High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study

Kishore Polireddy, Ruochen Dong, Gregory Reed, Jun Yu, Ping Chen, Stephen Williamson, Pierre-Christian Violet, Ziyan Pessetto, Andrew K Godwin, Fang Fan, Mark Levine, Jeanne A Drisko, Qi Chen, Kishore Polireddy, Ruochen Dong, Gregory Reed, Jun Yu, Ping Chen, Stephen Williamson, Pierre-Christian Violet, Ziyan Pessetto, Andrew K Godwin, Fang Fan, Mark Levine, Jeanne A Drisko, Qi Chen

Abstract

Pancreatic cancer is among the most lethal cancers with poorly tolerated treatments. There is increasing interest in using high-dose intravenous ascorbate (IVC) in treating this disease partially because of its low toxicity. IVC bypasses bioavailability barriers of oral ingestion, provides pharmacological concentrations in tissues, and exhibits selective cytotoxic effects in cancer cells through peroxide formation. Here, we further revealed its anti-pancreatic cancer mechanisms and conducted a phase I/IIa study to investigate pharmacokinetic interaction between IVC and gemcitabine. Pharmacological ascorbate induced cell death in pancreatic cancer cells with diverse mutational backgrounds. Pharmacological ascorbate depleted cellular NAD+ preferentially in cancer cells versus normal cells, leading to depletion of ATP and robustly increased α-tubulin acetylation in cancer cells. While ATP depletion led to cell death, over-acetylated tubulin led to inhibition of motility and mitosis. Collagen was increased, and cancer cell epithelial-mesenchymal transition (EMT) was inhibited, accompanied with inhibition in metastasis. IVC was safe in patients and showed the possibility to prolong patient survival. There was no interference to gemcitabine pharmacokinetics by IVC administration. Taken together, these data revealed a multi-targeting mechanism of pharmacological ascorbate's anti-cancer action, with minimal toxicity, and provided guidance to design larger definitive trials testing efficacy of IVC in treating advanced pancreatic cancer.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Ascorbate inhibited pancreatic cancer growth and metastasis in vitro. (A) Dose responses of pancreatic cancer cells and normal cells to ascorbate. Cells were exposed to 0–20 mM ascorbate and cell viability was detected at 24 h by MTT assays. +cat, pre-incubation with 600 U/ml catalase. Data represents Mean ± SD of 3 experiments each done in triplicates. (B) Colony formation of pancreatic cancer cells under ascorbate treatment. Cells were exposed to 5 mM of ascorbate and colonies were counted at 21–28 days. Data shows % of colonies relative to untreated control (Mean ± SD of ≥3 replicates). (C) Matrigel invasion assays for pancreatic cancer cell migration and invasion. PANC-1 cells were seeded at 1 × 104 cells/insert and were exposed to 1 mM ascorbate (Asc). Cell migration (without Matrigel) and invasion (with Matrigel) were detected at 24 hrs. Bar graph shows the average number of migrated/invaded cells per field. Data represents ≥ 3 experiments each done in triplicates. (D) qRT-PCR for changes in EMT markers. PANC-1 cells were treated with 1.5 mM Asc for 24 hrs. Data was normalized to 18s rRNA, and then compared to control PANC-1 cells for fold change. Data represent Mean ± SD of 2–6 independent experiments. (E) Western blot in PANC-1 cells showing expression of E-cadherin (E-Cad), vimentin (Vim) and Snail after ascorbate treatment. Vinculin was a loading control. Left panel shows representative blots. Bar graph shows relative band density normalized to vinculin, analyzed by Image J. (F) qRT-PCR for MMP-2 mRNA expression in PANC-1 cells treated with ascorbate for 24 hrs. Data represents Mean ± SD of 3 independent experiments. (G) Gelatin zymography assay for MMP-2 enzymatic activity after ascorbate treatment. After ascorbate treatment cell lysate were used for RNA isolation, reverse transcription and qRT-PCR. Supernatant media was used for detection of MMP-2 activity using gelatin zymography. Data represents Mean ± SD of 3 independent experiments.
