Iodine and doxorubicin, a good combination for mammary cancer treatment: antineoplastic adjuvancy, chemoresistance inhibition, and cardioprotection

Yunuen Alfaro, Guadalupe Delgado, Alfonso Cárabez, Brenda Anguiano, Carmen Aceves, Yunuen Alfaro, Guadalupe Delgado, Alfonso Cárabez, Brenda Anguiano, Carmen Aceves

Abstract

Background: Although mammary cancer (MC) is the most common malignant neoplasia in women, the mortality for this cancer has decreased principally because of early detection and the use of neoadjuvant chemotherapy. Of several preparations that cause MC regression, doxorubicin (DOX) is the most active, first-line monotherapeutic. Nevertheless, its use is limited due to the rapid development of chemoresistance and to the cardiotoxicity caused by free radicals. In previous studies we have shown that supplementation with molecular iodine (I2) has a powerful antineoplastic effect in methylnitrosourea (MNU)-induced experimental models of MC. These studies also showed a consistent antioxidant effect of I2 in normal and tumoral tissues.

Methods: Here, we analyzed the effect of I2 in combination with DOX treatment in female Sprague Dawley rats with MNU-induced MC. In the first experiment (short) animals were treated with the therapeutic DOX dose (16 mg/kg) or with lower doses (8 and 4 mg/Kg), in each case with and without 0.05% I2 in drinking water. Iodine treatment began on day 0, a single dose of DOX was injected (ip) on day 2, and the analysis was carried out on day 7. In the second experiment (long) animals with and without iodine supplement were treated with one or two injections of 4 mg/kg DOX (on days 0 and 14) and were analyzed on day 56.

Results: At all DOX doses, the short I2 treatment induced adjuvant antineoplastic effects (decreased tumor size and proliferating cell nuclear antigen level) with significant protection against body weight loss and cardiotoxicity (creatine kinase MB, cardiac lipoperoxidation, and heart damage). With long-term I2, mammary tumor tissue became more sensitive to DOX, since a single injection of the lowest dose of DOX (4 mg/Kg) was enough to stop tumor progression and a second DOX4 injection on day 14 caused a significant and rapid decrease in tumor size, decreased the expression of chemoresistance markers (Bcl2 and survivin), and increased the expression of the apoptotic protein Bax and peroxisome proliferator-activated receptor type gamma.

Conclusions: The DOX-I2 combination exerts antineoplastic, chemosensitivity, and cardioprotective effects and could be a promising strategy against breast cancer progression.

Figures

Figure 1
Figure 1
Effect of molecular iodine(I2) and different doses of doxorubicin (DOX) on body weight and tumor size in MNU-induced rats. Animals with tumors (2–3 cm3) received single injections of DOX and 0.05% I2 treatment (drinking water; beginning 2 days before the DOX injections). Data are expressed as the mean ± SD (n = 10). * Indicates p < 0.05 compared to control or **compared with DOX group.
Figure 2
Figure 2
Proliferation rate. Immunohistochemical presence of PCNA-positive cells in tumors from control and DOX16-treated animals with and without iodine supplement for one week. PCNA-positive cells were revealed with diaminobenzidine (brown stain) and counterstained with hematoxylin (purple stain). Data are expressed as the mean ± SD (n = 6). Means with different letters indicate statistically significant differences (p < 0.05).
Figure 3
Figure 3
Short-term effect on gene expression. Animals with tumors (2–3 cm3) received a single injection of DOX16 and were treated with 0.05% I2 for one week. mRNA expression was measured by qPCR. β-actin mRNA was amplified to check for RNA quantity and integrity. The experiments were repeated three times with independent RNA samples. Values are expressed as mean ± SD. Means with different letters indicate statistically significant differences (p < 0.05).
Figure 4
Figure 4
Cardiac responses. CK-MB serum concentration (A), cardiac lipoperoxidation [malondialdehyde (MDA); B] and catalase activity [U* = μmoles of H2O2 consumed/min; C] were measured in MNU-treated animals injected with saline (Control) or DOX exposed or not to 0.05% I2 for one week. Values are expressed as mean ± SD. Means with different letters indicate statistically significant differences (p < 0.05). Basal group in CK-MB corresponds to control animals without MNU treatment.
Figure 5
Figure 5
Iodine antioxidant power. The antioxidant capacity of different chemical forms of iodine was analyzed with the ferric reducing/antioxidant power assay (FRAP). Lugol: solution of I2:KI (1:3). Ascorbic acid was used as positive control. Values are expressed as mean ± SD (n = 4).
Figure 6
Figure 6
Long-term effect on tumor size (% change). Animals with tumors (2–3 cm3) received one (day 0) and/or two (days 0 and 14) injections of saline (Control) or DOX4 with and without I2 supplement, and tumor size was measured at 56 days. Single injection on day 0 of DOX4 (DOX4); I2 alone (I2); DOX4 + I2 (DOX4/I2); DOX4 + second injection (day 14) of DOX4 (DOX4 + DOX4); I2 with only the day 14 injection of DOX4 (I2 + DOX4), and DOX/I2 with the second injection of DOX4 on day 14 (DOX4/I2 + DOX4). Vertical arrows indicated DOX4 injections days. * Indicates p < 0.05 compared to control, **compared with I2 and DOX4/I2 groups.
Figure 7
Figure 7
Long-term I2 effect on gene expression. Animals with tumors (2–3 cm3) received two injections of saline (Control) or DOX4 without (DOX4 + DOX4) or with continuous I2 supplement (DOX4/I2 + DOX4) for 56 days. mRNA expression was measured by qPCR. β-actin mRNA was amplified to check for RNA quantity and integrity. The experiments were repeated three times with independent RNA samples. Values are expressed as mean ± SD. Means with different letters are statistically significantly (p < 0.05).

