Advertisement
No access
Research Article
Cancer

Metabolic Modulation of Glioblastoma with Dichloroacetate

Science Translational Medicine
12 May 2010
Vol 2, Issue 31
p. 31ra34

Metabolic Modulators in Cancer

Cancer cells are optimized for growth, not performance. Their metabolism is geared to provide the raw materials needed for new cells, and that package includes resistance to apoptosis and mitochondria that run on aerobic glycolysis, not oxidative phosphorylation. In a small study, Michelakis et al. have taken advantage of these characteristics seen in glioblastoma, a deadly brain cancer, and examined the effects of a metabolic regulator, dichloroacetate, on tumor cells in culture and in patients, with some promising results.
Dichloroacetate activates pyruvate dehydrogenase, which in turn increases the flow of carbohydrates, in the form of pyruvate, into the mitochondria, where it enhances glucose oxidation by oxidative phosphorylation and decreases glycolysis. When the authors applied this agent to freshly excised glioblastoma tissue from 49 patients, the mitochondrial membrane potential (an index of mitochondrial function) improved, while it did not affect mitochondria from normal brain tissue. They then treated five glioblastoma patients with dichloroacetate, three of whom had recurring disease after traditional chemotherapy and two of whom were newly diagnosed. Three of the patients showed evidence of tumor regression on brain imaging scans, and four were clinically stable 15 months after therapy was initiated. Follow-up studies on cells taken from these patients before and after treatment showed that dichloroacetate normalized several mitochondrial functions, promoted apoptosis, and had other biochemical effects consistent with antitumor activity. Dichloroacetate appears to be safe to give to humans at doses that are required for pyruvate dehydrogenase inhibition. Now, this agent can be added to a growing group of metabolic modulators that may prove useful in cancer therapy.

Abstract

Solid tumors, including the aggressive primary brain cancer glioblastoma multiforme, develop resistance to cell death, in part as a result of a switch from mitochondrial oxidative phosphorylation to cytoplasmic glycolysis. This metabolic remodeling is accompanied by mitochondrial hyperpolarization. We tested whether the small-molecule and orphan drug dichloroacetate (DCA) can reverse this cancer-specific metabolic and mitochondrial remodeling in glioblastoma. Freshly isolated glioblastomas from 49 patients showed mitochondrial hyperpolarization, which was rapidly reversed by DCA. In a separate experiment with five patients who had glioblastoma, we prospectively secured baseline and serial tumor tissue, developed patient-specific cell lines of glioblastoma and putative glioblastoma stem cells (CD133+, nestin+ cells), and treated each patient with oral DCA for up to 15 months. DCA depolarized mitochondria, increased mitochondrial reactive oxygen species, and induced apoptosis in GBM cells, as well as in putative GBM stem cells, both in vitro and in vivo. DCA therapy also inhibited the hypoxia-inducible factor–1α, promoted p53 activation, and suppressed angiogenesis both in vivo and in vitro. The dose-limiting toxicity was a dose-dependent, reversible peripheral neuropathy, and there was no hematologic, hepatic, renal, or cardiac toxicity. Indications of clinical efficacy were present at a dose that did not cause peripheral neuropathy and at serum concentrations of DCA sufficient to inhibit the target enzyme of DCA, pyruvate dehydrogenase kinase II, which was highly expressed in all glioblastomas. Metabolic modulation may be a viable therapeutic approach in the treatment of glioblastoma.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Materials and Methods
Results
Discussion
Fig. S1. Molecular characterization of GBM tumors.
Fig. S2. Evolution of tumor response in patient 1.
Fig. S3. Evolution of tumor response in patient 4.
Fig. S4. Evolution of tumor response in patient 5.
Fig. S5. GBM MRI from patient 3.
Fig. S6. Mitochondrial membrane potential in GBM-SC from freshly excised GBM tissue.
Fig. S7. Characterization of primary GBM cells and GBM-SC.
Fig. S8. HXKII in GBM cells derived from patients before and after chronic DCA treatment.
Fig. S9 Effects of DCA therapy on GBM-SC and vascular apoptosis.
Fig. S10. Effects of DCA on angiogenesis in vitro.
Fig. S11. Effects of DCA treatment on p53 and p21 activity in vivo.
Fig. S12. A proposed comprehensive mechanism for the anticancer effects of DCA in GBM (see Supplementary Discussion).
Table S1. Laboratory and clinical parameters of five GBM patients before and after DCA treatment.
References