Figure 2
Figure 2
Ascorbate depleted NAD+ in pancreatic cancer cells and enhanced tubulin acetylation. (A) Western blot analysis of acetylated α-tubulin in pancreatic cancer cells (BxPC-3 and PANC-1), and an immortalized non-cancerous pancreatic ductal epithelial cell line hTERT-HPNE. Cells were treated for 4 hrs. (B) Immunofluorescence with confocal microscopy showing acetylated α-tubulin in cells treated with ascorbate (Asc) 2.5 mM or hydrogen peroxide (H2O2) 500 µM for 4 hrs. Asc + Cat, co-treatment of ascorbate and 600 U/ml catalase for 4 hrs. Cell nuclei were counter-stained blue with hoechst33342 (2 mg/mL). (C) Changes of NAD+ and (D) ATP in PANC-1, BxPC-3 and hTERT-HPNE cells treated with Asc for 4 hrs. NAD+ and ATP were detected by HPLC-UV analysis and normalized to protein contents. Data represents Mean ± SD of 2–4 independent experiments. (E) Supplementation of NAD+ protected PANC-1 cellular NAD+ levels with Asc treatment. (F) Western blot showing tubulin acetylation was prevented with supplementation of NAD+. (G) ATP in PANC-1 was protected with supplementation of NAD+. (H) Colony formation was protected with supplementation of NAD+. PANC-1 cells were pre-incubated with NAD+ for 30 min, and then treated with Asc and seeded into 2-layer soft agar for colony formation. Colonies were counted after 21 days of incubation. NAD+, tubulin acetylation, and ATP were detected at 4 h of treatment. Data represents Mean ± SD of ≥3 independent experiments.
Figure 3
Figure 3
Ascorbate over-stabilized polymerized tubulin by increased tubulin acetylation. (A) Tubulin Polymerization detected by native PAGE in PANC-1 and BxPC-3 cells. After 4 h of Asc or paclitaxel treatment, cells were lysed in RIPA buffer and subject to native PAGE. α-tubulin acetylation was confirmed by SDS PAGE and western blot as shown in the bottom panels. MW, molecular weight in kDa. (B) Cold induced microtubule depolymerization assay in PANC-1 and BxPC-3 pancreatic cancer cells. After Asc treatment for 4 h cell lysates were exposed to either 37 °C or sit on ice (4 °C) for indicated time, and were then subject to native PAGE. (C) Correlation between tubulin acetylation and cell death induced by ascorbate. All results are representatives of ≥3 independent experiments.
Figure 4
Figure 4
Ascorbate inhibited pancreatic cancer growth and metastasis in vivo. (A) Bioluminescence images of mice bearing orthotopic pancreatic xenografts treated with ascorbate (Asc), gemcitabine (Gem) or the combination of Asc+Gem. Day 0 indicated the beginning of treatment which was 2 weeks post orthotopic injection of luciferase expressing PANC-1-Leu cells into mouse pancreas. Day 45 was the end of the experiment. Asc, ascorbate treatment at intraperitoneal dose of 4 g/kg/day. Gem, gemcitabine at intraperitoneal dose of 40 mg/kg/week. Control (Ctrl) mice were treated with saline that had the same osmolarity as the ascorbate injections. (B) Total tumor burden per mouse by imaging was quantified as photons/sec/cm2 (Mean ± SEM). (C) Total tumor weight (Mean ± SEM), and (D) number of metastatic lesions in each mouse, determined by necropsy at Day 45. (E) Immunohistochemical analysis of proliferating cell nuclear antigen (PCNA) with formalin fixed tumor samples. Bar graph (right) represents the average number of PCNA positive cells per field. 15 fields from 3 different tumors from each group were analyzed. (F) Histological analysis of mitosis on H&E stained tumor slices. Bar graph (right) shows mitotic index, which was the average number of mitoses from 4 separate fields. Tumors from 4 mice in each group were examined. (G) Masson’s trichrome staining for collagen content in tumor tissues. Collagen was stained blue, and cytoplasm pink. Bar graph represents Mean ± SD of % area collagen/cross section. 15 fields from 3 different tumors from each group were analyzed. (H) H&E staining for analysis of desmoplasia in mouse tumor tissues. Bar graph shows desmoplasia represented as % of area contains desmoplastic response. Tumors from 4 mice in each group were examined (Mean ± SD). (I) Masson’s trichrome staining for collagen and fibrosis of livers from control and ascorbate treated mice. No collagen or fibrosis was seen. (J) qRT-PCR detection of 24 kinds of collagen transcripts in mouse tumor samples. Five tumors from each group were detected in duplicates. Bar shows fold changes compared to control mice in Mean ± SD. *P < 0.05; **P < 0.01; and ***P < 0.001. (K) Western blot in mouse tumor samples showing changes in CK-19 and Snail. Vinculin was used as loading control. (L) Immunohistochemistry in mouse tumor samples showing α-tubulin acetylation. Bar graph represents Mean ± SD of % area of positive staining for acetylated α-tubulin per cross section. 11–17 fields from 3 different tumors from each group were analyzed. (M) A simplified scheme showing mechanisms of ascorbate inhibiting pancreatic cancer growth and metastasis.
