Tumor cells have decreased ability to metabolize H2O2: Implications for pharmacological ascorbate in cancer therapy

Claire M Doskey, Visarut Buranasudja, Brett A Wagner, Justin G Wilkes, Juan Du, Joseph J Cullen, Garry R Buettner, Claire M Doskey, Visarut Buranasudja, Brett A Wagner, Justin G Wilkes, Juan Du, Joseph J Cullen, Garry R Buettner

Abstract

Ascorbate (AscH-) functions as a versatile reducing agent. At pharmacological doses (P-AscH-; [plasma AscH-] ≥≈20mM), achievable through intravenous delivery, oxidation of P-AscH- can produce a high flux of H2O2 in tumors. Catalase is the major enzyme for detoxifying high concentrations of H2O2. We hypothesize that sensitivity of tumor cells to P-AscH- compared to normal cells is due to their lower capacity to metabolize H2O2. Rate constants for removal of H2O2 (kcell) and catalase activities were determined for 15 tumor and 10 normal cell lines of various tissue types. A differential in the capacity of cells to remove H2O2 was revealed, with the average kcell for normal cells being twice that of tumor cells. The ED50 (50% clonogenic survival) of P-AscH- correlated directly with kcell and catalase activity. Catalase activity could present a promising indicator of which tumors may respond to P-AscH-.

Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Normal cells have a more robust capacity to remove extracellular H2O2than tumor cells. The rate constants, kcell, at which 10 normal cell lines and 15 cancer cell lines remove H2O2 were measured (listed in Table 1). There was a wide range of capacities for removal of H2O2 across all cell types. On average, normal cells had a 2-fold higher rate constant for the removal of H2O2 than tumor cells (p<0.05).
Fig. 2
Fig. 2
Catalase activity varies across cancer cell lines and correlates with the rate constant for removal of H2O2(kcell). (A) Catalase activity for cell lines of different tissue origins (i.e. pancreas (purple), breast (green), lung (red), and liver (blue)) were determined and used to calculate the effective number of fully active catalase monomers per cell. This number varied 5-fold across the different cancer cell lines: from 101,000 monomers per cell (MIA PaCa-2) to 538,000 monomers per cell (339) (n =3–9, error bars are standard error of the mean). (B) There is a strong correlation between the rate constant at which these cell lines remove extracellular H2O2 and the effective number of fully active catalase molecules per cell (R2 =0.88). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
Fig. 3
Fig. 3
Catalase plays a major role in removal of H2O2. (A) Treatment of HepG2 cells with 100 µM buthionine sulfoximine (BSO) 24 h prior to the H2O2-removal assay to inhibit glutathione synthesis did not result in any change in the rate constant by which these cells remove H2O2. However, treatment of HepG2 cells with 20 mM 3-AT for 1 h to inhibit catalase resulted in a four-fold decrease in the rate constant by which HepG2 cells remove extracellular H2O2 (n =4, error bars are standard error of the mean). (B) There is a direct correlation between the number of active catalase molecules per cell and the rate constant for removal of H2O2 following transduction of MIA PaCa-2 cells with adenovirus catalase (0–25 MOI) (R2 =0.91).
Fig. 4
Fig. 4
Dose of ascorbate is better specified on a per cell basis (pmol cell−1) than as initial concentration in the medium (mM). MIA PaCa-2 cells at varying cell densities (45,000–543,000 cells/3.0 mL medium) were treated with 5 mM ascorbate 1 h; ATP was measured immediately after. Dose of ascorbate is expressed as: (A) initial concentration of ascorbate in the medium; and (B) absolute amount of ascorbate (pmol) per cell.
