 11, 1737. [Google Scholar] [CrossRef] [Green Version]
- Fan, C.; Wang, Y.; Liu, Z.; Sun, Y.; Wang, X.; Wei, G.; Wei, J. Metformin exerts anticancer effects through the inhibition of the Sonic hedgehog signaling pathway in breast cancer. Int. J. Mol. Med. 2015, 36, 204–214. [Google Scholar] [CrossRef] [Green Version]
- Dunphy, K.A.; Seo, J.-H.; Kim, D.J.; Roberts, A.L.; Tao, L.; DiRenzo, J.; Balboni, A.L.; Crisi, G.M.; Hagen, M.J.; Chandrasekaran, T.; et al. Oncogenic transformation of mammary epithelial cells by transforming growth factor beta independent of mammary stem cell regulation. Cancer Cell Int. 2013, 13, e74. [Google Scholar] [CrossRef] [Green Version]
- Woosley, A.N.; Dalton, A.C.; Hussey, G.S.; Howley, B.V.; Mohanty, B.K.; Grelet, S.; Dincman, T.; Bloos, S.; Olsen, S.K.; Howe, P.H. TGFβ promotes breast cancer stem cell self-renewal through an ILEI/LIFR signaling axis. Oncogene 2019, 38, 3794–3811. [Google Scholar] [CrossRef]
- Konge, J.; Leteurtre, F.; Goislard, M.; Biard, D.; Morel-Altmeyer, S.; Vaurijoux, A.; Gruel, G.; Chevillard, S.; Lebeau, J. Breast cancer stem cell-like cells generated during TGFβ-induced EMT are radioresistant. Oncotarget 2018, 9, 23519–23531. [Google Scholar] [CrossRef] [Green Version]
- Karicheva, O.; Rodriguez-Vargas, J.M.; Wadier, N.; Martin-Hernandez, K.; Vauchelles, R.; Magroun, N.; Tissier, A.; Schreiber, V.; Dantzer, F. PARP3 controls TGFβ and ROS driven epithelial-to-mesenchymal transition and stemness by stimulating a TG2-Snail-E-cadherin axis. Oncotarget 2016, 7, 64109–64123. [Google Scholar] [CrossRef] [Green Version]
- Wahdan-Alaswad, R.; Harrell, J.C.; Fan, Z.; Edgerton, S.M.; Liu, B.; Thor, A.D. Metformin attenuates transforming growth factor beta (TGF-β) mediated oncogenesis in mesenchymal stem-like/claudin-low triple negative breast cancer. Cell Cycle 2016, 15, 1046–1059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leonel, C.; Borin, T.F.; de Carvalho Ferreira, L.; Moschetta, M.G.; Bajgelman, M.C.; Viloria-Petit, A.M.; de Campos Zuccari, D.A.P. Inhibition of Epithelial-Mesenchymal Transition and Metastasis by Combined TGFbeta Knockdown and Metformin Treatment in a Canine Mammary Cancer Xenograft Model. J. Mammary Gland Biol. Neoplasia 2017, 22, 27–41. [Google Scholar] [CrossRef] [PubMed]
- Vazquez-Martin, A.; Oliveras-Ferraros, C.; Cufí, S.; Del Barco, S.; Martin-Castillo, B.; Menendez, J.A. Metformin regulates breast cancer stem cell ontogeny by transcriptional regulation of the epithelial-mesenchymal transition (EMT) status. Cell Cycle 2010, 9, 3807–3814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, M.; Wang, Y.; Du, C.; Liu, Y.; Zhang, N.; Luo, F. Aspirin and metformin exhibit antitumor activity in murine breast cancer. Oncol. Rep. 2018, 39, 1414–1422. [Google Scholar] [CrossRef] [Green Version]
- Iliopoulos, D.; Hirsch, H.A.; Struhl, K. An epigenetic switch involving NF-kappaB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell 2009, 139, 693–706. [Google Scholar] [CrossRef] [Green Version]
- Hirsch, H.A.; Iliopoulos, D.; Struhl, K. Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Proc. Natl. Acad. Sci. USA 2013, 110, 972–977. [Google Scholar] [CrossRef] [Green Version]
