27
Views
0
CrossRef citations to date
0
Altmetric
Review

The possibility of dexrazoxane to prevent ovarian damage caused by toxicity

, &
Pages 269-275 | Received 17 Feb 2016, Accepted 18 May 2016, Published online: 10 Jun 2016

References

  • Salih SM, Elsarrag SZ, Prange E, et al. Evidence to incorporate inclusive reproductive health measures in guidelines for childhood and adolescent cancer survivors. J Pediatr Adolesc Gynecol. 2015;28(2):95–101.
  • Woodruff TK. The oncofertility consortium-addressing fertility in young people with cancer. Nat Rev Clin Oncol. 2010;7(8):466–475.
  • Anderson RA, Nelson SM, Wallace WHB. Measuring anti-Mullerian hormone for the assessment of ovarian reserve: when and for whom is it indicated? Maturitas. 2012;71(1):28–33.
  • Dunlop CE, Anderson RA. Uses of anti-Mullerian hormone (AMH) measurement before and after cancer treatment in women. Maturitas. 2015;80(3):245–250.
  • Brougham MFH, Crofton PM, Johnson EJ, et al. Anti-Mullerian Hormone is a marker of gonadotoxicity in pre- and postpubertal girls treated for cancer: a prospective study. J Clin Endocrinol Metabolism. 2012;97(6):2059–2067.
  • Meirow D, Biederman H, Anderson RA, et al. Toxicity of chemotherapy and radiation on female reproduction. Clin Obstet Gynecol. 2010;53(4):727–739.
  • Perez GI, Knudson CM, Leykin L, et al. Apoptosis-associated signaling pathways are required for chemotherapy-mediated female germ cell destruction. Nat Med. 1997;3(11):1228–1232.
  • Jurisicova A, Lee HJ, D’Estaing SG, et al. Molecular requirements for doxorubicin-mediated death in murine oocytes. Cell Death Differ. 2006;13(9):1466–1474.
  • Bar-Joseph H, Ben-Aharon I, Rizel S, et al. Doxorubicin-induced apoptosis in germinal vesicle (GV) oocytes. Reprod Toxicol. 2010;30(4):566–572.
  • Ben-Aharon I, Bar-Joseph H, Tzarfaty G, et al. Doxorubicin-induced ovarian toxicity. Reprod Biol Endocrinol. 2010;8:7.
  • Roti ECR, Leisman SK, Abbott DH, et al. Acute doxorubicin insult in the mouse ovary is cell- and follicle-type dependent. PLoS One. 2012;7(8):e42293.
  • Loren AW, Mangu PB, Beck LN, et al. Fertility preservation for patients with cancer: American society of clinical oncology clinical practice guideline update. J Clin Oncol. 2013;31(19):2500.
  • Roness H, Kalich-Philosoph L, Meirow D. Prevention of chemotherapy-induced ovarian damage: possible roles for hormonal and non-hormonal attenuating agents. Hum Reprod Update. 2014;20(5):759–774.
  • Roness H, Kashi O, Meirow D. Prevention of chemotherapy-induced ovarian damage. Fertil Steril. 2016;105(1):20–29.
  • Del Mastro L, Lambertini M. Temporary ovarian suppression with gonadotropin-releasing hormone agonist during chemotherapy for fertility preservation: toward the end of the debate? Oncologist. 2015;20(11):1233–1235.
  • Del Mastro L, Rossi G, Lambertini M, et al. New insights on the role of luteinizing hormone releasing hormone agonists in premenopausal early breast cancer patients. Cancer Treat Rev. 2016;42:18–23.
  • Gris-Martinez JM, Trillo-Urrutia L, Gomez-Cabeza JJ, et al. Protective effect of GnRH analogues on the reproductive capacity of women with neoplasia or autoimmune disease who require chemotherapy. Final results of a phase ii clinical trial. Med Clin (Barc). 2016;146(3):97–103.
  • Lambertini M, Ceppi M, Poggio F, et al. Ovarian suppression using luteinizing hormone-releasing hormone agonists during chemotherapy to preserve ovarian function and fertility of breast cancer patients: a meta-analysis of randomized studies. Ann Oncol. 2015;26(12):2408–2419.
  • Lambertini M, Poggio F, Levaggi A, et al. Protecting ovaries during chemotherapy through gonad suppression: a systematic review and meta-analysis. Obstet Gynecol. 2015;126(4):901.
  • Lambertini M, Del Mastro L, Pescio MC, et al. Cancer and fertility preservation: international recommendations from an expert meeting. BMC Med. 2016;14(1):1.
  • Moore HC, Unger JM, Phillips KA, et al. Goserelin for ovarian protection during breast-cancer adjuvant chemotherapy. N Engl J Med. 2015;372(10):923–932.
  • Moore HC, Unger JM, Albain KS. Ovarian protection during adjuvant chemotherapy. N Engl J Med. 2015;372(23):2269–2270.
