Metformin targets the metabolic achilles heel of human pancreatic cancer stem cells

Enza Lonardo, Michele Cioffi, Patricia Sancho, Yolanda Sanchez-Ripoll, Sara Maria Trabulo, Jorge Dorado, Anamaria Balic, Manuel Hidalgo, Christopher Heeschen, Enza Lonardo, Michele Cioffi, Patricia Sancho, Yolanda Sanchez-Ripoll, Sara Maria Trabulo, Jorge Dorado, Anamaria Balic, Manuel Hidalgo, Christopher Heeschen

Abstract

Pancreatic ductal adenocarcinomas contain a subset of exclusively tumorigenic cancer stem cells (CSCs), which are capable of repopulating the entire heterogeneous cancer cell populations and are highly resistant to standard chemotherapy. Here we demonstrate that metformin selectively ablated pancreatic CSCs as evidenced by diminished expression of pluripotency-associated genes and CSC-associated surface markers. Subsequently, the ability of metformin-treated CSCs to clonally expand in vitro was irreversibly abrogated by inducing apoptosis. In contrast, non-CSCs preferentially responded by cell cycle arrest, but were not eliminated by metformin treatment. Mechanistically, metformin increased reactive oxygen species production in CSC and reduced their mitochondrial transmembrane potential. The subsequent induction of lethal energy crisis in CSCs was independent of AMPK/mTOR. Finally, in primary cancer tissue xenograft models metformin effectively reduced tumor burden and prevented disease progression; if combined with a stroma-targeting smoothened inhibitor for enhanced tissue penetration, while gemcitabine actually appeared dispensable.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Metformin targets pancreatic cancer stem…
Figure 1. Metformin targets pancreatic cancer stem cells.
(A) Primary PDAC cells, but not normal pancreas cells express organic cation transporter 1, 2, and 3 (n = 3). (B) Definition of the therapeutic range for metformin in primary PDAC cells. Number of cells grown in the presence of the indicated concentrations of metformin for 24 h (n = 6). (C) qPCR analysis of CSCs-associated genes in spheres treated with 3 mM of metformin for 7 days. Data are normalized to the housekeeping gene and are presented as fold change in gene expression relative to control cells (n = 6). (D) Representative Western blot illustrating reduced Oct4 protein expression in response to metformin treatment (n = 3). (E) Representative flow cytometry analysis for CSCs markers in spheres treated for 7 days with 3 mM of metformin as compared to untreated spheres (upper panel). Summary of data for PDAC-185, A6L, 215, 253, and 354 is shown (lower panel; n = 6).
Figure 2. Metformin diminishes in vitro and…
Figure 2. Metformin diminishes in vitro and in vivo tumorigenicity.
(A) Metformin decreases the size of spheres. Representative images of spheres obtained after treatment with metformin for 7 days. Quantification of sphere size (n≥6). (B) Sphere formation capacity in the presence or absence of metformin for 7 days (n≥6). (C) Self-renewal capacity of cancer stem cells isolated from tumors responding poorly in terms of first passage sphere forming capacity. Cells were continuously passage as secondary and tertiary spheres treated with metformin or vehicle only during first generation sphere formation (n = 6). (D) Colony formation for PDAC-185, A6L, 215, 253, and 354 evidenced by 0.05% crystal violet after 21 days (n = 3). (E) Rarefication of in vivo tumorigenic cancer stem cells in spheres treated with metformin as compared to vehicle. (F) Invasion of sphere-derived cells after 24 h of treatment with metformin or control (n = 3).