Figure 5
Figure 5
Overall survival (OS) and progression free survival (PFS) of the 12 participants who completed phase IIa study. Dotted lines showed median overall survival.
Figure 6
Figure 6
Pharmacokinetic parameters of gemcitabine, dFdU and ascorbate when IVC and gemcitabine were used alone or in combination. Cmax, AUC and t ½ of gemcitabine (A–C), dFdU (D–F) and ascorbate (G–I) were shown. Cmax and AUC were normalized to dose. Line graphs show parameters obtained for each subject from single drug administration (left) and combination administration (right). Box plots denote the median and the 5th, 25th, 75th, and 95th percentiles for all patients, and the outlying values.

References

    1. American Cancer Society. Cancer Facts and Figures 2017 (2017).
    1. Burris H, III, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. Journal of Clinical Oncology. 1997;15:2403. doi: 10.1200/JCO.1997.15.6.2403.
    1. Renouf D, Moore M. Evolution of systemic therapy for advanced pancreatic cancer. Expert Rev Anticancer Ther. 2010;10:529–540. doi: 10.1586/era.10.21.
    1. Kim R. FOLFIRINOX: a new standard treatment for advanced pancreatic cancer? Lancet Oncol. 2011;12:8–9. doi: 10.1016/S1470-2045(10)70237-0.
    1. Von Hoff DD, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–1703. doi: 10.1056/NEJMoa1304369.
    1. Moore MJ, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–1966. doi: 10.1200/JCO.2006.07.9525.
    1. Dahan L, et al. Combination 5-fluorouracil, folinic acid and cisplatin (LV5FU2-CDDP) followed by gemcitabine or the reverse sequence in metastatic pancreatic cancer: final results of a randomised strategic phase III trial (FFCD 0301) Gut. 2010;59:1527–1534. doi: 10.1136/gut.2010.216135.
    1. Oberstein PE, Saif MW. First-line treatment for advanced pancreatic cancer. Highlights from the “2011 ASCO Gastrointestinal Cancers Symposium”. San Francisco, CA, USA. January 20–22, 2011. JOP. 2011;12:96–100.
    1. Ma Y, et al. High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy. Science translational medicine. 2014;6:222ra218.
    1. Hoffer LJ, et al. Phase I clinical trial of i.v. ascorbic acid in advanced malignancy. Ann Oncol. 2008;19:1969–1974. doi: 10.1093/annonc/mdn377.
    1. Monti DA, et al. Phase I evaluation of intravenous ascorbic acid in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. PLoS One. 2012;7:e29794. doi: 10.1371/journal.pone.0029794.
    1. Welsh JL, et al. 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;71:765–775. doi: 10.1007/s00280-013-2070-8.
    1. Schoenfeld, J. D. et al. O2- and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate. Cancer Cell, 10.1016/j.ccell.2017.02.018 (2017).
    1. Yun J, et al. Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science. 2015;350:1391–1396. doi: 10.1126/science.aaa5004.
    1. Jones S, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–1806. doi: 10.1126/science.1164368.
    1. Graumlich JF, et al. Pharmacokinetic model of ascorbic acid in healthy male volunteers during depletion and repletion. Pharm Res. 1997;14:1133–1139. doi: 10.1023/A:1012186203165.
    1. Levine M, et al. Vitamin C pharmacokinetics in healthy volunteers: evidence for a recommended dietary allowance. Proc Natl Acad Sci USA. 1996;93:3704–3709. doi: 10.1073/pnas.93.8.3704.
    1. Padayatty SJ, et al. Vitamin C pharmacokinetics: implications for oral and intravenous use. Ann Intern Med. 2004;140:533–537. doi: 10.7326/0003-4819-140-7-200404060-00010.
    1. Chen Q, et al. Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc Natl Acad Sci USA. 2008;105:11105–11109. doi: 10.1073/pnas.0804226105.
    1. Chen Q, et al. Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo. Proc Natl Acad Sci USA. 2007;104:8749–8754. doi: 10.1073/pnas.0702854104.
    1. Chen Q, et al. Pharmacologic ascorbic acid concentrations selectively kill cancer cells: action as a pro-drug to deliver hydrogen peroxide to tissues. Proc Natl Acad Sci USA. 2005;102:13604–13609. doi: 10.1073/pnas.0506390102.
