Metformin kills and radiosensitizes cancer cells and preferentially kills cancer stem cells

Chang W Song, Hyemi Lee, Ruud P M Dings, Brent Williams, John Powers, Troy Dos Santos, Bo-Hwa Choi, Heon Joo Park, Chang W Song, Hyemi Lee, Ruud P M Dings, Brent Williams, John Powers, Troy Dos Santos, Bo-Hwa Choi, Heon Joo Park

Abstract

The anti-cancer effects of metformin, the most widely used drug for type 2 diabetes, alone or in combination with ionizing radiation were studied with MCF-7 human breast cancer cells and FSaII mouse fibrosarcoma cells. Clinically achievable concentrations of metformin caused significant clonogenic death in cancer cells. Importantly, metformin was preferentially cytotoxic to cancer stem cells relative to non-cancer stem cells. Metformin increased the radiosensitivity of cancer cells in vitro, and significantly enhanced the radiation-induced growth delay of FSaII tumors (s.c.) in the legs of C3H mice. Both metformin and ionizing radiation activated AMPK leading to inactivation of mTOR and suppression of its downstream effectors such as S6K1 and 4EBP1, a crucial signaling pathway for proliferation and survival of cancer cells, in vitro as well as in the in vivo tumors.

Conclusion: Metformin kills and radiosensitizes cancer cells and eradicates radioresistant cancer stem cells by activating AMPK and suppressing mTOR.

Figures

Figure 1. Clonogenic death of FSaII and…
Figure 1. Clonogenic death of FSaII and MCF-7 cells treated with metformin alone or with radiation.
(A, B) Cells were incubated for 1–48 h with 0.5–10 mM metformin and the clonogenic survival was determined. (C) Cells were cultured in media containing 5–100 µM of metformin for 10 days for FSaII cells and 20 days for MCF-7 days. (D, E) Cells were incubated with 1 or 5 mM metformin for 24 h, irradiated with various doses of X-rays, further incubated with metformin for 24 h and then the clonogenic survival was determined. The radiation survival curves were normalized for the cell death caused by metformin alone. All data points are means of 5–7 independent experiments ± 1 S.E.
Figure 2. Western blotting for the effect…
Figure 2. Western blotting for the effect of metformin, 4 Gy irradiation or combined on the signals associated with AMPK/mTOR pathway in MCF-7 and FSaII cells.
Whole cell lysates were prepared and Western blot analysis was conducted using anti-p-AMPK, -AMPK, -p-ACC, -ACC, -p-mTOR,-mTOR, -p-S6K1, -S6K1, -p-4EBP1 and -4EBP1 antibodies. Experiments were repeated 4-5 times and the representative results are shown. The numbers under each blot are intensity of the blot relative to that of untreated control.
Figure 3. Effects of AMPK siRNA on…
Figure 3. Effects of AMPK siRNA on the cytotoxic and radiosensitizing effects of metformin.
(A) Transfection of MCF-7 and FSaII ells with siRNA against AMPK markedly reduced the AMPK levels in the cells. (B) Transfection of cells with AMPK siRNA markedly reduced the clonogenic cell death caused by 48 h incubation with various concentrations of metformin. (C) Transfection of cells with AMPK siRNA reduced the efficacy of metformin to sensitize the cells to radiation. Cells were incubated with 1 or 5 mM metformin for 24 h, irradiated with various doses of X-rays, further incubated with metformin for 24 h and then cultured for colony formation. The radiation survival curves were normalized for the cell death caused by metformin alone.
Figure 4. Effect of metformin on the…