Resources

File (2-31ra34_sm.pdf)

References and Notes

1
Wen P. Y., Kesari S., Malignant gliomas in adults. N. Engl. J. Med. 359, 492–507 (2008).
2
Yung W. K., Albright R. E., Olson J., Fredericks R., Fink K., Prados M. D., Brada M., Spence A., Hohl R. J., Shapiro W., Glantz M., Greenberg H., Selker R. G., Vick N. A., Rampling R., Friedman H., Phillips P., Bruner J., Yue N., Osoba D., Zaknoen S., Levin V. A., A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse. Br. J. Cancer 83, 588–593 (2000).
3
Bonnet S., Archer S. L., Allalunis-Turner J., Haromy A., Beaulieu C., Thompson R., Lee C. T., Lopaschuk G. D., Puttagunta L., Bonnet S., Harry G., Hashimoto K., Porter C. J., Andrade M. A., Thebaud B., Michelakis E. D., A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell 11, 37–51 (2007).
4
Michelakis E. D., Webster L., Mackey J. R., Dichloroacetate (DCA) as a potential metabolic-targeting therapy for cancer. Br. J. Cancer 99, 989–994 (2008).
5
Stacpoole P. W., The pharmacology of dichloroacetate. Metabolism 38, 1124–1144 (1989).
6
O. Warburg, Ueber den Stoffwechsel der Tumoren (Constable, London, 1930).
7
Weinhouse S., The Warburg hypothesis fifty years later. Z. Krebsforsch. Klin. Onkol. Cancer Res. Clin. Oncol. 87, 115–126 (1976).
8
Vander Heiden M. G., Cantley L. C., Thompson C. B., Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).
9
Pan J. G., Mak T. W., Metabolic targeting as an anticancer strategy: Dawn of a new era? Sci. STKE 2007, pe14 (2007).
10
Kim J. W., Dang C. V., Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res. 66, 8927–8930 (2006).
11
Gatenby R. A., Gillies R. J., Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 4, 891–899 (2004).
12
Kim J. W., Dang C. V., Multifaceted roles of glycolytic enzymes. Trends Biochem. Sci. 30, 142–150 (2005).
13
Chen L. B., Mitochondrial membrane potential in living cells. Annu. Rev. Cell Biol. 4, 155–181 (1988).
14
Kroemer G., Galluzzi L., Brenner C., Mitochondrial membrane permeabilization in cell death. Physiol. Rev. 87, 99–163 (2007).
15
Zamzami N., Kroemer G., The mitochondrion in apoptosis: How Pandora’s box opens. Nat. Rev. Mol. Cell Biol. 2, 67–71 (2001).
16
Cairns R. A., Papandreou I., Sutphin P. D., Denko N. C., Metabolic targeting of hypoxia and HIF1 in solid tumors can enhance cytotoxic chemotherapy. Proc. Natl. Acad. Sci. U.S.A. 104, 9445–9450 (2007).
17
Cao W., Yacoub S., Shiverick K. T., Namiki K., Sakai Y., Porvasnik S., Urbanek C., Rosser C. J., Dichloroacetate (DCA) sensitizes both wild-type and over expressing Bcl-2 prostate cancer cells in vitro to radiation. Prostate 68, 1223–1231 (2008).
18
Dhar S., Lippard S. J., Mitaplatin, a potent fusion of cisplatin and the orphan drug dichloroacetate. Proc. Natl. Acad. Sci. U.S.A. 106, 22199–22204 (2009).
19
Sun R. C., Fadia M., Dahlstrom J. E., Parish C. R., Board P. G., Blackburn A. C., Reversal of the glycolytic phenotype by dichloroacetate inhibits metastatic breast cancer cell growth in vitro and in vivo. Breast Cancer Res. Treat. 120, 253–260 (2010).
20
Wong J. Y., Huggins G. S., Debidda M., Munshi N. C., De Vivo I., Dichloroacetate induces apoptosis in endometrial cancer cells. Gynecol. Oncol. 109, 394–402 (2008).
21
