Ribonucleotide reductase inhibition restores platinum-sensitivity in platinum-resistant ovarian cancer: a Gynecologic Oncology Group Study

Charles Kunos, Tomas Radivoyevitch, Fadi W Abdul-Karim, James Fanning, Ovadia Abulafia, Albert J Bonebrake, Lydia Usha, Charles Kunos, Tomas Radivoyevitch, Fadi W Abdul-Karim, James Fanning, Ovadia Abulafia, Albert J Bonebrake, Lydia Usha

Abstract

Background: The potent ribonucleotide reductase (RNR) inhibitor 3-aminopyridine-2-carboxyaldehyde-thiosemicarbazone (3-AP) was tested as a chemosensitizer for restored cisplatin-mediated cytotoxicity in platinum-resistant ovarian cancer.

Methods: Preclinical in vitro platinum-resistant ovarian cancer cell survival, RNR activity, and DNA damage assays were done after cisplatin or cisplatin plus 3-AP treatments. Six women with platinum-resistant ovarian cancer underwent four-day 3-AP (96 mg/m(2), day one to four) and cisplatin (25 mg/m(2), day two and three) infusions every 21 days until disease progression or adverse effects prohibited further therapy. Pre-therapy ovarian cancer tissues were analyzed by immunohistochemistry for RNR subunit expression as an indicator of cisplatin plus 3-AP treatment response.

Results: 3-AP preceding cisplatin exposure in platinum-resistant ovarian cancer cells was not as effective as sequencing cisplatin plus 3-AP together in cell survival assays. Platinum-mediated DNA damage (i.e., γH2AX foci) resolved quickly after cisplatin-alone or 3-AP preceding cisplatin exposure, but persisted after a cisplatin plus 3-AP sequence. On trial, 25 four-day overlapping 3-AP and cisplatin cycles were administered to six women (median 4.2 cycles per patient). 3-AP-related methemoglobinemia (range seven to 10%) occurred in two (33%) of six women, halting trial accrual.

Conclusions: When sequenced cisplatin plus 3-AP, RNR inhibition restored platinum-sensitivity in platinum-resistant ovarian cancers. 3-AP (96 mg/m(2)) infusions produced modest methemoglobinemia, the expected consequence of ribonucleotide reductase inhibitors disrupting collateral proteins containing iron.

Trial registry: ClinicalTrials.gov NCT00081276.

Figures

Figure 1
Figure 1
3-aminopyridine-2-carboxyaldehyde-thiosemicarbazone (3-AP) lowered 6-hour deoxycytidine triphosphates (dCTP) pools and was associated with elevated 24-hour posttherapy M2 and M2b protein. Panel A: SKOV3 and OVCAR3 cells were treated with cisplatin (5 μM) and/or 3-AP (5 μM) for 6 hours and assayed for dCTP pool by a DNA polymerase extension assay. Cisplatin treatment elevated 6-hour post therapy dCTP levels, indirectly suggesting a rise in RNR activity. dCTP levels 6 hours after cisplatin treatment (and 18 hours after start of 3-AP treatment) were indistinguishable from dCTP levels in cisplatin alone treated cells (P = 1.0). When 3-AP was administered alone or co-administered with cisplatin, substantial reduction in dCTP levels were detected 6 hours post therapy (P < 0.001, star). Means (± standard error) are reported. Panel B: Immunoblots for M2 and M2b protein and corresponding dCTP level are depicted for control and treated cells (t = 0 h [before] and 24 h [after start of cisplatin]). M2 and M2b showed moderate increase after cisplatin treatment or after ribonucleotide reductase blockade by 3-AP. Corresponding 24-hour dCTP level after cisplatin, an indicator of rise in ribonucleotide reductase activity in response to cisplatin-mediated DNA damage, was higher than baseline (P < 0.01, star). Increased ribonucleotide reductase activity occurred in the 3-AP then cisplatin treatment (P < 0.01, star). In the three 3-AP treatment groups, 24-hour recovery of ribonucleotide reductase activity after 3-AP inactivation was found. Whether return to baseline or elevated recovery of activity happens because of transcriptional replacement of the M2 and/or M2b subunits or other mechanism is not discerned. Means (± standard error) are reported for RNR activity.
Figure 2
Figure 2
3-aminopyridine-2-carboxyaldehyde-thiosemicarbazone (3-AP) protracts resolution of γH2AX foci. Panel A: Manual counts (mean ± standard error) of retained γH2AX foci at 24 hours post therapy (i.e., start of cisplatin exposure) are depicted for SKOV3 and OVCAR3 treated cells. Cisplatin damages DNA resulting in visible γH2AX foci [19]. Cisplatin plus 3-AP treated cells demonstrated an increased number of γH2AX foci, compared to cisplatin alone (P < 0.001, star) or 3-AP preceding cisplatin (P < 0.001, star) treatment, indicative of retained cisplatin-mediated DNA damage.
Figure 3
Figure 3
3-aminopyridine-2-carboxyaldehyde-thiosemicarbazone (3-AP) effectively restored cisplatin sensitivity in “platinum-resistant” SKOV3 and OVCAR3 ovarian cancer cells. Panel A: Cells were treated with cisplatin (5 μM) and/or 3-AP (5 μM) for 6 hours and assayed by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) for cell mitochondrial viability at 24 hours after the start of cisplatin exposure. Compared to cisplatin alone, 3-AP alone, and 3-AP preceding cisplatin treatment, a significant cisplatin plus 3-AP interaction was found (P < 0.01, star). Panel B: 14-day clonogenic ovarian cancer cell survival was done using cisplatin (5 μM) and a wider therapeutic range of 3-AP (1, 5, or 10 μM). Here too a significant cisplatin plus 3-AP enhancement of cytotoxicity was seen (P < 0.001, star), when compared to 3-AP alone or 3-AP preceding cisplatin. Means (± standard error) are reported.
Figure 4
Figure 4
Pretherapy ovarian cancer ribonucleotide reductase (RNR) M1, M2, and M2b (p53R2) protein is associated with 3-aminopyridine-2-carboxyaldehyde-thiosemicarbazone (3-AP) then cisplatin treatment response. Panel A: Two pathologists blinded to treatment and outcome scored intensity of ribonucleotide reductase subunit staining. Corresponding best treatment responses are listed (S = stable, PD = progressive disease, PR = partial response, U = undetermined). Panel B: Progression-free survival (PFS) and overall survival are illustrated for six treated patients by 3-AP (96 mg/m2) given days 1 through 4 and cisplatin (25 mg/m2) given on days 2 and 3 every 21 days. A median 14-week PFS and median 28-week overall survival were observed.