Fig. 5
Fig. 5
Sensitivity to ascorbate varies across pancreatic cancer cell lines and correlates with the capacity to remove extracellular H2O2(k1-cell). (A) The ED50 of ascorbate was determined in MIA PaCa-2, AsPC-1, 403, 339, and PANC-1 cell lines using a clonogenic survival assay. The dose of ascorbate needed to decrease clonogenic survival by 50% varied across pancreatic cancer cell lines. When the rate constants for removal of extracellular H2O2 by a cell (k1-cell) for these 5 different pancreatic cancer cell lines are plotted against the ED50 of P-AscH− there is a direct correlation between sensitivity to P-AscH− and the rate at which cells remove H2O2 (R2 =0.69). The rate constant k1-cell represents the capacity of a single cell to remove extracellular H2O2. It is determined by: k1-cell (s−1) = kcell (s−1 cell−1 L) x 1 (cell L−1). (B) Transduction of MIA PaCa-2 cells with adenovirus catalase at increasing MOIs increases resistance to ascorbate as seen by ED50. MIA PaCa-2 cells were transduced with adenovirus catalase at 0–25 MOI and then exposed to ascorbate (0–50 pmol cell−1). The dose that decreased clonogenic survival by 50% was determined at each transduction-MOI of adenovirus catalase (0–25 MOI). Catalase activity was measured after transduction with adenovirus catalase. The resulting ED50 correlated with catalase activity at varying MOI of adenovirus catalase (R2 =0.94).
Fig. 6
Fig. 6
Inhibition of catalase with 3-amino-1,2,4-triazole sensitizes PANC-1 cells to ascorbate parallels the decrease inkcell. PANC-1 cells were treated with 20 mM 3-AT for 1 h prior to treatment with 0–17 pmol cell−1 ascorbate (350,000 cells; 0–2 mM) for 1 h. Cells were then plated for a clonogenic survival assay. 3-AT sensitized PANC-1 cells to ascorbate. The ED50 of ascorbate was 1.5-fold less with 3-AT treatment than without (n =3, error bars are standard error of the mean).
Fig. 7
Fig. 7
H2O2generated by ascorbate induces damage to nDNA and mtDNA in MIA PaCa-2 cells and depletes intracellular ATP. (A) MIA PaCa-2 cells were treated with ascorbate (3.5–28 pmol cell−1) for 1 h and then the frequency of DNA lesions was quantified with QPCR. Ascorbate treatment caused dose-dependent damage to nDNA and mtDNA (for nDNA: n =4, mean±SEM, * p<0.05 vs. 3.5 pmol cell−1; for mtDNA: n =8, mean±SEM, ** p<0.001 vs. 3.5 pmol cell−1). (B) MIA PaCa-2 cells were incubated with ascorbate (14 pmol cell−1), or ascorbate (14 pmol cell−1) and bovine catalase (200 units mL−1), or bovine catalase (200 units mL−1) alone for 1 h. QPCR analysis revealed no DNA damage from ascorbate when catalase is present in the medium indicating that the DNA damage is caused by H2O2 (nDNA: n =4; mean±SEM; * p<0.01 vs. ascorbate; mtDNA: n =4; mean±SEM; ** p<0.001 vs. ascorbate). (C) MIA PaCa-2 cells were treated with ascorbate (14 pmol cell−1), combination of ascorbate (14 pmol cell−1) and bovine catalase (200 units mL−1), or bovine catalase (200 units mL−1) for 1 h and then intracellular ATP was determined. ATP was depleted upon treatment with ascorbate, but was unchanged when catalase was present in the medium (n =4; mean±SEM; * p<0.001 vs. P-AscH−).