    1. Parrow NL, Leshin JA, Levine M. Parenteral ascorbate as a cancer therapeutic: a reassessment based on pharmacokinetics. Antioxid Redox Signal. 2013;19:2141–2156. doi: 10.1089/ars.2013.5372.
    1. Du J, et al. Mechanisms of ascorbate-induced cytotoxicity in pancreatic cancer. Clin Cancer Res. 2010;16:509–520. doi: 10.1158/1078-0432.CCR-09-1713.
    1. Verrax J, Calderon PB. Pharmacologic concentrations of ascorbate are achieved by parenteral administration and exhibit antitumoral effects. Free Radic Biol Med. 2009;47:32–40. doi: 10.1016/j.freeradbiomed.2009.02.016.
    1. Pollard HB, Levine MA, Eidelman O, Pollard M. Pharmacological ascorbic acid suppresses syngeneic tumor growth and metastases in hormone-refractory prostate cancer. In Vivo. 2010;24:249–255.
    1. Takemura Y, et al. High dose of ascorbic acid induces cell death in mesothelioma cells. Biochem Biophys Res Commun. 2010;394:249–253. doi: 10.1016/j.bbrc.2010.02.012.
    1. Deubzer B, et al. H(2)O(2)-mediated cytotoxicity of pharmacologic ascorbate concentrations to neuroblastoma cells: potential role of lactate and ferritin. Cell Physiol Biochem. 2010;25:767–774. doi: 10.1159/000315098.
    1. Espey MG, et al. Pharmacologic ascorbate synergizes with gemcitabine in preclinical models of pancreatic cancer. Free Radic Biol Med. 2011;50:1610–1619. doi: 10.1016/j.freeradbiomed.2011.03.007.
    1. Rhim AD, et al. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148:349–361. doi: 10.1016/j.cell.2011.11.025.
    1. Ellenrieder V, et al. Role of MT-MMPs and MMP-2 in pancreatic cancer progression. Int J Cancer. 2000;85:14–20. doi: 10.1002/(SICI)1097-0215(20000101)85:1<14::AID-IJC3>;2-O.
    1. Chiarugi A, Moskowitz MA. Cell biology. PARP-1–a perpetrator of apoptotic cell death? Science. 2002;297:200–201.
    1. Pero RW, et al. Oxidative stress induces DNA damage and inhibits the repair of DNA lesions induced by N-acetoxy-2-acetylaminofluorene in human peripheral mononuclear leukocytes. Cancer Res. 1990;50:4619–4625.
    1. Warburg O, Wind F, Negelein E. The Metabolism of Tumors in the Body. J Gen Physiol. 1927;8:519–530. doi: 10.1085/jgp.8.6.519.
    1. Sauve AA, Youn DY. Sirtuins: NAD(+)-dependent deacetylase mechanism and regulation. Current opinion in chemical biology. 2012;16:535–543. doi: 10.1016/j.cbpa.2012.10.003.
    1. Perdiz D, Mackeh R, Pous C, Baillet A. The ins and outs of tubulin acetylation: more than just a post-translational modification? Cellular signalling. 2011;23:763–771. doi: 10.1016/j.cellsig.2010.10.014.
    1. Hammond JW, Cai D, Verhey KJ. Tubulin modifications and their cellular functions. Current opinion in cell biology. 2008;20:71–76. doi: 10.1016/j.ceb.2007.11.010.
    1. Kirkland WL, Burton PR. Cyclic adenosine monophosphate-mediated stabilization of mouse neuroblastoma cell neuritis microtubules exposed to low temperature. Nat New Biol. 1972;240:205–207. doi: 10.1038/newbio240205a0.
    1. Marangon E, et al. Simultaneous determination of gemcitabine and its main metabolite, dFdU, in plasma of patients with advanced non-small-cell lung cancer by high-performance liquid chromatography-tandem mass spectrometry. J Mass Spectrom. 2008;43:216–223. doi: 10.1002/jms.1293.
    1. Doskey CM, et al. Tumor cells have decreased ability to metabolize H2O2: Implications for pharmacological ascorbate in cancer therapy. Redox biology. 2016;10:274–284. doi: 10.1016/j.redox.2016.10.010.
    1. Du J, Wagner BA, Buettner GR, Cullen JJ. Role of labile iron in the toxicity of pharmacological ascorbate. Free Radic Biol Med. 2015;84:289–295. doi: 10.1016/j.freeradbiomed.2015.03.033.