References

    1. Armstrong D, Bundy B, Wenzel L, Huang HQ, Baergen R, Lele S, Copeland LJ, Walker JL, Burger RA. Gynecologic Oncology Group. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med. 2006;354:34–43. doi: 10.1056/NEJMoa052985.
    1. Markman M, Kennedy A, Webster K, Peterson G, Kulp B, Belinson J. Combination chemotherapy with carboplatin and docetaxel in the treatment of cancers of the ovary and fallopian tube and primary carcinoma of the peritoneum. J Clin Oncol. 2001;19:1901–1905.
    1. Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, Burger RA, Mannel RS, DeGeest K, Hartenbach EM, Baergen R. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2003;21:3194–3200. doi: 10.1200/JCO.2003.02.153.
    1. du Bois A, Herrstedt J, Hardy-Bessard AC, Muller HH, Harter P, Kristensen G, Joly F, Huober J, Avall-Lundquist E, Weber B, Kurzeder C, Jelic S, Pujade-Lauraine E, Burges A, Pfisterer J, Gropp M, Staehle A, Wimberger P, Jackisch C, Sehouli J. Phase III trial of carboplatin plus paclitaxel with or without gemcitabine in first-line treatment of epithelial ovarian cancer. J Clin Oncol. 2010;28:4162–4169. doi: 10.1200/JCO.2009.27.4696.
    1. Brewer CA, Blessing JA, Nagourney RA, Morgan M, Hanjani P. Cisplatin plus gemcitabine in platinum-refractory ovarian or peritoneal cancer: a phase II study of the Gynecologic Oncology Group. Gynecol Oncol. 2006;103:446–450. doi: 10.1016/j.ygyno.2006.03.018.
    1. Peng P, Shen K, Yang JX, Wu M, Huang HF, Pan LY, Lang JH. Phase II study of gemcitabine combined with platinum chemotherapy for recurrent epithelial ovarian cancer. Chin Med Sci J. 2007;22:177–182.
    1. Kunos CA, Waggoner S, Von Gruenigen V, Eldermire E, Pink J, Dowlati A, Kinsella TJ. Phase I trial of intravenous 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, NSC #663249) in combination with pelvic radiation therapy and weekly cisplatin chemotherapy for locally advanced cervical cancer. Clin Cancer Res. 2010;16:1298–1306. doi: 10.1158/1078-0432.CCR-09-2469.
    1. Dueñas-González A, Zárba J, Patel F, Alcedo JC, Beslija S, Casanova L, Pattaranutaporn P, Hameed S, Blair JM, Barraclough H, Orlando M. A phase III, open-label, randomized study comparing concurrent gemcitabine plus cisplatin and radiation followed by adjuvant gemcitabine and cisplatin versus concurrent cisplatin and radiation in patients with stage IIB to IVA carcinoma of the cervix. J Clin Oncol. 2011;29:1678–1685. doi: 10.1200/JCO.2009.25.9663.
    1. Rose P, DeGeest K, McMeekin S, Fusco Nl. A phase I study of gemcitabine followed by cisplatin concurrent with whole pelvic radiation therapy in locally advanced cervical cancer: A Gynecologic Oncology Group study. Gynecol Oncol. 2007;107:274–279. doi: 10.1016/j.ygyno.2007.06.012.
    1. Kunos C, Radivoyevitch T, Pink J, Chiu SM, Stefan T, Jacobberger J, Kinsella TJ. Ribonucleotide reductase inhibition enhances chemoradiosensitivity of human cervical cancers. Radiation Res. 2010;174:574–581. doi: 10.1667/RR2273.1.
    1. Kunos CA, Chiu SM, Pink J, Kinsella TJ. Modulating radiation resistance by inhibiting tribonucleotide reductase in cancers with virally or mutationally silenced p53 protein. Radiation Res. 2009;172:666–676. doi: 10.1667/RR1858.1.