- Zhao, W.; Zhang, X.; Liu, J.; Sun, B.; Tang, H.; Zhang, H. miR-27a-mediated antiproliferative effects of metformin on the breast cancer cell line MCF-7. Oncol. Rep. 2016, 36, 3691–3699. [Google Scholar] [CrossRef] [Green Version]
- Truong Do, M.; Gyun Kim, H.; Ho Choi, J.; Gwang Jeong, H. Metformin induces microRNA-34a to downregulate the Sirt1/Pgc-1α/Nrf2 pathway, leading to increased susceptibility of wild-type p53 cancer cells to oxidative stress and therapeutic agents. Free Radic. Biol. Med. 2014, 74, 21–34. [Google Scholar] [CrossRef]
- Oliveras-Ferraros, C.; Cufí, S.; Vazquez-Martin, A.; Torres-Garcia, V.Z.; Del Barco, S.; Martin-Castillo, B.; Menendez, J.A. Micro(mi)RNA expression profile of breast cancer epithelial cells treated with the anti-diabetic drug metformin: Induction of the tumor suppressor miRNA let-7a and suppression of the TGFβ-induced oncomiR miRNA-181a. Cell Cycle 2011, 10, 1144–1151. [Google Scholar] [CrossRef]
- Wahdan-Alaswad, R.S.; Cochrane, D.R.; Spoelstra, N.S.; Howe, E.N.; Edgerton, S.M.; Anderson, S.M.; Thor, A.D.; Richer, J.K. Metformin-induced killing of triple-negative breast cancer cells is mediated by reduction in fatty acid synthase via miRNA-193b. Horm Cancer 2014, 5, 374–389. [Google Scholar] [CrossRef] [Green Version]
- Feng, F.; Zhang, J.; Fan, X.; Yuan, F.; Jiang, Y.; Lv, R.; Ma, Y. Downregulation of Rab27A contributes to metformin-induced suppression of breast cancer stem cells. Oncol. Lett. 2017, 14, 2947–2953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Janzer, A.; German, N.J.; Gonzalez-Herrera, K.N.; Asara, J.M.; Haigis, M.C.; Struhl, K. Metformin and phenformin deplete tricarboxylic acid cycle and glycolytic intermediates during cell transformation and NTPs in cancer stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, 10574–10579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, S.L.; Kolozsvary, A.; Isrow, D.M.; Al Feghali, K.; Lapanowski, K.; Jenrow, K.A.; Kim, J.H. A Novel Mechanism of High Dose Radiation Sensitization by Metformin. Front. Oncol. 2019, 9, 247. [Google Scholar] [CrossRef] [Green Version]
- Chan, D.K.; Miskimins, W.K. Metformin and phenethyl isothiocyanate combined treatment in vitro is cytotoxic to ovarian cancer cultures. J. Ovarian Res. 2012, 5, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christensen, M.M.; Hojlund, K.; Hother-Nielsen, O.; Stage, T.B.; Damkier, P.; Beck-Nielsen, H.; Brosen, K. Steady-state pharmacokinetics of metformin is independent of the OCT1 genotype in healthy volunteers. Eur. J. Clin. Pharm. 2015, 71, 691–697. [Google Scholar] [CrossRef]
- Kinaan, M.; Ding, H.; Triggle, C.R. Metformin: An Old Drug for the Treatment of Diabetes but a New Drug for the Protection of the Endothelium. Med. Princ. Pract. 2015, 24, 401–415. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.; Park, H.J.; Park, C.-S.; Oh, E.-T.; Choi, B.-H.; Williams, B.; Lee, C.K.; Song, C.W. Response of Breast Cancer Cells and Cancer Stem Cells to Metformin and Hyperthermia Alone or Combined. PLoS ONE 2014, 9, e87979. [Google Scholar] [CrossRef] [PubMed]
