99
Views
2
CrossRef citations to date
0
Altmetric
Review

Potential pharmacological interventions against hematotoxicity: an overview

&

References

  • Bloom JC, Brandt JT. Toxic responses of blood. Casarett’s and Doull’s toxicology: The basic science of poisons. The McGraw-Hill Companies; USA: 2008p. 455-84
  • Smith EP. Hematologic disorders after solid organ transplantation. Hematology Am Soc Hematol Educ Program 2010;1:281-6
  • Verrotti A, Scaparrotta A, Grosso S, et al. Anticonvulsant drugs and hematological disease. Neurol Sci 2014;35(7):983-93
  • Berger I, Seqal I, Shmueli D, et al. The effect of antiepileptic drugs on mitochondrial activity: a pilot study. J Child Neurol 2010;25(5):541-5
  • Orasan O, Cozma A, Rednic N, et al. Anemia – a complication of antiviral treatment in chronic viral hepatitis C. Rom J Intern Med 2009;47(3):217-25
  • Gribaldo L, Malerba I, Collotta A, et al. Inhibition of CFU-E/BFU-E by 3′-azido-3′-deoxythymidine, chlorpropamide, and protoporphirin IX zinc (II): a comparison between direct exposure of progenitor cells and long-term exposure of bone marrow cultures. Toxicol Sci 2000;58(1):96-101
  • de Vocht F, Vlaanderen J, Povey AC, et al. Environmental and occupational toxicants. IARC Sci Publ 2011;163-163:74
  • Umarani V, Muvvala S, Ramesh A, et al. Rutin potentially attenuates fluoride induced oxidative stress mediated cardiotoxicity, blood toxicity and dyslipidemia in rats. Toxicol Mech Methods 2015. [Epub ahead of print]
  • Van Den Heuvel RL, Leppens H, Schoeters GE. Use of in vitro assays to assess hematotoxic effects of environmental compounds. Cell Biol Toxicol 2001;17(2):107-16
  • Graeme KA, Pollack CV. Heavy metal toxicity, Part I: arsenic and mercury. J Emerg Med 1998;16(1):45-56
  • Ramadhas M, Palanisamy K, Sudhagar M, et al. Ameliorating effect of Phoenix dactylifera on lambda cyhalothrin induced biochemical, hematological and hepatopathological alterations in male wistar rats. Biomed Aging Pathol 2014;4(3):273-9
  • Chatterjee S, Basak P, Chaklader M, et al. Pesticide induced alterations in marrow physiology and depletion of stem and stromal progenitor population: an experimental model to study the toxic effects of pesticide. Environ Toxicol 2014;29(1):84-97
  • Halfdanarson TR, Litzow MR, Murray JA. Hematologic manifestations of celiac disease. Blood 2007;109(2):412-21
  • Albella B, Faircloth G, López-Lázaro L, et al. In vitro toxicity of ET-743 and aplidine, two marine-derived antineoplastics, on human bone marrow haematopoietic progenitors. comparison with the clinical results. Eur J Cancer 2002;38(10):1395-404
  • Ziepert M, Schmits R, Trümper L, et al. Prognostic factors for hematotoxicity of chemotherapy in aggressive non-Hodgkin’s lymphoma. Ann Oncol 2008;19(4):752-62
  • Moiseeva I, Zinov’ev AI, Ionicheva LV, et al. Comparative study of the hemato-protective effect of Dicarbamine and Leykostime in experimental radiation damage to the blood system. Vopr Onkol 2013;59(2):71-4
  • Tannehill SP, Mehta MP. Amifostine and radiation therapy: past, present, and future. Semin Oncol 1996;23(4 Suppl 8):69-77
  • Kurbacher CM, Mallmann PK. Chemoprotection in anticancer therapy: the emerging role of amifostine (WR-2721). Anticancer Res 1998;18(3C):2203-10
  • Lv W, Zhang M, Zhang Z, et al. Amifostine acts upon mitochondria to stimulate growth of bone marrow and regulate cytokines. Adv Exp Med Biol 2013;789:195-201
  • Dobrenis K, Gauthier LR, Barroca V, et al. G-CSF off-target effect on nerve outgrowth promotes prostate cancer development. Int J Cancer 2015;136(4):982-8