Figure 3. Metformin specifically eliminates cancer stem…
Figure 3. Metformin specifically eliminates cancer stem cells.
(A) Number of cells grown in the presence of the indicated concentrations of metformin for 24 h. (n = 3). (B) Quantification for Ki67 and DAPI in adherent cells after 7 d of treatment with metformin or control (n = 3). (C) qPCR analysis for CyclinD1 in adherent and sphere-derived cells after 7 d of treatment with metformin or control. Data are normalized to the housekeeping gene and are presented as fold change in gene expression relative to untreated cells (n = 6). (D) Cell cycle analysis determined by Propidium Iodide staining in adherent cells and spheres after 7 d of treatment with metformin or control (n = 3). (E) Cytometry analysis of apoptotic cells by double staining for Annexin V/DAPI after treatment with metformin or control for adherent versus sphere-derived cells (n = 3).
Figure 4. Mechanism of action.
Figure 4. Mechanism of action.
(A) Cellular ATP levels in adherent cells and spheres after 7 d of treatment with metformin or control (n = 3). (B) Upper panel: Western blot analysis of pAMPK, AMPK, and GAPDH in spheres treated with metformin or control. Lower panel: Western blot analysis of pAMPK, pp70S6K, and GAPDH in adherent cells and spheres treated with metformin or control for 7 days (n = 3). (C) Overall effect of metformin (3 mM), inhibition of mTOR (rapamycin; 100 ng/ml), and direction activation of AMPK (A769662; 10 µM) on sphere formation and (n = 6). (D) colony formation for PDAC-215, 253, and 354 cells (n = 3). (E) Total and mitochondrial (Mito) ROS production after 8 hours of control or metformin treatment. (F) Mitochondrial transmembrane potential after 8 hours of control or metformin treatment.
Figure 5. Metformin stalls PDAC progression in…
Figure 5. Metformin stalls PDAC progression in vivo.
(A) Left panel: PDAC-215 tissue was implanted and treatment was allocated after initial tumor take was verified. Mice were treated with either gemcitabine (Gem) or metformin (Met). Metformin was discontinued on d100 to test the potential of the remaining lesions to initiate disease relapse. After documented disease relapse, metformin treatment was re-administered. Right panel: Body weight during treatment. (B) Left panel: Histological analysis: Hematoxylin & Eosin (H&E), CyclinD1, and Caspase3. Right panel: Content for CD44+ cells. (C) Effects of in vitro treatment as indicated on sphere formation capacity. (D) PDAC-A6L tissue implanted in mice and treated as indicated. (E) Content for CD44+ cells (upper panel) and sphere formation capacity (lower panel). (F) Histological analysis: H&E and cytokeratin19 (CK19; n = 6).
Figure 6. Additional targeting of the stroma…
Figure 6. Additional targeting of the stroma prevents tumor relapse.
(A) PDAC-185 tissue implanted in mice and treated as indicated including the smoothened inhibitor SIBI-C1. (B) Content for EpCAM+ and CD133+ cells, respectively, (upper panel) and sphere formation capacity (lower panel). (C) Histological analysis: Hematoxylin&Eosin (H&E) and cytokeratin19 (CK19). (D) mTOR inhibitor resistant PDAC-253 tissue implanted in mice and treated as indicated (n = 6).