References

    1. Goldhirsch A, Wood WC, Senn H-J, Glick GH, Gelbe RD. International consensus panel on the treatment of primary breast cancer. Eur J Cancer. 1995;31:1754–1759.
    1. Wang J, Song Y, Xu S, Zhang Q, Li Y, Tang D, Jin S. Down-regulation of ICBP90 contributes to doxorubicin resistance. Eur J Pharma. 2011;656:33–38.
    1. Weiss RB. The anthracyclines: will we ever find a better doxorubicin? Semin Oncol. 1992;19:670–686.
    1. Gewirtz D. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem Pharmacol. 1999;57:727–741.
    1. Deavall DG, Martin EA, Horner JM, Roberts R. Drug-induced oxidative stress and toxicity. J Toxicol. 2012;45:610–618.
    1. Herman JF, Mangala LS, Mehta K. Implications of increased tissue transglutaminase (TG2) expression in drug-resistant breast cancer (MCF-7) cells. Oncogene. 2006;25:3049–3058.
    1. Woo CC, Loo SY, Gee V, Yap CW, Sethi G, Kumar AP, Tan KHB. Anticancer activity of thymoquinone in breast cancer cells: Possible involvement of PPAR-g pathway. Biochem Pharmacol. 2011;82:464–475.
    1. Rios-Petrarca C, Brunetto AT, Duval V, Brondani A, Pereira-Carvalho G, Garicochea B. Survivin as a predictive biomarker of complete pathologic response to neoadjuvant chemotherapy in patients with stage II and stage III breast cancer. Clin Breast Cancer. 2011;11:83–88.
    1. Li T, Danelisen I, Singal PK. Early changes in myocardial antioxidant enzymes in rats treated with adriamycin. Moll Cell Biochem. 2002;232:19–26.
    1. Lipshultz SE, Colan SD, Gelber RD, Pérez-Atayde AR, Sallan SE, Sander SP. Late cardiac effect of doxorubicin therapy for acute lymphoblastic leukemia in chilhood. N Engl J Med. 1991;324:808–815.
    1. Swain SM, Whaley FS, Ewer MS. Congestive heart failure in patients treated with doxorubicin. Am Cancer Soc. 2003;97:2869–2879.
    1. Stearns V, Singh B, Tsangaris T, Crawford JG, Novielli A, Ellis MJ, Isaacs C, Pennanen M, Tibery C, Farhad A, Slack R, Hayes DF. A prospective randomized pilot study to evaluate predictors of response in serial core biopsies to single agent neoadjuvant doxorubicin or paclitaxel for patients with locally advanced breast cancer. Clin Cancer Res. 2003;9:124–133.
    1. Takemura G, Fujiwara H. Doxorubicin-induced cardiomyopathy from the cardiotoxic mechanism to management. Prog Cadiovasc Dis. 2007;49:330–352.
    1. Teas J, Pino S, Critchley A, Braverman LE. Variability of iodine content in common commercially available edible seaweeds. Thyroid. 1983;14:836–841.
    1. Cann SA, Van Netten JP, Van Netten C. Hypothesis: Iodine, selenium and the development of breast Cancer. Cancer Causes Control. 2000;11:121–127.
    1. Funahashi H, Imai T, Tanaka Y, Tsukamara K, Kilumori T. Wakame seaweed suppresses the proliferation of 7, 12-dimethylbenz [a]-anthracene-induced mammary tumors in rats. Jpn Cancer Res. 1999;90:922–927.
    1. Venturi S. Evolutionary significance of iodine. Curr Chem Biol. 2011;5:155–162.
    1. Torremante PE, Rosner H. Antiproliferative effects of molecular iodine in cancers. Curr Chem Biol. 2011;5:168–176.
    1. Anguiano B, Aceves C. Iodine in mammary and prostate pathologies. Curr Chem Biol. 2011;5:177–182.
    1. Ghent W, Eskin B, Low D, Hill L. Iodine replacement in fibrocystic disease of the breast. Canadian J Surg. 1993;36:453–460.
    1. Kessler J. In: Comprehensive Handbook of Iodine. Nutritional, endocrine and pathological aspects. Preedy VR, Burrow GN, Watson RR, editor. San Diego, CA: Academic; 2009. Are there side effects when using supraphysiological levels of iodine in treatment regimens; pp. 801–810.
    1. Garcia-Solis P, Alfaro Y, Anguiano B, Delgado G, Guzman RC, Nandi S, Diaz-Muñoz M, Vazquez-Martinez O, Aceves C. Inhibition of N-methyl-N-nitrosourea induced mammary carcinogenesis by molecular iodine (I2) but not by iodide (KI) treatment Evidence that I2 prevents cancer promotion. Mol Cell Endocrinol. 2005;236:49–57.