Fig. 8
Fig. 8
Pharmacological ascorbate slows growth of MIA PaCa-2 tumors in comparison to PANC-1 tumorsin vivo.(A) MIA PaCa-2 (kcell =1.1×10−12 s−1 cell−1 L; 101,000 active catalase monomers per cell) cells and (B) PANC-1 (kcell =5.1×10−12 s−1 cell−1 L; 459,000 active catalase monomers per cell) cells were injected into mice and formed tumors. Mice were treated with IP ascorbate (4 g/kg) twice daily for two weeks. Tumors were measured on day 3, 7, 10, and 14 following first treatment with ascorbate. P-AscH− slowed the growth rate of PANC-1 xenograft tumors to 42% of the controls; with MIA PaCa-2 tumor xenografts P-AscH− slowed growth to just 9% of controls. The ratio of kcell(PANC-1)/kcell(MIA PaCa-2) =4.6; the ratio for the relative growth rates compared to controls is essentially identical, 42%/9% =4.7, a remarkable quantitative comparison. (C) MIA PaCa-2 tumor catalase immunofluorescence, and (D) PANC-1 tumor catalase immunofluorescence. Tumor samples were fixed with 4% paraformaldehyde at 4 °C, and blocked with 5% goat serum for 30 min at 20 °C. The samples were incubated with catalase antibody (1:50) for 20 h at 4 °C. An Alexa Fluor 488 nm goat anti-Rabbit (1:200) was used as secondary antibody. DAPI was used to stain the cell nuclei. The samples were examined using a Zeiss confocal microscope. Scale bar, 20 µm. Tissue samples for PANC-1-derived tumors show considerably more immunofluorescence due to the presence of catalase enzyme than tissue samples from MIA PaCa-2 tumors. (Normalized fluorescent intensity of PANC-1 vs. MIA PaCA-2 is 100±27 vs. 2.0±0.5.).

References

    1. Chen Q., Espey M.G., Krishna M.C., Mitchell J.B., Corpe C.P., Buettner G.R., Shacter E., Levine M. Ascorbic acid at pharmacologic concentrations selectively kills cancer cells: ascorbic acid as a pro-drug for hydrogen peroxide delivery to tissues. Proc. Natl. Acad. Sci. USA. 2005;102:13604–13609. (PMID: 16157892)
    1. Chen Q., Espey M.G., Sun A.Y., Lee J.H., Krishna M.C., Shacter E., Choyke P.L., Pooput C., Kirk K.L., Buettner G.R., Levine M. Ascorbic acid in pharmacologic concentrations: a pro-drug for selective delivery of ascorbate radical and hydrogen peroxide to extracellular fluid in vivo. Proc. Natl. Acad. Sci. USA. 2007;104:8749–8754. (PMID: 17502596)
    1. Du J., Martin S.M., Levine M., Wagner B.A., Buettner G.R., Wang S.H., Taghiyev A.F., Du C., Knudson C.M., Cullen J.J. Mechanisms of ascorbate-induced cytotoxicity in pancreatic cancer. Clin. Cancer Res. 2010;16(2):509–520. (PMID: 20068072)
    1. Rawal M., Schroeder S.R., Wagner B.A., Cushing C.M., Welsh J., Button A.M., Du J., Sibenaller Z.A., Buettner G.R., Cullen J.J. Manganoporphyrins increase ascorbate-induced cytotoxicity by enhancing H2O2 generation. Cancer Res. 2013;73(16):5232–5241. (PMID: 23764544)
    1. Sestili P., Brandi G., Brambilla L., Cattabeni F., Cantoni O. Hydrogen peroxide mediates the killing of U937 tumor cells elicited by pharmacologically attainable concentrations of ascorbic acid: cell death prevention by extracellular catalase or catalase from cocultured erythrocytes or fibroblasts. J. Pharm. Exp. Ther. 1996;277(3):1719–1725. (PMID: 8667243)