- Shi, P.; Liu, W.; Tala; Wang, H.; Li, F.; Zhang, H.; Wu, Y.; Kong, Y.; Zhou, Z.; Wang, C.; et al. Metformin suppresses triple-negative breast cancer stem cells by targeting KLF5 for degradation. Cell Discov. 2017, 3, 17010. [Google Scholar] [CrossRef]
- Cuyàs, E.; Martin-Castillo, B.; Bosch-Barrera, J.; Menendez, J.A. Metformin inhibits RANKL and sensitizes cancer stem cells to denosumab. Cell Cycle 2017, 16, 1022–1028. [Google Scholar] [CrossRef] [Green Version]
- Tan, W.; Tang, H.; Jiang, X.; Ye, F.; Huang, L.; Shi, D.; Li, L.; Huang, X.; Li, L.; Xie, X.; et al. Metformin mediates induction of miR-708 to inhibit self-renewal and chemoresistance of breast cancer stem cells through targeting CD47. J. Cell. Mol. Med. 2019, 23, 5994–6004. [Google Scholar] [CrossRef]
- Song, C.W.; Lee, H.; Dings, R.P.M.; Williams, B.; Powers, J.; Santos, T.D.; Choi, B.-H.; Park, H.J. Metformin kills and radiosensitizes cancer cells and preferentially kills cancer stem cells. Sci. Rep. 2012, 2, 362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, T.H.; Suh, D.H.; Kim, M.-K.; Song, Y.S. Metformin against Cancer Stem Cells through the Modulation of Energy Metabolism: Special Considerations on Ovarian Cancer. BioMed Res. Int. 2014, 2014, e132702. [Google Scholar] [CrossRef] [PubMed]
- Iliopoulos, D.; Hirsch, H.A.; Struhl, K. Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types. Cancer Res. 2011, 71, 3196–3201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bednar, F.; Simeone, D.M. Metformin and cancer stem cells: Old drug, new targets. Cancer Prev. Res. 2012, 5, 351–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rattan, R.; Ali Fehmi, R.; Munkarah, A. Metformin: An emerging new therapeutic option for targeting cancer stem cells and metastasis. J. Oncol. 2012, 2012, 928127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aponte, P.M.; Caicedo, A. Stemness in Cancer: Stem Cells, Cancer Stem Cells, and Their Microenvironment. Stem Cells Int. 2017, 2017, e5619472. [Google Scholar] [CrossRef] [PubMed]
- Bozic, I.; Reiter, J.G.; Allen, B.; Antal, T.; Chatterjee, K.; Shah, P.; Moon, Y.S.; Yaqubie, A.; Kelly, N.; Le, D.T.; et al. Evolutionary dynamics of cancer in response to targeted combination therapy. eLife 2013, 2, e00747. [Google Scholar] [CrossRef]
- Banerjee, A.; Birts, C.N.; Darley, M.; Parker, R.; Mirnezami, A.H.; West, J.; Cutress, R.I.; Beers, S.A.; Rose-Zerilli, M.J.J.; Blaydes, J.P. Stem cell-like breast cancer cells with acquired resistance to metformin are sensitive to inhibitors of NADH-dependent CtBP dimerization. Carcinogenesis 2019, 40, 871–882. [Google Scholar] [CrossRef] [Green Version]
- Oliveras-Ferraros, C.; Vazquez-Martin, A.; Cuyàs, E.; Corominas-Faja, B.; Rodríguez-Gallego, E.; Fernández-Arroyo, S.; Martin-Castillo, B.; Joven, J.; Menendez Menendez, J. Acquired resistance to metformin in breast cancer cells triggers transcriptome reprogramming toward a degradome-related metastatic stem-like profile. Cell Cycle 2014, 13, 1132–1144. [Google Scholar] [CrossRef] [Green Version]