Wang S., Leonard S. S., Ye J., Ding M., Shi X., The role of hydroxyl radical as a messenger in Cr(VI)-induced p53 activation. Am. J. Physiol. Cell Physiol. 279, C868–C875 (2000).
22
Huang C., Zhang Z., Ding M., Li J., Ye J., Leonard S. S., Shen H. M., Butterworth L., Lu Y., Costa M., Rojanasakul Y., Castranova V., Vallyathan V., Shi X., Vanadate induces p53 transactivation through hydrogen peroxide and causes apoptosis. J. Biol. Chem. 275, 32516–32522 (2000).
23
Schmid T., Zhou J., Köhl R., Brüne B., p300 relieves p53-evoked transcriptional repression of hypoxia-inducible factor-1 (HIF-1). Biochem. J. 380, 289–295 (2004).
24
Blagosklonny M. V., An W. G., Romanova L. Y., Trepel J., Fojo T., Neckers L., p53 inhibits hypoxia-inducible factor-stimulated transcription. J. Biol. Chem. 273, 11995–11998 (1998).
25
Kim J. W., Tchernyshyov I., Semenza G. L., Dang C. V., HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 3, 177–185 (2006).
26
Semenza G. L., Artemov D., Bedi A., Bhujwalla Z., Chiles K., Feldser D., Laughner E., Ravi R., Simons J., Taghavi P., Zhong H., ‘The metabolism of tumours’: 70 years later. Novartis Found. Symp. 240, 251–260 (2001).
27
Weir E. K., López-Barneo J., Buckler K. J., Archer S. L., Acute oxygen-sensing mechanisms. N. Engl. J. Med. 353, 2042–2055 (2005).
28
Denko N. C., Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat. Rev. Cancer 8, 705–713 (2008).
29
Semenza G. L., Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721–732 (2003).
30
MacKenzie E. D., Selak M. A., Tennant D. A., Payne L. J., Crosby S., Frederiksen C. M., Watson D. G., Gottlieb E., Cell-permeating α-ketoglutarate derivatives alleviate pseudohypoxia in succinate dehydrogenase-deficient cells. Mol. Cell. Biol. 27, 3282–3289 (2007).
31
Bowker-Kinley M. M., Davis W. I., Wu P., Harris R. A., Popov K. M., Evidence for existence of tissue-specific regulation of the mammalian pyruvate dehydrogenase complex. Biochem. J. 329 (Pt 1), 191–196 (1998).
32
Stacpoole P. W., Kerr D. S., Barnes C., Bunch S. T., Carney P. R., Fennell E. M., Felitsyn N. M., Gilmore R. L., Greer M., Henderson G. N., Hutson A. D., Neiberger R. E., O’Brien R. G., Perkins L. A., Quisling R. G., Shroads A. L., Shuster J. J., Silverstein J. H., Theriaque D. W., Valenstein E., Controlled clinical trial of dichloroacetate for treatment of congenital lactic acidosis in children. Pediatrics 117, 1519–1531 (2006).
33
Stacpoole P. W., Lorenz A. C., Thomas R. G., Harman E. M., Dichloroacetate in the treatment of lactic acidosis. Ann. Intern. Med. 108, 58–63 (1988).
34
Stacpoole P. W., Kurtz T. L., Han Z., Langaee T., Role of dichloroacetate in the treatment of genetic mitochondrial diseases. Adv. Drug Deliv. Rev. 60, 1478–1487 (2008).
35
Kaufmann P., Engelstad K., Wei Y., Jhung S., Sano M. C., Shungu D. C., Millar W. S., Hong X., Gooch C. L., Mao X., Pascual J. M., Hirano M., Stacpoole P. W., DiMauro S., De Vivo D. C., Dichloroacetate causes toxic neuropathy in MELAS: A randomized, controlled clinical trial. Neurology 66, 324–330 (2006).
36
Stacpoole P. W., Gilbert L. R., Neiberger R. E., Carney P. R., Valenstein E., Theriaque D. W., Shuster J. J., Evaluation of long-term treatment of children with congenital lactic acidosis with dichloroacetate. Pediatrics 121, e1223–e1228 (2008).
37