    1. Chen Q., Espey M.G., Sun A.Y., Pooput C., Kirk K.L., Krishna M.C., Khosh D.B., Drisko J., Levine M. Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc. Natl. Acad. Sci. USA. 2008;32(105):11105–11109. (PMID: 18678913)
    1. Tian J., Peehl D.M., Knox S.J. Metalloporphyrin synergizes with ascorbic acid to inhibit cancer cell growth through fenton chemistry. Cancer Biother. Radiopharm. 2010;25(4):439–448. (PMID: 20735206)
    1. Espey M.G., Chen P., Chalmers B., Drisko J., Sun A.Y., Levine M., Chen Q. Pharmacologic ascorbate synergizes with gemcitabine in preclinical models of pancreatic cancer. Free Radic. Biol. Med. 2011;50(11):1610–1619. (PMID: 21402145)
    1. Ranzato E., Biffo S., Burlando B. Selective ascorbate toxicity in malignant mesothelioma: a redox Trojan mechanism. Am. J. Respir. Cell Mol. Biol. 2011;44(1):108–117. (PMID: 20203294)
    1. Chen P., Yu J., Chalmers B., Drisko J., Yang J., Li B., Chen Q. Pharmacological ascorbate induces cytotoxicity in prostate cancer cells through ATP depletion and induction of autophagy. Anticancer Drugs. 2012;23(4):437–444. (PMID: 22205155)
    1. Klingelhoeffer C., Kämmerer U., Koospal M., Mühling B., Schneider M., Kapp M., Kübler A., Germer C.T., Otto C. Natural resistance to ascorbic acid induced oxidative stress is mainly mediated by catalase activity in human cancer cells and catalase-silencing sensitizes to oxidative stress. BMC Complement. Alter. Med. 2012;12(61) (PMID: 22551313)
    1. Shatzer A.N., Espey M.G., Chavez M., Tu H., Levine M., Cohen J.I. Ascorbic acid kills Epstein-Barr virus positive Burkitt lymphoma cells and Epstein-Barr virus transformed B-cells in vitro, but not in vivo. Leuk. Lymphoma. 2013;54(5):1069–1078. (PMID: 23067008)
    1. Ma Y., Chapman J., Levine M., Polireddy K., Drisko J., Chen Q. High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy. Sci. Transl. Med. 2014;6(222) 222ra18 (PMID: 24500406)
    1. Cho C.S., Lee S., Lee G.T., Woo H.A., Choi E.J., Rhee S.G. Irreversible inactivation of glutathione peroxidase 1 and reversible inactivation of peroxiredoxin II by H2O2 in red blood cells. Antioxid. Redox Signal. 2010;12(11):1235–1246. (PMID: 2007018)
    1. Du J., Cullen J.J., Buettner G.R. Ascorbic acid: chemistry, biology and the treatment of cancer. Biochim. Biophys. Acta: Rev. Cancer. 2012;1826:443–457. (PMID: 22728050)
    1. Johnson R.M., HoY-S Yu.D.-Y., Kuypers F.A., Ravindranath Y., Goyette G.W. The effects of disruption of genes for peroxiredoxin-2, glutathione peroxidase-1, and catalase on erythrocyte oxidative metabolism. Free Radic. Biol. Med. 2010;48:519–525. (PMID: 19969073)
    1. Benfeitas R., Selvaggio G., Antunes F., Coelho P.M., Salvador A. Hydrogen peroxide metabolism and sensing in human erythrocytes: a validated kinetic model and reappraisal of the role of peroxiredoxin II. Free Radic. Biol. Med. 2014;74:35–49. (PMID: 24952139)
    1. Nicholls P. Activity of catalase in the red cell. Biochim Biophys. Acta. 1965;99:286–297. (PMID: 14336065)