- Scherbakov, A.M.; Sorokin, D.V.; Tatarskiy Jr, V.V.; Prokhorov, N.S.; Semina, S.E.; Berstein, L.M.; Krasil’nikov, M.A. The phenomenon of acquired resistance to metformin in breast cancer cells: The interaction of growth pathways and estrogen receptor signaling. IUBMB Life 2016, 68, 281–292. [Google Scholar] [CrossRef]
- Menendez, J.A.; Oliveras-Ferraros, C.; Cufi, S.; Corominas-Faja, B.; Joven, J.; Martin-Castillo, B.; Vazquez-Martin, A. Metformin is synthetically lethal with glucose withdrawal in cancer cells. Cell Cycle 2012, 11, 2782–2792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zordoky, B.N.; Bark, D.; Soltys, C.L.; Sung, M.M.; Dyck, J.R. The anti-proliferative effect of metformin in triple-negative MDA-MB-231 breast cancer cells is highly dependent on glucose concentration: Implications for cancer therapy and prevention. Biochim. Biophys. Acta 2014, 1840, 1943–1957. [Google Scholar] [CrossRef] [PubMed]
- Snima, K.S.; Jayakumar, R.; Unnikrishnan, A.G.; Nair, S.V.; Lakshmanan, V.-K. O-Carboxymethyl chitosan nanoparticles for metformin delivery to pancreatic cancer cells. Carbohydr. Polym. 2012, 89, 1003–1007. [Google Scholar] [CrossRef]
- Graham, G.G.; Punt, J.; Arora, M.; Day, R.O.; Doogue, M.P.; Duong, J.; Furlong, T.J.; Greenfield, J.R.; Greenup, L.C.; Kirkpatrick, C.M.; et al. Clinical Pharmacokinetics of Metformin. Clin. Pharmacokinet. 2011, 50, 81–98. [Google Scholar] [CrossRef] [PubMed]
- Cetin, M.; Sahin, S. Microparticulate and nanoparticulate drug delivery systems for metformin hydrochloride. Drug Deliv. 2016, 23, 2796–2805. [Google Scholar] [CrossRef]
- Javidfar, S.; Pilehvar-Soltanahmadi, Y.; Farajzadeh, R.; Lotfi-Attari, J.; Shafiei-Irannejad, V.; Hashemi, M.; Zarghami, N. The inhibitory effects of nano-encapsulated metformin on growth and hTERT expression in breast cancer cells. J. Drug Deliv. Sci. Technol. 2018, 43, 19–26. [Google Scholar] [CrossRef]
- Baldassari, S.; Solari, A.; Zuccari, G.; Drava, G.; Pastorino, S.; Fucile, C.; Marini, V.; Daga, A.; Pattarozzi, A.; Ratto, A.; et al. Development of an Injectable Slow-Release Metformin Formulation and Evaluation of Its Potential Antitumor Effects. Sci. Rep. 2018, 8, 3929. [Google Scholar] [CrossRef]
- De Jong, W.H.; Borm, P.J.A. Drug delivery and nanoparticles:applications and hazards. Int. J. Nanomed. 2008, 3, 133–149. [Google Scholar] [CrossRef] [Green Version]
- Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Dié, V.; Hegg, R.; Im, S.-A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef]
- Kuwayama, T.; Nakamura, S.; Hayashi, N.; Takano, T.; Tsugawa, K.; Sato, T.; Kitani, A.; Okuyama, H.; Yamauchi, H. Randomized Multicenter Phase II Trial of Neoadjuvant Therapy Comparing Weekly Nab-paclitaxel Followed by FEC With Docetaxel Followed by FEC in HER2− Early-stage Breast Cancer. Clin. Breast Cancer 2018, 18, 474–480. [Google Scholar] [CrossRef]