Stacpoole P. W., Henderson G. N., Yan Z., James M. O., Clinical pharmacology and toxicology of dichloroacetate. Environ. Health Perspect. 106 (Suppl. 4), 989–994 (1998).
38
Sanai N., Alvarez-Buylla A., Berger M. S., Neural stem cells and the origin of gliomas. N. Engl. J. Med. 353, 811–822 (2005).
39
Zindy F., Uziel T., Ayrault O., Calabrese C., Valentine M., Rehg J. E., Gilbertson R. J., Sherr C. J., Roussel M. F., Genetic alterations in mouse medulloblastomas and generation of tumors de novo from primary cerebellar granule neuron precursors. Cancer Res. 67, 2676–2684 (2007).
40
Hide T., Takezaki T., Nakamura H., Kuratsu J., Kondo T., Brain tumor stem cells as research and treatment targets. Brain Tumor Pathol. 25, 67–72 (2008).
41
Calabrese C., Poppleton H., Kocak M., Hogg T. L., Fuller C., Hamner B., Oh E. Y., Gaber M. W., Finklestein D., Allen M., Frank A., Bayazitov I. T., Zakharenko S. S., Gajjar A., Davidoff A., Gilbertson R. J., A perivascular niche for brain tumor stem cells. Cancer Cell 11, 69–82 (2007).
42
Pallini R., Ricci-Vitiani L., Banna G. L., Signore M., Lombardi D., Todaro M., Stassi G., Martini M., Maira G., Larocca L. M., De Maria R., Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin. Cancer Res. 14, 8205–8212 (2008).
43
Laks D. R., Masterman-Smith M., Visnyei K., Angenieux B., Orozco N. M., Foran I., Yong W. H., Vinters H. V., Liau L. M., Lazareff J. A., Mischel P. S., Cloughesy T. F., Horvath S., Kornblum H. I., Neurosphere formation is an independent predictor of clinical outcome in malignant glioma. Stem Cells 27, 980–987 (2009).
44
Gilbertson R. J., Rich J. N., Making a tumour’s bed: Glioblastoma stem cells and the vascular niche. Nat. Rev. Cancer 7, 733–736 (2007).
45
Pastorino J. G., Hoek J. B., Hexokinase II: The integration of energy metabolism and control of apoptosis. Curr. Med. Chem. 10, 1535–1551 (2003).
46
Pastorino J. G., Hoek J. B., Shulga N., Activation of glycogen synthase kinase 3β disrupts the binding of hexokinase II to mitochondria by phosphorylating voltage-dependent anion channel and potentiates chemotherapy-induced cytotoxicity. Cancer Res. 65, 10545–10554 (2005).
47
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., Joshua B., Kaplan M. J., Wapnir I., Dirbas F. M., Somlo G., Garberoglio C., Paz B., Shen J., Lau S. K., Quake S. R., Brown J. M., Weissman I. L., Clarke M. F., Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 458, 780–783 (2009).
48
Yan H., Parsons D. W., Jin G., McLendon R., Rasheed B. A., Yuan W., Kos I., Batinic-Haberle I., Jones S., Riggins G. J., Friedman H., Friedman A., Reardon D., Herndon J., Kinzler K. W., Velculescu V. E., Vogelstein B., Bigner D. D., IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 360, 765–773 (2009).
49
Thompson C. B., Metabolic enzymes as oncogenes or tumor suppressors. N. Engl. J. Med. 360, 813–815 (2009).
50
Dang L., White D. W., Gross S., Bennett B. D., Bittinger M. A., Driggers E. M., Fantin V. R., Jang H. G., Jin S., Keenan M. C., Marks K. M., Prins R. M., Ward P. S., Yen K. E., Liau L. M., Rabinowitz J. D., Cantley L. C., Thompson C. B., Vander Heiden M. G., Su S. M., Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744 (2009).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science Translational Medicine
Volume 2 | Issue 31
May 2010