  • Blumenfeld Z, Zur H, Dann EJ. Gonadotropin-releasing hormone agonist cotreatment during chemotherapy may increase pregnancy rate in survivors. Oncologist. 2015;20(11):1283–1289.
  • Blumenfeld Z, Evron A. Preserving fertility when choosing chemotherapy regimens - the role of gonadotropin-releasing hormone agonists. Expert Opin Pharmacother. 2015;16(7):1009–1020.
  • Huser M, Smardova L, Janku P, et al. Fertility status of Hodgkin lymphoma patients treated with chemotherapy and adjuvant gonadotropin-releasing hormone analogues. J Assist Reprod Genet. 2015;32(8):1187–1193.
  • Bildik G, Akin N, Senbabaoglu F, et al. GnRH agonist leuprolide acetate does not confer any protection against ovarian damage induced by chemotherapy and radiation in vitro. Hum Reprod. 2015;30(12):2912–2925.
  • Sun X, Dongol S, Jiang J, et al. Protection of ovarian function by GnRH agonists during chemotherapy: a meta-analysis. Int J Oncol. 2014;44(4):1335–1340.
  • Shen YW, Zhang XM, Lv M, et al. Utility of gonadotropin-releasing hormone agonists for prevention of chemotherapy-induced ovarian damage in premenopausal women with breast cancer: a systematic review and meta-analysis. Onco Targets Ther. 2015;8:3349–3359.
  • Horicks F, Van Den Steen G, Houben S, et al. Folliculogenesis is not fully inhibited during GnRH analogues treatment in mice challenging their efficiency to preserve the ovarian reserve during chemotherapy in this model. PLoS One. 2015;10(9):e0137164.
  • Elgindy E, Sibai H, Abdelghani A, et al. Protecting ovaries during chemotherapy through gonad suppression: a systematic review and meta-analysis. Obstet Gynecol. 2015;126(1):187–195.
  • Parlakgumus HA, Kilicdag EB, Bolat FA, et al. GNRH agonists and antagonists in rescue for cyclophosphamide-induced ovarian damage: friend or foe? Arch Gynecol Obstet. 2015;291(6):1403–1410.
  • Karimi-Zarchi M, Forat-Yazdi M, Vafaeenasab MR, et al. Evaluation of the effect of GnRH agonist on menstrual reverse in breast cancer cases treated with cyclophosphamide. Eur J Gynaecol Oncol. 2014;35(1):59–61.
  • Wallace WH, Kelsey TW, Anderson RA. Fertility preservation in pre-pubertal girls with cancer: the role of ovarian tissue cryopreservation. Fertil Steril. 2016;105(1):6–12.
  • Minotti G, Menna P, Salvatorelli E, et al. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004;56(2):185–229.
  • Doroshow JH. Dexrazoxane for the prevention of cardiac toxicity and treatment of extravasation injury from the anthracycline antibiotics. Curr Pharm Biotechnol. 2012;13(10):1949–1956.
  • Gewirtz DA. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics Adriamycin and daunorubicin. Biochem Pharmacol. 1999;57(7):727–741.
  • Sterba M, Popelova O, Vavrova A, et al. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxid Redox Signal. 2013;18(8):899–929.
  • Beretta GL, Zunino F. Molecular mechanisms of anthracycline activity. Top Curr Chem. 2008;283:1–19.
  • Vavrova A, Jansova H, Mackova E, et al. Catalytic inhibitors of topoisomerase II differently modulate the toxicity of anthracyclines in cardiac and cancer cells. Plos One. 2013;8:10.
  • Andoh T. Bis(2,6-dioxopiperazines), catalytic inhibitors of DNA topoisomerase II, as molecular probes, cardioprotectors and antitumor drugs. Biochimie. 1998;80(3):235–246.
  • Lyu YL, Kerrigan JE, Lin C-P, et al. Topoisomerase II beta-Mediated DNA double-strand breaks: Implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Res. 2007;67(18):8839–8846.
  • Deng S, Yan T, Jendrny C, et al. Dexrazoxane may prevent doxorubicin-induced DNA damage via depleting both topoisomerase II isoforms. BMC Cancer. 2014;14:842.
  • Van Dalen EC, Caron HN, Dickinson HO, et al. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database System Rev. 2011;6:CD003917.
  • Classen S, Olland S, Berger JM. Structure of the topoisomerase II ATPase region and its mechanism of inhibition by the chemotherapeutic agent ICRF-187. Proc Natl Acad Sci U S A. 2003;100(19):10629–10634.
  • Weiss G, Loyevsky M, Gordeuk VR. Dexrazoxane (ICRF-187). Gen Pharmacol. 1999;32(1):155–158.
  • Swain SM, Whaley FS, Gerber MC, et al. Cardioprotection with dexrazoxane for doxorubicin-containing therapy in advanced breast cancer. J Clin Oncol. 1997;15(4):1318–1332.