- Farajzadeh, R.; Pilehvar-Soltanahmadi, Y.; Dadashpour, M.; Javidfar, S.; Lotfi-Attari, J.; Sadeghzadeh, H.; Shafiei-Irannejad, V.; Zarghami, N. Nano-encapsulated metformin-curcumin in PLGA/PEG inhibits synergistically growth and hTERT gene expression in human breast cancer cells. Artif. Cells Nanomed. Biotechnol. 2018, 46, 917–925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shukla, S.K.; Kulkarni, N.S.; Chan, A.; Parvathaneni, V.; Farrales, P.; Muth, A.; Gupta, V. Metformin-Encapsulated Liposome Delivery System: An Effective Treatment Approach against Breast Cancer. Pharmaceutics 2019, 11, e559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aljofan, M.; Riethmacher, D. Anticancer activity of metformin: A systematic review of the literature. Future Sci. OA 2019, 5, FSO410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DeVito, N.C.; Goodman, M.D.; Caplain, J.; Rajagopal, S.; Popowich, D.; Orkin, B.A.; Grimm, E.; Tsichlis, P.N.; Martell, R.E.; Saif, W.M. A pilot study evaluating the safety and impact of pretreatment with metformin on colorectal cancer stem cells (CCSC) in patients undergoing resection. J. Clin. Oncol. 2014, 32, e14581. [Google Scholar] [CrossRef]
- Brown, J.R.; Chan, D.K.; Shank, J.J.; Griffith, K.A.; Fan, H.; Szulawski, R.; Yang, K.; Reynolds, R.K.; Johnston, C.; McLean, K.; et al. Phase II clinical trial of metformin as a cancer stem cell-targeting agent in ovarian cancer. JCI Insight 2020, 5, e133247. [Google Scholar] [CrossRef]
Cell Lines | Drug Intervention | BCSC Marker Status | Pathways/Factors Involved in Drug Resistance | Reference | |
---|---|---|---|---|---|
CD44+/CD24neg/low | ALDH+ | ||||
MCF7 | PTX, DOX, Epi, 5-FU, FAC, Mito | ↑ | --- | Notch, NRF2, NF-κB/IL6 | [192,203,204,205,206,207] |
PTX, DOX | --- | ↑ | ALDH, PHGDH | [192,208,209] | |
T47D | DOX, Cis, 5-FU, FAC | ↑ | --- | Hh, MCL1, XIAP, NF-κB/IL6 | [192,206,210,211,212] |
BT474 | FAC, Trastuzumab | ↑ | --- | NF-κB/IL6, IL6R | [192,206,213] |
SUM190 | PTX, DOX, 5-FU | ↑ | --- | NF-κB/STAT3 | [192,205] |
SKBR3 | DTX, 5-FU | ↑ | --- | Hh, XIAP | [192,210,211] |
MDA-MB-231 | DOX, Epi, Noco, FAC | ↑ | --- | GRP78, NF-κB/IL6 | [192,203,206,214] |
PTX, DTX, DOX | --- | ↑ | FAK, autophagy, DNMT, PHGDH, HIF1α, HIF2 α | [156,192,215,216,217,218,219] | |
SUM159 | --- | ↑ | ALDH, PI3K/Akt/mTOR, NF-κB, Hh, CXCR1, HIF1α | [192,219,220,221,222] |
Serial No: | Type of Cancer | Total Number of Registered Trials | Completed | Active, Not Recruiting | Active, Recruiting/Not Yet Recruiting | Terminated | Withdrawn/Suspended | Unknown Status# | Has Results * |
---|---|---|---|---|---|---|---|---|---|
1 | All Cancers | 353 | 142 | 29 | 79/14 | 34 | 12 / 7 | 38 | 41 |
2 | Lung Cancer | 20 | 5 | 1 | 6/1 | 6 | -- | 1 | 5 |
3 | Breast Cancers | 48 | 17 | 4 | 14/2 | 5 | 1 | 5 | 6 |
4 | Colorectal Cancer | 18 | 5 | -- | 7 | 6 | -- | -- | 3 |
5 | Prostate Cancer | 27 | 7 | 3 | 8/1 | 2 | 4 | 2 | 4 |
6 | Liver Cancer | 9 | 2 | -- | 2/1 | 3 | -- | 1 | -- |
7 | Bladder Cancer | 6 | 1 | -- | 4 | -- | -- | 1 | -- |
8 | Pancreatic Cancer | 21 | 9 | 1 | 4/3 | 1 | 1 | 2 | -- |
9 | Ovarian Cancer | 8 | 2 | 1 | 3/1 | -- | 1 | -- | 1 |
10 | Endometrial Cancer | 15 | 6 | 5 | 2 | -- | 1 | 1 | -- |
11 | Head and Neck Cancers | 10 | 2 | 2 | 3/1 | 2 | -- | -- | 2 |
12 | Oral Cancers | 8 | -- | 2 | 4 | 1 | -- | 1 | 2 |