    1. Cohen G., Hochstein P. Glutathione peroxidase: the primary agent for the elimination of hydrogen peroxide in erythrocytes. Biochemistry. 1963;2:1420–1428. (PMID: 14093920)
    1. Jones D.P., Eklöw L., Thor H., Orrenius S. Metabolism of hydrogen peroxide in isolated hepatocytes: relative contributions of catalase and glutathione peroxidase in decomposition of endogenously generated H2O2. Arch. Biochem. Biophys. 1981;210(2):505–516. (PMID: 7305340)
    1. Makino N., Mochizuki Y., Bannai S., Sugita Y. Kinetic studies on the removal of extracellular hydrogen peroxide by cultured fibroblasts. J. Biol. Chem. 1994;269(2):1020–1025. (PMID: 8288557)
    1. Sasaki K., Bannai S., Makino N. Kinetics of hydrogen peroxide elimination by human umbilical vein endothelial cells in culture. Biochim. Biophys. Acta. 1998;1380(2):275–288. (PMID: 9565698)
    1. Winterbourn C.C. Reconciling the chemistry and biology of reactive oxygen species. Nat. Chem. Biol. 2008;4(5):278–286. (PMID: 18421291)
    1. Makino N., Sasaki K., Hashida K., Sakakura Y. A metabolic model describing the H2O2 elimination by mammalian cells including H2O2 permeation through cytoplasmic and peroxisomal membranes: comparisons with experimental data. Biochim. Biophys. Acta. 2004;1673:149–159. (PMID: 15279886)
    1. Mitozo P.A., de Souza L.F., Loch-Neckel G., Flesch S., Maris A.F., Figueiredo C.P., Dos Santos A.R., Farina M., Dafre A.L. A study of the relative importance of the peroxiredoxin-, catalase-, and glutathione-dependent systems in neural peroxide metabolism. Free Radic. Biol. Med. 2011;51(1):69–77. (PMID: 21440059)
    1. Johnson R.M., Goyette G., Jr, Ravindranath Y., Ho Y.S. Hemoglobin autoxidation and regulation of endogenous H2O2 levels in erythrocytes. Free Radic. Biol. Med. 2005;39(11):1407–1417. (PMID: 16274876)
    1. De Duve C., Baudhuin P. Peroxisomes (microbodies and related particles) Physiol. Rev. 1966;46(2):323–357. (PMID: 5325972)
    1. Marklund S.L., Westman N.G., Lundgreen E., Roos G. Copper- and zinc-containing superoxide dismutase, catalase, and glutathione peroxidase in normal and neoplastic cell lines and normal human tissues. Cancer Res. 1982;42:1955–1961. (PMID: 7066906)
    1. Oberley T.D., Oberley L.W. Antioxidant enzyme levels in cancer. Histol. Histopathol. 1997;12:525–535. (PMID: 9151141)
    1. Oberley L.W., Buettner G.R. The role of superoxide dismutase in cancer: a review. Cancer Res. 1979;39:1141–1149. (PMID: 217531)
    1. Roy I., Zimmerman N.P., Mackinnon A.C., Tsai S., Evans D.B., Dwinell M.B. CXCL12 chemokine expression suppresses human pancreatic cancer growth and metastasis. PLoS One. 2014;9(3):e90400. (PMID: 24594697)
    1. Du J., Cieslak J.A., 3rd, Welsh J.L., Sibenaller Z.A., Allen B.G., Wagner B.A., Kalen A.L., Doskey C.M., Strother R.K., Button A.M., Mott S.L., Smith B., Tsai S., Mezhir J., Goswami P.C., Spitz D.R., Buettner G.R., Cullen J.J. Pharmacological ascorbate radiosensitizes pancreatic cancer. Cancer Res. 2015;75(16):3314–3326. (PMID: 26081808)
    1. Wagner B.A., Witmer J.R., van ‘t Erve T.J., Buettner G.R. An assay for the rate of removal of extracellular hydrogen peroxide by cells. Redox Biol. 2013;1:210–217. (PMID: 23936757)