  • Makita K, Ohta K, Mugitani A, et al. Acute myelogenous leukemia in a donor after granulocyte colony-stimulating factor-primed peripheral blood stem cell harvest. Bone Marrow Transplant 2004;33(6):661-5
  • Pentz RD. Healthy sibling donation of G-CSF primed stem cells: a call for research. Pediatr Blood Cancer 2006;46(4):407-8
  • Donadieu J. Is G-CSF-mobilized peripheral stem cell harvest harmful? Pediatr Blood Cancer 2007;48(5):594-5
  • Sytkowski AJ. Does erythropoietin have a dark side? Epo signaling and cancer cells. Sci STKE 2007;395:38
  • Lapierre A, Souquet PJ. Use of erythropoiesis stimulating agents. Rev Mal Respir 2014;31(2):162-72
  • Pratheeshkumar P, Budhraja A, Son YO, et al. Quercetin inhibits angiogenesis mediated human prostate tumor growth by targeting VEGFR- 2 regulated AKT/mTOR/P70S6K signaling pathways. PLoS One 2012;7(10):e47516
  • Larocca LM, Teofili L, Leone G, et al. Antiproliferative activity of quercetin on normal bone marrow and leukaemic progenitors. Br J Haematol 1991;79(4):562-6
  • Kim H, Moon JY, Ahn KS, et al. Quercetin induces mitochondrial mediated apoptosis and protective autophagy in human glioblastoma U373MG cells. Oxid Med Cell Longev 2013;2013:596496
  • Youn H, Jeong JC, Jeong YS, et al. Quercetin potentiates apoptosis by inhibiting nuclear factor-kappaB signaling in H460 lung cancer cells. Biol Pharm Bull 2013;36(6):944-51
  • Wang H, Yuan Z, Chen Z, et al. Effect of quercetin on glioma cell U87 apoptosis and feedback regulation of MDM2-p53. Nan Fang Yi Ke Da Xue Xue Bao 2014;34(5):686-9
  • Nabavi SM, Nabavi SF, Eslami S, et al. In vivo protective effects of quercetin against sodium fluoride-induced oxidative stress in the hepatic tissue. Food Chem 2012;132(2):931-5
  • Tabaczar S, Pieniążek A, Czepas J, et al. Quercetin attenuates oxidative stress in the blood plasma of rats bearing DMBA-induced mammary cancer and treated with a combination of doxorubicin and docetaxel. Gen Physiol Biophys 2013;32(4):535-43
  • Orsolic N, Car N. Quercetin and hyperthermia modulate cisplatin-induced DNA damage in tumor and normal tissues in vivo. Tumour Biol 2014;35(7):6445-54
  • Carl H, Chandni A, Neha K, et al. Curcumin as a modulator of oxidative stress during storage: a study on plasma. Transfus Apher Sci 2014;50(2):288-93
  • El-Bahr SM. Curcumin regulates gene expression of insulin like growth factor, B-cell CLL/lymphoma 2 and antioxidant enzymes in streptozotocin induced diabetic rats. BMC Complement Altern Med 2013;13:368
  • Gao S, Duan X, Wang X, et al. Curcumin attenuates arsenic-induced hepatic injuries and oxidative stress in experimental mice through activation of Nrf2 pathway, promotion of arsenic methylation and urinary excretion. Food Chem Toxicol 2013;59:739-47
  • Nabavi SF, Daglia M, Moghaddam AH, et al. Curcumin and liver disease: from chemistry to medicine. Comprehensive Rev Food Sci Food Safety 2014;13(1):62-77
  • Anitha A, Sreeranganathan M, Chennazhi KP, et al. In vitro combinatorial anticancer effects of 5-fluorouracil and curcumin loaded N,O-carboxymethyl chitosan nanoparticles toward colon cancer and in vivo pharmacokinetic studies. Eur J Pharm Biopharm 2014;88(1):238-51
  • Palange AL, Di Mascolo D, Carallo C, et al. Lipid-polymer nanoparticles encapsulating curcumin for modulating the vascular deposition of breast cancer cells. Nanomedicine 2014;10(5):991-1002
  • Garufi A, Trisciuoglio D, Porru M, et al. A fluorescent curcumin-based Zn(II)-complex reactivates mutant (R175H and R273H) p53 in cancer cells. J Exp Clin Cancer Res 2013;32(1):72
  • Storka A, Vcelar B, Klickovic U, et al. Effect of liposomal curcumin on red blood cells in vitro. Anticancer Res 2013;33(9):3629-34