References

    1. Jemal A, Siegel R, Xu J, Ward E (2010) Cancer statistics, 2010. CA Cancer J Clin 60: 277–300.
    1. Lowenfels AB, Maisonneuve P (2006) Epidemiology and risk factors for pancreatic cancer. Best Pract Res Clin Gastroenterol 20: 197–209.
    1. Li D (2012) Diabetes and pancreatic cancer. Mol Carcinog 51: 64–74.
    1. White E (2012) Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer 12: 401–410.
    1. Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD (2005) Metformin and reduced risk of cancer in diabetic patients. BMJ 330: 1304–1305.
    1. Sadeghi N, Abbruzzese JL, Yeung SC, Hassan M, Li D (2012) Metformin Use Is Associated with Better Survival of Diabetic Patients with Pancreatic Cancer. Clin Cancer Res
    1. Michaud DS (2004) Epidemiology of pancreatic cancer. Minerva Chir 59: 99–111.
    1. Shaw RJ, Lamia KA, Vasquez D, Koo SH, Bardeesy N, et al. (2005) The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 310: 1642–1646.
    1. Meric-Bernstam F, Gonzalez-Angulo AM (2009) Targeting the mTOR signaling network for cancer therapy. J Clin Oncol 27: 2278–2287.
    1. Mueller MT, Hermann PC, Witthauer J, Rubio-Viqueira B, Leicht SF, et al. (2009) Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology 137: 1102–1113.
    1. Anisimov VN, Berstein LM, Egormin PA, Piskunova TS, Popovich IG, et al. (2005) Effect of metformin on life span and on the development of spontaneous mammary tumors in HER-2/neu transgenic mice. Exp Gerontol 40: 685–693.
    1. Martin-Castillo B, Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA (2010) Metformin and cancer: doses, mechanisms and the dandelion and hormetic phenomena. Cell Cycle 9: 1057–1064.
    1. Kisfalvi K, Eibl G, Sinnett-Smith J, Rozengurt E (2009) Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res 69: 6539–6545.
    1. Bao B, Wang Z, Ali S, Ahmad A, Azmi AS, et al. (2011) Metformin inhibits cell proliferation, migration and invasion by attenuating CSC function mediated by deregulating miRNAs in pancreatic cancer cells. Cancer Prev Res (Phila) 5: 355–364.
    1. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, et al. (2007) Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1: 313–323.
    1. Jones D, Wagers A (2008) No place like home: anatomy and function of the stem cell niche. Nat Rev Mol Cell Biol 9: 11–21.
    1. Rubio-Viqueira B, Jimeno A, Cusatis G, Zhang X, Iacobuzio-Donahue C, et al. (2006) An in vivo platform for translational drug development in pancreatic cancer. Clin Cancer Res 12: 4652–4661.
    1. Lonardo E, Hermann PC, Mueller M-T, Huber S, Balic A, et al. (2011) Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy. Cell Stem Cell
    1. Ouslimani N, Peynet J, Bonnefont-Rousselot D, Therond P, Legrand A, et al. (2005) Metformin decreases intracellular production of reactive oxygen species in aortic endothelial cells. Metabolism 54: 829–834.
    1. Vander Heiden MG, Cantley LC, Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324: 1029–1033.
    1. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, et al. (2009) Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324: 1457–1461.
    1. Lonardo E, Frias-Aldeguer J, Hermann PC, Heeschen C (2012) Pancreatic stellate cells form a niche for cancer stem cells and promote their self-renewal and invasiveness. Cell Cycle 11: 1282–1290.
    1. Garrido-Laguna I, Tan AC, Uson M, Angenendt M, Ma WW, et al. (2010) Integrated preclinical and clinical development of mTOR inhibitors in pancreatic cancer. Br J Cancer 103: 649–655.
    1. Hess G, Herbrecht R, Romaguera J, Verhoef G, Crump M, et al. (2009) Phase III study to evaluate temsirolimus compared with investigator's choice therapy for the treatment of relapsed or refractory mantle cell lymphoma. J Clin Oncol 27: 3822–3829.
    1. Rehman J (2010) Empowering self-renewal and differentiation: the role of mitochondria in stem cells. J Mol Med (Berl) 88: 981–986.
    1. Owen MR, Doran E, Halestrap AP (2000) Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J 348 Pt 3: 607–614.
    1. Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K (2009) Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res 69: 7507–7511.
    1. Hidalgo M (2010) Pancreatic cancer. N Engl J Med 362: 1605–1617.
    1. Hamada S, Masamune A, Takikawa T, Suzuki N, Kikuta K, et al. (2012) Pancreatic stellate cells enhance stem cell-like phenotypes in pancreatic cancer cells. Biochem Biophys Res Commun 421: 349–354.
    1. Jimeno A, Feldmann G, Suarez-Gauthier A, Rasheed Z, Solomon A, et al. (2009) A direct pancreatic cancer xenograft model as a platform for cancer stem cell therapeutic development. Mol Cancer Ther 8: 310–314.
    1. Gallmeier E, Hermann PC, Mueller MT, Machado JG, Ziesch A, et al. (2011) Inhibition of ATR Function Abrogates the in Vitro and in Vivo Tumorigenicity of Human Colon Cancer Cells Through Depletion of the CD133+ Tumor-Initiating Cell Fraction. Stem Cells 29: 418–429.
    1. Lonardo E, Hermann PC, Mueller MT, Huber S, Balic A, et al. (2011) Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy. Cell Stem Cell 9: 433–446.

Source: PubMed

3
Sottoscrivi