Figure 4. Effect of metformin on the CSCs or CS-like cells.
Cells were incubated with different concentrations of metformin for 48 h, dispersed to single cells, washed, and the fraction of CSCs or CS-like cells were analyzed. (A) FACS for CD44high/CD24low cells of MCF-7 cells treated with 0.5–5.0 mM metformin for 48 h. (B) % of CD44high/CD24low MCF-7 cells in the surviving cells after treating with different concentrations of metformin for 48 h. Means of 7 experiments ± 1 S.E are shown. The declines in % of CD44high/CD24low cells by 0.5–10 mM metformin were statistically significant (P < 0.05). (C) FACS for ALDH1-positive FSaII cells treated with metformin for 48 h. (D) % of ALDH1-positive cells of FSaII cells treated with different concentrations of metformin for 48 h. Means of 5–7 experiments ± 1 S.E are shown. The declines in % of ALDH1-positive cells by 0.1–1.0 mM metformin were statistically significant (P < 0.05). (E) Changes in % of SP cells in FSaII cells treated with different concentrations of metformin for 48 h.
Figure 5. Effect of metformin on the…
Figure 5. Effect of metformin on the growth of spheres of MCF-7 and FSaII cells.
Cells were cultured for 8 days in ultralow attachment wells with sphere media containing different concentrations of metformin. (A) MCF-7 spheres grown with or without 0.1 mM metformin. (B) The numbers of sphere with > 50 µm in diameter obtained from 103 cells. Means of 5 experiments ± 1 S.E are shown. *P < 0.05 between control and metformin treated groups.
Figure 6. Effects of metformin on the…
Figure 6. Effects of metformin on the radiosensitivity of CSCs or CS-like cells.
(A) MCF-7 cells were irradiated, incubated with or without 1 mM metformin for 48 h and analyzed for CD44high/CD24low cells. (B) % of CD44high/CD24low MCF-7 cells after irradiation and 48 h incubation with 1 mM metformin. Means of 5 experiments ± 1 S.E. *P < 0.05 for IR-induced increases. **P < 0.05 between IR and IR + Metformin. (C) % of ALDH1-positive FSaII cells after irradiation and 48 h incubation with 1 mM metformin. Means of 5 experiment ± 1 S.E. *P < 0.05 for IR-induced increases. **P < 0.05 between IR and IR + 1 mM Metformin groups.
Figure 7. Growth and immunohistochemistry of FSaII…
Figure 7. Growth and immunohistochemistry of FSaII tumors.
(A) Tumors grown (s.c.) to 150 mm3 in the hind legs of C3H mice were treated as follows. (a) Control. (b) Metformin: mice were injected i.p. twice a day with metformin at 25 mg/kg/dose from Day 0. (c) Tumors were irradiated with 20 Gy of X-rays in a single exposure. (d) 20 Gy + Metformin: Tumors were irradiated with 20 Gy of X-rays in a single exposure 1 h after the first metformin treatment. Data points are means of 7–10 tumors ± 1 S.E. (B–E) Immunohistological study for the expression of p-AMPK and p-mTOR in FSaII tumors. Tumors were treated with metformin and 20 Gy alone or combined as the tumors were treated for the tumor growth study (A), and tumors were excised on day 6. The cross-sections were stained and quantified for p-AMPK (B, C) and p-mTOR (D, E). All results are expressed as mean pixel counts per image ± standard error from 20 images derived from 3 tumors per group (magnification 200x). Representative images illustrating the average amount of staining are depicted in panels B and D for each treatment modality as indicated. *P < 0.001 treatment group vs. vehicle; #P < 0.05 metformin and radiation group vs. metformin alone.