  • Deng SL, Chen WF, Zhou B, et al. Protective effects of curcumin and its analogues against free radical-induced oxidative haemolysis of human red blood cells. Food Chem 2006;98(1):112-19
  • Yunos NM, Beale P, Yu JQ, et al. Synergism from sequenced combinations of curcumin and epigallocatechin-3-gallate with cisplatin in the killing of human ovarian cancer cells. Anticancer Res 2011;31(4):1131-40
  • Day RM, Davis TA, Barshishat-Kupper M, et al. Enhanced hematopoietic protection from radiation by the combination of genistein and captopril. Int Immunopharmacol 2013;15(2):348-56
  • Singh VK, Grace MB, Parekh VI, et al. Effects of genistein administration on cytokine induction in whole-body gamma irradiated mice. Int Immunopharmacol 2009;9(12):1401-10
  • Colado Simao AN, Suzukawa AA, Casado MF, et al. Genistein abrogates pre-hemolytic and oxidative stress damage induced by 2,2′-Azobis (amidinopropane). Life Sci 2006;78(11):1202-10
  • Zhou Y, Mi MT. Genistein stimulates hematopoiesis and increases survival in irradiated mice. J Radiat Res 2005;46(4):425-33
  • Ishimi Y, Yoshida M, Wakimoto S, et al. Genistein, a soybean isoflavone, affects bone marrow lymphopoiesis and prevents bone loss in castrated male mice. Bone 2002;31(1):180-5
  • Aufderklamm S, Miller F, Galasso A, et al. Chemoprevention of prostate cancer by isoflavonoids. Recent Results Cancer Res 2014;202:101-8
  • Damle AA, Pawar YP, Narkar AA. Anticancer activity of betulinic acid on MCF-7 tumors in nude mice. Indian J Exp Biol 2013;51(7):485-91
  • Li Q, Li Y, Wang X, et al. Co-treatment with ginsenoside Rh2 and betulinic acid synergistically induces apoptosis in human cancer cells in association with enhanced capsase-8 activation, bax translocation, and cytochrome c release. Mol Carcinog 2011;50(10):760-9
  • Liu Y, Luo W. Betulinic acid induces Bax/Bak-independent cytochrome c release in human nasopharyngeal carcinoma cells. Mol Cells 2012;33(5):517-24
  • Emmerich D, Vanchanagiri K, Baratto LC, et al. Synthesis and studies of anticancer properties of lupane-type triterpenoid derivatives containing a cisplatin fragment. Eur J Med Chem 2014;75:460-6
  • Park SY, Kim HJ, Kim KR, et al. Betulinic acid, a bioactive pentacyclic triterpenoid, inhibits skeletal-related events induced by breast cancer bone metastases and treatment. Toxicol Appl Pharmacol 2014;275(2):152-62
  • Kumar S, Gautam S, Sharma A. Antimutagenic and antioxidant properties of plumbagin and other naphthoquinones. Mutat Res 2013;755(1):30-41
  • Luo P, Wong YF, Ge L, et al. Anti-inflammatory and analgesic effect of plumbagin through inhibition of nuclear factor-kappaB activation. J Pharmacol Exp Ther 2010;335(3):735-42
  • Li YC, He SM, He ZX, et al. Plumbagin induces apoptotic and autophagic cell death through inhibition of the PI3K/Akt/mTOR pathway in human non-small cell lung cancer cells. Cancer Lett 2014;344(2):239-59
  • Tian L, Yin D, Ren Y, et al. Plumbagin induces apoptosis via the p53 pathway and generation of reactive oxygen species in human osteosarcoma cells. Mol Med Rep 2012;5(1):126-32
  • Sinha S, Pal K, Elkhanany A, et al. Plumbagin inhibits tumorigenesis and angiogenesis of ovarian cancer cells in vivo. Int J Cancer 2013;132(5):1201-12
  • Kawiak A, Zawacka-Pankau J, Lojkowska E. Plumbagin induces apoptosis in Her2-overexpressing breast cancer cells through the mitochondrial-mediated pathway. J Nat Prod 2012;75(4):747-51
  • Qiu JX, He YQ, Wang Y, et al. Plumbagin induces the apoptosis of human tongue carcinoma cells through the mitochondria-mediated pathway. Med Sci Monit Basic Res 2013;19:228-36
  • Son TG, Camandola S, Arumugam TV, et al. Plumbagin, a novel Nrf2/ARE activator, protects against cerebral ischemia. J Neurochem 2010;112(5):1316-26