    1. Aebi H. Catalase in vitro. Methods Enzym. 1984;105:121–126. (PMID: 6727660)
    1. Bonnichsen R. Blood catalase. Methods Enzym. 1955;2:781–784.
    1. Higashi T., Peters T., Jr. Studies of rat liver catalase. I. Combined immunochemical and enzymatic determination of catalase in liver cell fractions. J. Biol. Chem. 1963;238:3945–3951. (PMID: 14086728)
    1. Sies H., Bücher T., Oshino N., Chance B. Heme occupancy of catalase in hemoglobin-free perfused rat liver and of isolated rat liver catalase. Arch. Biochem. Biophys. 1973;154(1):106–116. (PMID: 4689773)
    1. Chance B., Sies H., Boveris A. Hydroperoxide metabolism in mammalian organs. Physiol. Rev. 1979;59(3):527–605. (PMID: 37532)
    1. Furda A., Santos J.H., Meyer J.N., Van Houten B. Quantitative PCR-based measurement of nuclear and mitochondrial DNA damage and repair in mammalian cells. Methods Mol. Biol. 2014;1105:419–437. (PMID: 24623245)
    1. Van Houten B., Cheng S., Chen Y. Measuring gene-specific nucleotide excision repair in human cells using quantitative amplification of long targets from nanogram quantities of DNA. Mutat. Res. 2000;460(2):81–94. (PMID: 10882849)
    1. Salazar J.J., Van Houten B. Preferential mitochondrial DNA injury caused by glucose oxidase as a steady generator of hydrogen peroxide in human fibroblasts. Mutat. Res. 1997;385(2):139–149. (PMID: 9447235)
    1. Euhus D.M., Hudd C., LaRegina M.C., Johnson F.E. Tumor measurement in the nude mouse. J. Surg. Oncol. 1986;31(4):229–234. (PMID: 3724177)
    1. Wagner B.A., Venkataraman S., Buettner G.R. The rate of oxygen utilization by cells. Free Radic. Biol. Med. 2011;51:700–712. (PMID: 21664270)
    1. Murphy M.P. How mitochondria produce reactive oxygen species. Biochem J. 2009;417(1):1–13. (PMID: 19061483)
    1. Olney K.E., Du J., van ‘t Erve T.J., Witmer J.R., Sibenaller Z.A., Wagner B.A., Buettner G.R., Cullen J.J. Inhibitors of hydroperoxide metabolism enhance ascorbate-induced cytotoxicity. Free Radic. Res. 2013;47(3):154–163. (PMID: 23205739)
    1. Buettner G.R., Ng C.F., Wang M., Rodgers V.G.J., Schafer F.Q. A new paradigm: manganese superoxide dismutase influences the production of H2O2 in cells and thereby their biological state. Free Radic. Biol. Med. 2006;41:1338–1350. (PMID: 17015180)
    1. De Duve C. The separation and characterization of subcellular particles. Harvey Lect. 1965;59:49–87. (PMID: 5337823)
    1. Doskey C.M., van ‘t Erve T.J., Wagner B.A., Buettner G.R. Moles of a substance per cell is a highly informative dosing metric in cell culture. PLoS One. 2015;10(7):e0132572. (PMID: 26172833)
    1. Spitz D.R., Elwell J.H., Sun Y., Oberley L.W., Oberley T.D., Sullivan S.J., Roberts R.J. Oxygen toxicity in control and H2O2-resistant Chinese hamster fibroblast cell lines. Arch. Biochem Biophys. 1990;279:249–260. (PMID:2350176)
    1. Gülden M., Jess A., Kammann J., Maser E., Seibert H. Cytotoxic potency of H2O2 in cell cultures: impact of cell concentration and exposure time. Free Radic. Biol. Med. 2010;49:1298–1305. (PMID: 20673847)
    1. Sobotta M.C., Barata A.G., Schmidt U., Mueller S., Millonig G., Dick T.P. Exposing cells to H2O2: a quantitative comparison between continuous low-dose and one-time high-dose treatments. Free Radic. Biol. Med. 2013;60:325–335. (PMID: 23485584)
    1. Du J., Cieslak J.A., 3rd, Welsh J.L., Sibenaller Z.A., Allen B.G., Wagner B.A., Kalen A.L., Doskey C.M., Strother R.K., Button A.M., Mott S.L., Smith B., Tsai S., Mezhir J., Goswami P.C., Spitz D.R., Buettner G.R., Cullen J.J. Pharmacological ascorbate radiosensitizes pancreatic cancer. Cancer Res. 2015;75(16):3314–3326. (PMID: 26081808)
    1. Martin D.S., Bertino J.R., Koutcher J.A. ATP depletion + pyrimidine depletion can markedly enhance cancer therapy: fresh insight for a new approach. Cancer Res. 2000;60(24):6776–6783. (PMID: 11156364)