References

    1. Witters L. A. The blooming of the french lilac. J Clin Metab. 1, 15–25 (2005).
    1. Hundal H. S., Ramlal T., Reyes R., Leiter L. A. & Klip A. Cellular mechanism of metformin action involves glucose transporter translocation from an intracellular pool to the plasma membrane in L6 muscle cells. Endocrinology. 131, 1165–1173 (1992).
    1. Dowling R. J. O., Goodwin R. J. & Stambolic V. Understanding the benefit of metformin use in cancer treatment. BMC Med. 9, 33 (2001).
    1. Sahra I. B., Marchand-Brustel Y., Tanti J. & Bost F. Metformin in cancer therapy: a new perspective for an old antidiabetic drug? Mol Cancer Ther. 9, 1092–1099 (2010).
    1. Gonzalez-Angulo A. M. & Meric-Bernstam F. Metformin: A therapeutic opportunity in breast cancer. Clin Cancer Res. 16, 1695–1700 (2010).
    1. Kourelis T. V. & Siegel R. D. Metformin and cancer: new applications for an old drug. Med Oncol. DOD 10.1007/s12032–011–9846–7.
    1. Micic D., Cvijovic G., Trajkovic V., Duntas L. H. & Polovina S. Metformin: Its emerging role in oncology. Hormones. 10, 5–15 (2011).
    1. Evans J. M. M., Donnely L. A., Emslie-Smith A., Alessi D. R. & Morris A. D. Metformin and reduced risk of cancer in diabetic patients. BMJ. 330, 1304–1305 (2005).
    1. Li D., Yeung S. J., Hassan M. M., Konopleva M. & Abbruzzese J. L. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 137, 482–488 (2009).
    1. Gagnon B., roseman M., Kasymjanova G., MacDonald H. & Kreisman D. Protective effect of metformin in lung cancer patients. J Clin Oncol. 27, No 15s Supplement e22063 (2009).
    1. Jiralerspong S. et al. Metformin and pathologic complete response to neoadjuvant chemotherapy in diabetic patients with breast cancer. J Clin Oncol. 20, 3297–3302 (2009).
    1. Dowling R. J., Zakikhani M., Fantus I. G., Pollak M. & Sonenberg N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 67, 10804–10812 (2007).
    1. Alimova I. N. et al. Metformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro. Cell Cycle. 8, 909–915 (2009).
    1. Zakikhani M., Dowling R. J., Sonenberg N. & Pollak M. The effects of adiponectin and metformin on prostate and colon neoplasia involve activation of AMP-Activated protein kinase. Cancer Prev Res. 1, 369–375 (2008).
    1. Liu B. et al. Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle. 8, 2031–2040 (2009).
    1. Kalender A. et al. Metformin, independent of AMPK, inhibits mTORC1 in a Rag GTPase-dependent manner. Cell Metab. 11, 390–401 (2010).
    1. Memmot R. M., Mercado J. R., Maier C. R., Kawabata S., Fox S. D. & Dennis P. A. Metformin prevents tobacco carcinogen-induced lung tumorigeneisis. Cancer Prev Res. 3, 1066–1076 (2010).
    1. Zhou J. et al. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. PNAS. 41, 16158–16163 (2007).
    1. Eyler C. E., Foo W. C., LaFiura K. M., McLendon R. E., Hjelmeland A. B. & Rich J. N. Brain cancer stem cells display preferential sensitivity to Akt inhibition. Stem Cells. 26, 3027–3036 (2008).
    1. Hill R. & Wu H. PTEN, stem cells, and cancer stem cells. J Biol Chem. 284, 11755–11759 (2009).
    1. Zakikhani M., Dowling R., Fantus I. G., Sonenberg N. & Pollak M. Metformin is an AMP Kinase-dependant growth inhibitor for breast cancer cells. Cancer Res. 66, 10269–10273 (2006).
    1. Martin-Castillon B., Vazquez-Martin A., Oliveras-Ferraros C. & Menendez J. A. Metformin and cancer: doses, mechanisms and the dandelion and hormetic phenomena. Cell Cycle. 9, 1057–1064 (2010).
    1. Karkaya H. et al. Regulation of mammary stem/progenitor cells by PTEN/Akt/B-catenin signaling. PLoS Biol. 7, e10000121 (2009).
    1. Rocha G. et al. Metformin amplifies chemotherapy-induced AMPK activation and antitumoral growth. Clin C ancer Res. 17, 3993–4005 (2011).
    1. Sanli T. et al. Ionizing radiation activates AMP-activated kinase (AMPK): a target for radiosensitization of human cancer cells. Int J Radiat Oncol Biol Phys. 78, 221–229 (2010).
    1. Rashid A. et al. Resveratrol enhances prostate caner cell response to ionizing radiation. Modulation of the AMPK, Akt and mTOR pathway. Radiat Oncol. 6, 144–155 (2011).
    1. Zannella V. E. et al. AMPK regulates metabolism and survival in response to ionizing radiation. Radiother Oncol. 99, 293–299 (2011).
    1. Al-Hajj M., Wicha M., Benito-Hernandes A., Morrison S. J. & Clarke M. F. Prospective identification of tumorigenic breast cancer cells. PNAS. 100, 3983–3988 (2003).
    1. Lobo N. A., Simono Y., Quian D. & Clarke M. F. The biology of cancer stem cells. Annu Rev Cell Dev Biol. 23, 675–699 (2007).