Submission history

Received: 11 November 2009
Accepted: 23 April 2010

Permissions

Request permissions for this article.

Acknowledgments

Funding: This study was funded by the Hecht Foundation (Vancouver, British Columbia, Canada; E.D.M.), the Canada Institutes for Health Research and the Canada Research Chairs Program (E.D.M.), and by public donations to the DCA program (received and managed by the Regents of the University of Alberta and the Faculty of Medicine). The authors would like to acknowledge the ort from the Alberta Health Services (D. Gordon, Senior Vice-President, Major Tertiary Hospitals). Author contributions: E.D.M. designed the studies, supervised the mechanistic studies, secured the funding, analyzed the data, and wrote the manuscript. K.C.P. co-designed the studies, supervised all clinical studies, and co-wrote the manuscript. G.S. and P.D. performed all the mechanistic studies and edited the manuscript. L.W. coordinated all studies, contributed to data acquisition, analyzed the clinical data, and edited the manuscript. A.H., E.N., C.M., T.-L.G., and M.S.M. contributed to data acquisition and data analysis and edited the manuscript. J.R.M., D.F., and B.A. co-designed the clinical studies, contributed to data acquisition, and edited the manuscript. Competing interests: E.D.M. is the co-owner of a pending use patent on the use of DCA as a cancer therapy. There has been no active or planned commercialization of this patent.

Authors

Affiliations

E. D. Michelakis* [email protected]
Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
G. Sutendra
Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
P. Dromparis
Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
L. Webster
Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
A. Haromy
Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
E. Niven
Department of Biomedical Engineering and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
C. Maguire
Department of Biomedical Engineering and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
T.-L. Gammer
Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
J. R. Mackey
Department of Oncology, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
D. Fulton
Department of Oncology, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
B. Abdulkarim
Department of Oncology, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
M. S. McMurtry
Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
K. C. Petruk
Department of Neurosurgery, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.

Notes

*
To whom correspondence should be addressed. E-mail: [email protected]

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Highlighted Advances in Therapies for Difficult-To-Treat Brain Tumours Such as Glioblastoma, Pharmaceutics, 15, 3, (928), (2023).https://doi.org/10.3390/pharmaceutics15030928
    Crossref
  2. The pyruvate dehydrogenase complex: Life’s essential, vulnerable and druggable energy homeostat, Mitochondrion, 70, (59-102), (2023).https://doi.org/10.1016/j.mito.2023.02.007
    Crossref
  3. Significance of mitochondrial activity in neurogenesis and neurodegenerative diseases, Neural Regeneration Research, 17, 4, (741), (2022).https://doi.org/10.4103/1673-5374.322429
    Crossref
  4. The Comparative Experimental Study of Sodium and Magnesium Dichloroacetate Effects on Pediatric PBT24 and SF8628 Cell Glioblastoma Tumors Using a Chicken Embryo Chorioallantoic Membrane Model and on Cells In Vitro, International Journal of Molecular Sciences, 23, 18, (10455), (2022).https://doi.org/10.3390/ijms231810455
    Crossref
  5. Connexins and Glucose Metabolism in Cancer, International Journal of Molecular Sciences, 23, 17, (10172), (2022).https://doi.org/10.3390/ijms231710172
    Crossref
  6. Pyruvate Dehydrogenase Kinase Inhibition by Dichloroacetate in Melanoma Cells Unveils Metabolic Vulnerabilities, International Journal of Molecular Sciences, 23, 7, (3745), (2022).https://doi.org/10.3390/ijms23073745
    Crossref
  7. Looking for the Holy Grail—Drug Candidates for Glioblastoma Multiforme Chemotherapy, Biomedicines, 10, 5, (1001), (2022).https://doi.org/10.3390/biomedicines10051001
    Crossref
  8. Global research trends and hotspots on glioma stem cells, Frontiers in Oncology, 12, (2022).https://doi.org/10.3389/fonc.2022.926025
    Crossref
  9. Targeting cancer-specific metabolic pathways for developing novel cancer therapeutics, Frontiers in Immunology, 13, (2022).https://doi.org/10.3389/fimmu.2022.955476
    Crossref
  10. Metabolic reprogramming and crosstalk of cancer-related fibroblasts and immune cells in the tumor microenvironment, Frontiers in Endocrinology, 13, (2022).https://doi.org/10.3389/fendo.2022.988295
    Crossref
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media