  • Babaei A, Arshami J, Haqhparast A, et al. Effects of saffron (Crocus sativus) petal ethanolic extract on hematology, antibody response, and spleen histology in rats. Avicenna J Phytomed 2014;4(2):103-9
  • Del Bo C, Martini D, Vendrame S, et al. Improvement of lymphocyte resistance against H(2)O(2)-induced DNA damage in Sprague–Dawley rats after eight weeks of a wild blueberry (Vaccinium angustifolium)-enriched diet. Mutat Res 2010;703(2):158-62
  • Bonarska-Kujawa D, Pruchnik H, Cyboran S, et al. Biophysical mechanism of the protective effect of blue honeysuckle (Lonicera caerulea L. var. kamtschatica Sevast.) polyphenols extracts against lipid peroxidation of erythrocyte and lipid membranes. J Membr Biol 2014;247(7):611-25
  • Nabavi SM, Nabavi SF, Setzer WN, et al. Interaction of different extracts of Primula heterochroma Stapf. with red blood cell membrane lipids and proteins: antioxidant and antihemolytic effects. J Diet Suppl 2012;9(4):285-92
  • Kropat C, Mueller D, Boettler U, et al. Modulation of Nrf2-dependent gene transcription by bilberry anthocyanins in vivo. Mol Nutr Food Res 2013;57(3):545-50
  • Mackenroth J, Hölig K, Kuhlisch E, et al. Influence of packed red cell transfusions on blood selenium concentration in pediatric hemato-oncology. Klin Padiatr 2008;220(3):153-8
  • Rahmanto AS, Davies MJ. Selenium-containing amino acids as direct and indirect antioxidants. IUBMB Life 2012;64(11):863-71
  • Richie JP, Das A, Calcagnotto AM, et al. Comparative effects of two different forms of selenium on oxidative stress biomarkers in healthy men: a randomized clinical trial. Cancer Prev Res (Phila) 2014;7(8):796-804
  • Hu YJ, Chen Y, Zhang YQ, et al. The protective role of selenium on the toxicity of cisplatin-contained chemotherapy regimen in cancer patients. Biol Trace Elem Res 1997;56(3):331-41
  • Panchuk R, Skorokhyd N, Chumak V, et al. Specific antioxidant compounds differentially modulate cytotoxic activity of doxorubicin and cisplatin: in vitro and in vivo study. Croat Med J 2014;55(3):206-17
  • Sieja K, Talerczyk M. Selenium as an element in the treatment of ovarian cancer in women receiving chemotherapy. Gynecol Oncol 2004;93(2):320-7
  • Sharma S, Raghuvanshi BP, Shukla S. Toxic effects of lead exposure in rats: involvement of oxidative stress, genotoxic effect, and the beneficial role of N-acetylcysteine supplemented with selenium. J Environ Pathol Toxicol Oncol 2014;33(1):19-32
  • Sonaa E, Usha S, Ja In J. An ex vivo study of selenium, genistein on the morphological and nuclear changes in anticancer drug-induced apoptosis in human peripheral blood lymphocytes. Biofactors 2013;39(3):279-93
  • Mohamed J, Wei WL, Husin NN, et al. Selenium supplementation reduced oxidative stress in diethylnitrosamine-induced hepatocellular carcinoma in rats. Pak J Biol Sci 2011;14(23):1055-60
  • Hu Y, Spengler ML, Kuropatwinski KK, et al. Selenium is a modulator of circadian clock that protects mice from the toxicity of a chemotherapeutic drug via upregulation of the core clock protein, BMAL1. Oncotarget 2011;2(12):1279-90
  • Taskin E, Dursun N. The protection of selenium on adriamycin-induced mitochondrial damage in rat. Biol Trace Elem Res 2012;147(1-3):165-71
  • de Rosa V, Erkekoğlu P, Forestier A, et al. Low doses of selenium specifically stimulate the repair of oxidative DNA damage in LNCaP prostate cancer cells. Free Radic Res 2012;46(2):105-16
  • Bhattacharjee A, Basu A, Ghosh P, et al. Protective effect of Selenium nanoparticle against cyclophosphamide induced hepatotoxicity and genotoxicity in Swiss albino mice. J Biomater Appl 2014;29(2):303-17

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.