Serial No: | Metformin + Drug */Radiation | Total Number of Registered Trials | Completed | Active, Not Recruiting | Active, Recruiting | Terminated | Withdrawn/Suspended | Unknown Status # |
---|---|---|---|---|---|---|---|---|
1 | 5-fluorouracil (5-FU) | 1 | -- | -- | 1 | -- | -- | -- |
2 | Carboplatin | 1 | -- | 0 | 1 | -- | -- | -- |
3 | Cyclophosphamide | 5 | 1 | 1 | 3 | -- | -- | -- |
4 | Docetaxel | 3 | -- | 1 | 2 | -- | -- | -- |
5 | Doxorubicin/adriamycin | 6 | 1 | 2 | 2 | 1 | -- | -- |
6 | Epirubicin | 1 | -- | -- | 1 | -- | -- | -- |
7 | Everolimus/Temsirolimus | 2 | 1 | -- | -- | 1 | -- | -- |
8 | Letrozole | 2 | 1 | -- | 1 | -- | -- | -- |
9 | Paclitaxel | 5 | -- | 2 | 1 | 1 | -- | 1 |
10 | Radiation/Radiotherapy | 4 | 1 | -- | 1 | 1 | -- | 1 |
Serial No: | Official Title | Trial Phase | Intervention Using Metformin | Objectives | Type of Cancer | Clinicaltrials.Gov ID (NCT Number)/Status, Actual Month and Year Related to the Status | Publications/References |
---|---|---|---|---|---|---|---|
1 | Impact of Pretreatment with Metformin on Colorectal Cancer Stem Cells (CCSC) and Related Pharmacodynamic Parameters | Phase I | Metformin | Primary outcome measures: Expression of CD133 in tumors from patients treated with metformin in comparison to patients not treated with metformin | Colon Cancer | NCT01440127/Terminated October 2012 | [299] (Abstract only) |
2 | A Phase II Evaluation of Metformin, Targeting Cancer Stem Cells for Prevention of Relapse in Patients with Stage IIC/III/IV Ovarian, Fallopian Tube, and Primary Peritoneal Cancer | Phase II | Metformin | Primary outcome measures: Recurrence-Free Survival Secondary outcome measures: Overall Survival | Ovarian, Fallopian Tube, and Primary Peritoneal Cancer | NCT01579812/Completed July 2017 | [300] |
3 | A Pharmacodynamic Study of Metformin in Patients with Resectable Pancreatic Cancer | Phase I | Metformin hydrochloride | Primary outcome measures: Pancreatic tumor cell proliferation and apoptosis as measured by the percentage of Ki67 positive, percentage of TUNEL positive and mitotic counts in tissue samples. Secondary outcome measures:
| Stage IA, IB, IIA, and IIB Pancreatic Cancer | NCT01954732/Completed March 2015 | No results posted |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Samuel, S.M.; Varghese, E.; Koklesová, L.; Líšková, A.; Kubatka, P.; Büsselberg, D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers 2020, 12, 2482. https://doi.org/10.3390/cancers12092482
Samuel SM, Varghese E, Koklesová L, Líšková A, Kubatka P, Büsselberg D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers. 2020; 12(9):2482. https://doi.org/10.3390/cancers12092482
Chicago/Turabian StyleSamuel, Samson Mathews, Elizabeth Varghese, Lenka Koklesová, Alena Líšková, Peter Kubatka, and Dietrich Büsselberg. 2020. "Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells" Cancers 12, no. 9: 2482. https://doi.org/10.3390/cancers12092482
APA StyleSamuel, S. M., Varghese, E., Koklesová, L., Líšková, A., Kubatka, P., & Büsselberg, D. (2020). Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers, 12(9), 2482. https://doi.org/10.3390/cancers12092482