  • Tebbi CK, London WB, Friedman D, et al. Dexrazoxane-associated risk for acute myeloid leukemia/myelodysplastic syndrome and other secondary malignancies in pediatric Hodgkin’s disease. J Clin Oncol. 2007;25(5):493–500.
  • Salzer WL, Devidas M, Carroll WL, et al. Long-term results of the pediatric oncology group studies for childhood acute lymphoblastic leukemia 1984-2001: a report from the children’s oncology group. Leukemia. 2010;24(2):355–370.
  • Schwartz CL, Wexler LH, Krailo MD, et al. Intensified chemotherapy with dexrazoxane cardioprotection in newly diagnosed nonmetastatic osteosarcoma: a report from the children’s oncology group. Pediatr Blood Cancer. 2016;63(1):54–61.
  • Shaikh F, Dupuis LL, Alexander S, et al. Cardioprotection and second malignant neoplasms associated with dexrazoxane in children receiving anthracycline chemotherapy: a systematic review and meta-analysis. J Natl Cancer Inst. 2016;108:4.
  • Chow EJ, Asselin BL, Schwartz CL, et al. Late mortality after dexrazoxane treatment: a report from the children’s oncology group. J Clin Oncol. 2015;33(24):2639–U2690.
  • Asselin BL, Devidas M, Chen L, et al. Cardioprotection and safety of dexrazoxane in patients treated for newly diagnosed T-cell acute lymphoblastic leukemia or advanced-stage lymphoblastic non-Hodgkin lymphoma: a report of the children’s oncology group randomized trial pediatric oncology group 9404. J Clin Oncol. 2016;34(8):854–862.
  • Salih SM, Ringelstetter AK, Elsarrag MZ, et al. Dexrazoxane abrogates acute doxorubicin toxicity in marmoset ovary. Biol Reprod. 2015;92(3):11.
  • Roti ECR, Salih SM. Dexrazoxane ameliorates doxorubicin-induced injury in mouse ovarian cells. Biol Reprod. 2012;86:3.
  • Kropp J, Roti ECR, Ringelstetter A, et al. Dexrazoxane diminishes doxorubicin-induced acute ovarian damage and preserves ovarian function and fecundity in mice. PLoS One. 2015;10:11.
  • Levi M, Tzabari M, Savion N, et al. Dexrazoxane exacerbates doxorubicin-induced testicular toxicity. Reproduction. 2015;150(4):357–366.
  • Attia SM, Ahmad SF, Bakheet SA. Impact of dexrazoxane on doxorubicin-induced aneuploidy in somatic and germinal cells of male mice. Cancer Chemother Pharmacol. 2016;77(1):27–33.
  • Har-Vardi I, Mali R, Breietman M, et al. DNA topoisomerases I and II in human mature sperm cells: characterization and unique properties. Hum Reprod. 2007;22(8):2183–2189.
  • St Pierre J, Wright DJ, Rowe TC, et al. DNA topoisomerase II distribution in mouse preimplantation embryos. Mol Reprod Dev. 2002;61(3):335–346.
  • Meyer-Ficca ML, Lonchar JD, Ihara M, et al. Poly(ADP-Ribose) polymerases PARP1 and PARP2 modulate topoisomerase II beta (TOP2B) function during chromatin condensation in mouse spermiogenesis. Biol Reprod. 2011;84(5):900–909.
  • Zhang YL, Yu C, Ji SY, et al. TOP2 beta is essential for ovarian follicles that are hypersensitive to chemotherapeutic drugs. Mol Endocrinol. 2013;27(10):1678–1691.
  • Deng SW, Yan TD, Nikolova T, et al. The catalytic topoisomerase II inhibitor dexrazoxane induces DNA breaks, ATF3 and the DNA damage response in cancer cells. Br J Pharmacol. 2015;172(9):2246–2257.
  • Wang JC. Cellular roles of DNA topoisomerases: A molecular perspective. Nat Rev Mol Biol. 2002;3(6):430–440.
  • Parker AS, Eckel-Passow JE, Serie D, et al. Higher expression of topoisomerase II alpha is an independent marker of increased risk of cancer-specific death in patients with clear cell renal cell carcinoma. Eur Urol. 2014;66(5):929–935.
  • Zhang S, Liu XB, Bawa-Khalfe T, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 2012;18(11):1639.
  • Jirkovska-Vavrova A, Roh J, Lencova-Popelova O, et al. Synthesis and analysis of novel analogues of dexrazoxane and its open-ring hydrolysis product for protection against anthracycline cardiotoxicity in vitro and in vivo. Toxicol Res. 2015;4(4):1098–1114.
  • Lencova-Popelova O, Jirkovsky E, Jansova H, et al. Cardioprotective effects of inorganic nitrate/nitrite in chronic anthracycline cardiotoxicity: comparison with dexrazoxane. J Mol Cell Cardiol. 2016;91:92–103.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.