    1. Kunos C, Ferris G, Pyatka N, Pink J, Radivoyevitch T. Deoxynucleoside salvage facilitates DNA repair during tibonucleotide reductase blockade in human cervical cancers. Radiat Res. 2011;176:425–433. doi: 10.1667/RR2556.1.
    1. Schöndorf T, Neumann R, Benz C, Becker M, Riffelmann M, Göhring UJ, Sartorius J, von König CH, Breidenbach M, Valter MM, Hoopman M, Di Nicolantonio F, Kurbacher CM. Cisplatin, doxorubicin and paclitaxel induce mdr1 gene transcription in ovarian cancer cell lines. Recent Results Cancer Res. 2003;161:111–116. doi: 10.1007/978-3-642-19022-3_10.
    1. Lemos C, Kathmann I, Giovannetti E, Beliën JAM, Scheffer GL, Calhau C, Jansen G, Peters GJ. Cellular folate status modulates the expression of BCRP and MRP multidrug transporters in cancer cell lines from different origins. Mol Cancer Ther. 2009;8:655–664. doi: 10.1158/1535-7163.MCT-08-0768.
    1. Yen Y, Margolin K, Doroshow J, Fishman M, Johnson B, Clairmont C, Sullivan D, Sznol M. A phase I trial of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone in combination with gemcitabine for patients with advanced cancer. Cancer Chemother Pharmacol. 2004;54:331–342.
    1. Murren J, Modiano M, Clairmont C, Lambert P, Savaraj N, Doyle T, Sznol M. Phase I and pharmacokinetic study of triapine, a potent ribonucleotide reductase inhibitor, administered daily for five days in patients with advanced solid tumors. Clin Cancer Res. 2003;9:4092–4100.
    1. Feun L, Modiano M, Lee K, Mao J, Marini A, Savaraj N, Plezia P, Almassian B, Colacino E, Fischer J, MacDonald S. Phase I and pharmacokinetic study of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) using a single intravenous dose schedule. Cancer Chemother Pharmacol. 2002;50:223–229. doi: 10.1007/s00280-002-0480-0.
    1. Wadler S, Makower D, Clairmont C, Lambert P, Fehn K, Sznol M. Phase I and pharmacokinetic study of the ribonucleotide reductase inhibitor, 3-aminopyridine-2-carboxaldehyde thiosemicarbazone, administered by 96-hour intravenous continuous infusion. J Clin Oncol. 2004;22:1553–1563. doi: 10.1200/JCO.2004.07.158.
    1. Olive P, Banáth J. Kinetics of H2AX phosphorylation after exposure to cisplatin. Cytometry B Clin Cytol. 2009;76:79–90.
    1. Olive P, Banáth J. Phosphorylation of histone H2AX as a measure of radiosensitivity. Int J Radiation Oncol Biol Phys. 2004;58:331–335. doi: 10.1016/j.ijrobp.2003.09.028.
    1. Mansouri A, Lurie AA. Concise review: methemoglobinemia. Amer J Hematol. 1993;42:7–12. doi: 10.1002/ajh.2830420104.
    1. Radivoyevitch T, Taverna P, Schupp J. et al.The linear-quadratic log-survival radiation dose response model: Confidence ellipses, drug-drug interactions, and brachytherapeutic gains. Med Hypotheses Res. 2004;1:23–28.
    1. Reece SY, Hodgkiss JM, Stubbe J, Nocera DG. Proton-coupled electron transfer: the mechanisitc underpinning for radical transport and catalysis in biology. Phil Trans R Soc B. 2006;361:1351–1364. doi: 10.1098/rstb.2006.1874.
    1. Hreshchyshyn MM, Aron BS, Boronow RC, Franklin EW, Shingleton HM, Blessing JA. Hydroxyurea or placebo combined with radiation to treat stages IIIB and IV cervical cancer confined to the pelvis. Int J Radiat Oncol Biol Phys. 1979;5:317–322. doi: 10.1016/0360-3016(79)91209-4.