    1. Venturelli S, et al. Epigenetic impacts of ascorbate on human metastatic melanoma cells. Frontiers in oncology. 2014;4:227. doi: 10.3389/fonc.2014.00227.
    1. Castro ML, McConnell MJ, Herst PM. 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. doi: 10.1016/j.freeradbiomed.2014.06.022.
    1. Levine M, Violet PC. Data Triumph at C. Cancer Cell. 2017;31:467–469. doi: 10.1016/j.ccell.2017.03.008.
    1. Sinnberg T, et al. The ROS-induced cytotoxicity of ascorbate is attenuated by hypoxia and HIF-1alpha in the NCI60 cancer cell lines. J Cell Mol Med. 2014;18:530–541. doi: 10.1111/jcmm.12207.
    1. Kim J, et al. Enhanced antitumor activity of vitamin C via p53 in cancer cells. Free Radic Biol Med. 2012;53:1607–1615. doi: 10.1016/j.freeradbiomed.2012.07.079.
    1. Hong SW, et al. SVCT-2 in breast cancer acts as an indicator for L-ascorbate treatment. Oncogene. 2013;32:1508–1517. doi: 10.1038/onc.2012.176.
    1. Ma E, et al. Pharmacologic ascorbate induces neuroblastoma cell death by hydrogen peroxide mediated DNA damage and reduction in cancer cell glycolysis. Free Radic Biol Med. 2017;113:36–47. doi: 10.1016/j.freeradbiomed.2017.09.008.
    1. Xu RH, et al. Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res. 2005;65:613–621.
    1. Ahmad IM, et al. Mitochondrial O2*- and H2O2 mediate glucose deprivation-induced stress in human cancer cells. J Biol Chem. 2005;280:4254–4263. doi: 10.1074/jbc.M411662200.
    1. Karnoub AE, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449:557–563. doi: 10.1038/nature06188.
    1. Merika EE, Syrigos KN, Saif MW. Desmoplasia in pancreatic cancer. Can we fight it? Gastroenterol Res Pract. 2012;2012:781765. doi: 10.1155/2012/781765.
    1. Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer. 2001;1:46–54. doi: 10.1038/35094059.
    1. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–1659. doi: 10.1056/NEJM198612253152606.
    1. Ozdemir BC, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–734. doi: 10.1016/j.ccr.2014.04.005.
    1. Chen Q, Polireddy K, Chen P, Dong R. The unpaved journey of vitamin C in cancer treatment. Can J Physiol Pharmacol. 2015;93:1055–1063. doi: 10.1139/cjpp-2014-0509.
    1. Zhang DS, et al. Phase I/II study of albumin-bound nab-paclitaxel plus gemcitabine administered to Chinese patients with advanced pancreatic cancer. Cancer Chemother Pharmacol. 2013;71:1065–1072. doi: 10.1007/s00280-013-2102-4.
    1. Goldstein, D. et al. nab-Paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst107, 10.1093/jnci/dju413 (2015).
    1. Chiorean EG, et al. CA19-9 decrease at 8 weeks as a predictor of overall survival in a randomized phase III trial (MPACT) of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone in patients with metastatic pancreatic cancer. Ann Oncol. 2016;27:654–660. doi: 10.1093/annonc/mdw006.
    1. Ueno H, et al. Phase I/II study of nab-paclitaxel plus gemcitabine for chemotherapy-naive Japanese patients with metastatic pancreatic cancer. Cancer Chemother Pharmacol. 2016;77:595–603. doi: 10.1007/s00280-016-2972-3.
    1. Eisenhauer E, et al. New response evaluation criteria in solid tumors: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:288–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Chen Q, Polireddy K, Chen P, Dong R. The unpaved journey of vitamin C in cancer treatment. Can J Physiol Pharmacol. 2015;93:1–9. doi: 10.1139/cjpp-2014-0394.
    1. Ma Y, et al. A Convenient Method for Measuring Blood Ascorbate Concentrations in Patients Receiving High-Dose Intravenous Ascorbate. Journal of the American College of Nutrition. 2013;32:187–193. doi: 10.1080/07315724.2013.791167.
    1. Bollu LR, et al. Involvement of de novo synthesized palmitate and mitochondrial EGFR in EGF induced mitochondrial fusion of cancer cells. Cell cycle. 2014;13:2415–2430. doi: 10.4161/cc.29338.

Source: PubMed

Подписаться