    1. O'Brien C. A., Kreso A. & Jamieson H. M. Cancer stem cells and self-renewal. Clin Cancer Res. 16, 3113–3120 (2010).
    1. Dalerba P., Cho R. W. & Clarke M. F. Cancer stem cells: Models and concept. Annu Rev Med. 58, 26–84 (2007).
    1. Lou H. & Dean M. Targeted therapy of cancer stem cells: the patched pathway and ABC Transporters. Oncogen. 26, 1357–1360 (2007).
    1. Zhou B. S., Zhang H., Damelin M., Geles K. G., Grindley J. C. & Dirks P. B. Tumor-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov 8, 806–823 (2009).
    1. Ma S., Lee T. K., Zheng B-J., Chan K. W. & Guan X-Y. CD133+ HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway. Oncogene. 27, 1749–1758 (2008).
    1. Frosina G. DNA Repair and Resistance of gliomas to chemotherapy and radiotherapy. Mol Cancer Res. 7, 989–999 (2009).
    1. Bao S. et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 444, 756–760 (2006).
    1. Phillips T. M., McBride W. H. & Pajonk F. The Response of CD24-/low/CD44+ breast cancer initiating cells to radiation. J Natl Cancer Inst. 98, 1777–1785 (2006).
    1. Baumann M., Krause M. & Hill R. Exploring the role of cancer stem cells in radioresistance. Nat Rev Cancer. 8, 545–554 (2008).
    1. 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. 69, 7507–7511 (2009).
    1. 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. 71, 3196–3201 (2011).
    1. Vazquerz-Martin A., Oliveras-Ferraros C., Barco S. D., Martin-Castillo B. & Menendez J. A. The anti-diabetic drug metformin suppresses self-renewal and proliferation of trastuzumbab-resistant tumor-initiating breast cancer stem cells. Breast Cancer Res Treat. 126, 355–364 (2011).
    1. Choi B. H. et al. Response of cancer stem cells to ionizing radiation and metformin alone and combined. Abstract # PS3.49, 56th Annual Meeting of the Radiation Research Society, Grand Wailea Resort Hotel and Spa, Maui, Hawaii. Sept 25th–29th (2010)
    1. Song C. W. et al. Metformin increases the response of tumors to radiotherapy by eliminating cancer stem cells. Abstract # PS3.57. 56th Annual Meeting of the Radiation Research Society, Grand Wailea Resort Hotel and Spa, Maui, Hawaii. Sept 25th–29th (2010).
    1. Charafe-Jauffret E., Ginesteir C. & Birnbaum D. Breast cancer stem cells: tools and models to rely on. BMC Cancer. 9, 1471–2407 (2009).
    1. Ginestier C. et al. ALDH1 is a marker of normal and malignant human mammary stem cells and predictor of poor clinical outcome. Cell Stem Cell. 1, 555–597 (2007).
    1. Li Y. et al. Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells. Clin Cancer Res. 16, 2580–2590 (2010).
    1. Levina V., Marrangoni A. M., DeMarco R., Gorelik E. & Lokshin A. E. Drug selected human lung cancer stem cells: cytokine network, tumorigenic and metastatic properties. PLoS One. 3, e3007 (2008).
    1. Nagata Y. et al. Effect of rapamycin, an mTOR inhibitor, on radiation sensitivity of lung cancer cells having different p53 gene status. Int J Oncol. 37, 1001–1010 (2010).
    1. Shinohaa E. T. et al. Enhanced radiation damage of tumor vasculature by mTOR inhibitiors. Oncogen. 24, 5414–5422 (2005).
    1. Braunstein S., Badura M. L., Xi Q., Formenti S. C. & Schneider R. J. Regulation of protein sysnthesis by ionizing radiation. Mol Cell Biol. 29, 5645–5656 (2009).
    1. Buzzai M. et al. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res. 67, 6745–6752 (2007).
    1. Kisfalvi K., Eibl G., Sinnett-Smith J. & Rozengurt E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res. 69, 6539–6545 (2009).
    1. Rattan R., Graham R. P., Maguire J. L., Giri S. & Shridhar V. Metformin suppresses ovarian cancer growth and metastasis with enhancement of cisplatin cytotoxicity in vivo. Neolasia. 13, 483–491 (2011).
    1. Park H. et al. Susceptibility of cancer cells to B-Lapachone is enhanced by ionizing radiation. Int J Radiat Oncol Biol Phys. 61, 212–219 (2005).
    1. Dings R. et al. Tumour thermotolerance, a physiological phenomenon involving vessel normalisation. Int J Hyperthermia. 27, 42–52 (2011).
    1. Dings R. et al. Inhibiting tumor growth by targeting tumor vasculature with galectin-1 antagonist anginex conjugated to the cytotoxic acylfulvene, 6-hydroxylpropylacylfulvene. Bioconjug Chem. 21, 20–7 (2010).
    1. Dings R. et al. Scheduling of radiation with angiogenesis inhibitors Anginex and Avastin improves therapeutic outcome via vessel normalization. Clin Cancer Res. 13, 3395–3402 (2007).

Source: PubMed

3
Prenumerera