    1. Whitney CW, Sause W, Bundy BN, Malfetano JH, Hannigan EV, Fowler WC, Clarke-Pearson DL, Liao SY. Randomized comparison of fluorouracil plus cisplatin versus hydroxyurea as an adjunct to radiation therapy in stage IIB-IVA carcinoma of the cervix with negative para-aortic lymph nodes: a Gynecologic Oncology Group and Southwest Oncology Group study. J Clin Oncol. 1999;17:1339–1348.
    1. Rose P, Ali S, Watkins E, Thigpen JT, Deppe G, Clarke-Pearson DL, Insalaco S. Gynecologic Oncology Group. Long-term follow-up of a randomized trial comparing concurrent single agent cisplatin, cisplatin-based combination chemotherapy, or hydroxyurea during pelvic irradiation for locally advanced cervical cancer: A Gynecologic Oncology Group Study. J Clin Oncol. 2007;25:2804–2810. doi: 10.1200/JCO.2006.09.4532.
    1. Schelman W, Morgan-Meadows S, Marnocha R, Lee F, Eickhoff J, Huang W, Pomplun M, Jiang Z, Alberti D, Kolesar JM, Ivy P, Wilding G, Traynor AM. A phase I study of Triapine in combination with doxorubicin in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2009;63:1147–1156. doi: 10.1007/s00280-008-0890-8.
    1. Traynor AM, Lee JW, Bayer GK, Tate JM, Thomas SP, Mazurczak M, Graham DL, Kolesar JM, Schiller JH. A phase II trial of triapine (NSC #663249) and gemcitabine as second line treatment of advanced non-small cell lung cancer: Eastern Cooperative Oncology Group Study 1503. Invest New Drugs. 2010;28:91–97. doi: 10.1007/s10637-009-9230-z.
    1. Swinnen LJ, Rankin C, Carraway H, Albain KS, Townsend JJ, Budd GT, Kish JA, Rivkin SE, Blumenthal DT. A phase II study of cisplatin preceded by a 12-h continuous infusion of concurrent hydroxyurea and cytosine arabinoside (Ara-C) for adult patients with malignant gliomas (Southwest Oncology Group S9149) J Neurooncol. 2008;86:353–358. doi: 10.1007/s11060-007-9483-3.
    1. Piver M, Khalil M, Emrich L. Hydroxyurea plus pelvic irradiation versus placebo plus pelvic irradiation in nonsurgically staged stage IIIB cervical cancer. J Surg Oncol. 1989;42:120–125. doi: 10.1002/jso.2930420211.
    1. Chabes A, Thelander L. Controlled protein degradation regulates ribonucleotide reductase activity in proliferating mammalian cells during the normal cell cycle and in response to DNA damage and replication blocks. J Biol Chem. 2000;275:17747–17753. doi: 10.1074/jbc.M000799200.
    1. Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4:307–320. doi: 10.1038/nrd1691.
    1. Chaston TB, Lovejoy DB, Watts RN, Richardson DR. Examination of the antiproliferative activity of iron chelators: multiple cellular targets and the different mechanism of action of triapine compared with desferrioxamine and the potent pyridoxal isonicotinoyl hydrazone analogue 311. Clin Cancer Res. 2003;9(1):402–414.
    1. Popović-Bijelić A, Kowol CR, Lind ME, Luo J, Himo F, Enyedy EA, Arion VB, Gräslund A. Ribonucleotide reductase inhibition by metal complexes of Triapine (3-aminopyridine-2-carboxaldehyde thiosemicarbazone): A combined experimental and theoretical study. J Inorg Biochem. 2011;105(11):1422–1431. doi: 10.1016/j.jinorgbio.2011.07.003.
    1. do Nascimento TS, Pereira RO, de Mello HL, Costa J. Methemoglobinemia: from diagnosis to treatment. Rev Bras Anestesiol. 2008;58:651–64. doi: 10.1590/S0034-70942008000600011.

Source: PubMed

3
S'abonner