    1. Zong W.X., Ditsworth D., Bauer D.E., Wang Z.Q., Thompson C.B. Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev. 2004;18(11):1272–1282. (PMID: 15145826)
    1. Herst P.M., Broadley K.W., Harper J.L., McConnell M.J. Pharmacological concentrations of ascorbate radiosensitize glioblastoma multiforme primary cells by increasing oxidative DNA damage and inhibiting G2/M arrest. Free Radic. Biol. Med. 2012;52(8):1486–1493. (PMID: 22342518)
    1. Castro M.L., McConnell M.J., Herst P.M. Radiosensitisation by pharmacological ascorbate in glioblastoma multiforme cells, human glial cells, and HUVECs depends on their antioxidant and DNA repair capabilities and is not cancer specific. Free Radic. Biol. Med. 2014;74:200–209. (PMID: 24992837)
    1. Cieslak J.A., Strother R.K., Rawal M., Du J., Doskey C.M., Schroeder S.R., Button A., Wagner B.A., Buettner G.R., Cullen J.J. Manganoporphyrins and ascorbate enhance gemcitabine cytotoxicity in pancreatic cancer. Free Radic. Biol. Med. 2015;83:227–237. (PMID: 25725418)
    1. Uetaki M., Tabata S., Nakasuka F., Soga T., Tomita M. Metabolomic alterations in human cancer cells by vitamin C-induced oxidative stress. Sci. Rep. 2015;5:13896. (PMID: 26350063)
    1. Yun J., Mullarky E., Lu C., Bosch, Kavalier A., Rivera K., Roper J., Chio I.I.C., Giannopoulou E.G., Rago C., Muley A., Asara J.M., Paik J., Elemento O., Chen Z., Pappin D.J., Dow L.E., Papadopoulos N., Gross S.S., Cantley L.C. Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science. 2015;350(6266):1391–1396. (PMID: 26541605)
    1. Du J., Wagner B.A., Buettner G.R., Cullen J.J. The role of labile iron in the toxicity of pharmacological ascorbate. Free Radic. Biol. Med. 2015;84:289–295. (PMID: 25857216)
    1. Mojić M., Bogdanović Pristov J., Maksimović-Ivanić D., Jones D.R., Stanić M., Mijatović S., Spasojević I. Extracellular iron diminishes anticancer effects of vitamin C: an in vitro study. Sci. Rep. 2014;4:5955. (PMID: 25092529)
    1. Schafer F.Q., Buettner G.R. Redox state of the cell as viewed though the glutathione disulfide/glutathione couple. Free Radic. Biol. Med. 2001;30:1191–1212. (PMID: 11368918)
    1. Welsh J.L., Wagner B.A., van ‘t Erve T.J., Zehr P.S., Berg D.J., Halfdanarson T.R., Yee N.S., Bodeker K.L., Du J., Roberts L.J., 2nd, Drisko J., Levine M., Buettner G.R., Cullen J.J. Pharmacological ascorbate with gemcitabine for the control of metastatic and node-positive pancreatic cancer (PACMAN): results from a phase I clinical trial. Cancer Chemother. Pharmacol. 2013;71(3):765–775. (PMID: 23381814)
    1. J. Cullen, D. Berg, J. Buatti, G. Buettner, M. Smith, C. Anderson, W. Sun, B. Allen, W. Rockey, D. Spitz, B. Wagner, S. Schroeder, R. HohlGemcitabine, ascorbate, and radiation therapy for pancreatic cancer, Phase 1, at The University of Iowa, (Start date 01/2014) (NCT01852890)
    1. Scheit K., Bauer G. Direct and indirect inactivation of tumor cell protective catalase by salicylic acid and anthocyanidins reactivated intracellular ROS signaling and allows for synergistic effects. Carcinogenesis. 2015;36(3):400–411. (PMID: 25653236)
    1. Scheit K., Bauer G. Synergistic effects between catalase inhibitors and modulators of nitric oxide metabolism on tumor cell apoptosis. Anticancer Res. 2014;34:5337–5350. (PMID: 25275027)
    1. Yakes F.M., Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc. Natl. Acad. Sci. USA. 1997;94(2):514–519. (PMID: 9012815)
    1. Cline S.D. Mitochondrial DNA damage and its consequences for mitochondrial gene expression. Biochim. Biophys. Acta. 2012;1819(9–10):979–991. (PMID: 22728831)
    1. Mini E., Nobili S., Caciagli B., Landini I., Mazzei T. Cellular pharmacology of gemcitabine. Ann. Oncol. 2016;17(Suppl 5):v7–v12. (PMID: 16807468)

Source: PubMed

3
Prenumerera