    1. Arroyo-Helguera O, Delgado G, Anguiano B, Aceves C. Uptake and antiproliferative effect of molecular iodine in the MCF-7 breast cancer cell line. Endocr Related Cancer. 2006;13:1147–1158.
    1. Shrivastava A, Tiwari M, Sinha RA, Kumar A, Balapure AK, Bajpai VK, Sharma R, Mitra K, Tandon A, Godbole MM. Molecular iodine induces caspase-independent apoptosis in human breast carcinoma cells involving mitochondria-mediated pathway. J Biol Chem. 2006;281:19762–19771.
    1. Gartner R, Rank P, Ander B. The role of iodine and delta-iodolactone in growth and apoptosis of malignant thyroid epithelial cells and breast cancer cells. Hormones (Athens) 2010;9:60–66.
    1. Dugrillon A, Uedelhoven W, Pisarev M, Bechtner G, Gartner R. Identification of delta-iodolactone in iodide treated human goiter and its inhibitory effect on proliferation of human thyroid follicles. Horm Metabol Res. 1994;26:465–469.
    1. Juvenal GJ, Thomasz L, Oglio R, Perona M, Pisarev MA, Rossich L, Salvarredi L. Thyroid: iodine beyond the thyronines. Curr Chem Biol. 2011;5:163–167.
    1. Arroyo-Helguera O, Rojas E, Delgado G, Aceves C. Characterization of the cell cycle arrest and signaling pathways involved in the antiproliferative effect of molecular iodine in normal and tumoral breast cells: evidence that 6-iodolactone mediates apoptotic effects. Endocr Related Cancer. 2008;15:1003–1011.
    1. Aceves C, García-Solís P, Arroyo-Helguera O, Vega-Riveroll L, Delgado G, Anguiano B. Antineoplastic effect of iodine in mammary cancer. Participation of 6-iodolactone (6-IL) and peroxisome proliferator-activated receptors (PPAR) Mol Cancer. 2009;8:33–36.
    1. Nuñez-Anita RE, Arroyo-Helguera O, Cajero-Juárez M, López-Bojorquez L, Aceves C. A complex between 6-iodolactone and the peroxisome proliferator-activated receptor type gamma may mediate the antineoplastic effect of iodine in mammary cancer. Prostaglands Other Lipid Mediat. 2009;89:34–42.
    1. Mansure JJ, Nassim R, Kassouf W. Peroxisome proliferator-activated receptor γ in bladder cancer. A promising therapeutic target. Cancer Biol Ther. 2009;8:1–9.
    1. Núñez-Anita RE, Cajero-Juárez M, Aceves C. Peroxisome proliferator-activated receptors. Role of isoform gamma in the antineoplasic effect of iodine in mammary cancer. Curr Cancer Drug Tar. 2011;11:775–786.
    1. Aiello A, Pandini G, Frasca F, Conte E, Murabito A, Sacco A, Genua M, Vigneri R, Belfiore A. Peroxisomal proliferator-activated receptor-gamma agonists induce partial reversion of epithelial-mesenchymal transition in anaplastic thyroid cancer cells. Endocrinology. 2006;47:4463–4475.
    1. Penault-Llorca F, Cayre A, Bouchet-Mishellany F, Amat S, Feillel V, Le Bouedec G, Ferriere JP, DeLatour M, Chollet P. Induction chemotherapy for breast carcinoma: predictive markers and relation with outcome. Int J Oncol. 2003;22:1319–1325.
    1. Doroshow JH, Gershon YL, Myers CE. Enzymatic defenses of the mouse heart against reactive oxygen metabolites. J Clin Invest. 1980;65:128–135.
    1. Hossam MA, Abd-Ellah M, Hafez FH. Abatement by naringenin of doxorubicin-induced cardiac toxicity in rats. J Egyptian Nat Cancer. 2005;17:291–300.
    1. Meischl C, Buermans HP, Hazes T, Zuidwijk MJ, Musters RJ, Boer C, Van Lingen A, Simonides WS, Blankestein MA, Dupuy C, Paulus WJ, Hack CE, Ris-Stalpers C, Roos D, Niessen HW. H9c2 cardiomyoblasts produce thyroid hormone. Am J Physiol Cell Physiol. 2008;294:1227–1233.
    1. Bocca C, Bozzo F, Francica S, Colombatto S, Miglietta A. Involvement of PPAR gamma and E-cadherin/beta-catenin pathway in the antiproliferative effect of conjugated linoleic acid in MCF-7 cells. Int J Cancer. 2007;121:248–256.
    1. Reka AK, Kurapati H, Narala VR, Bommer G, Chen J, Standiford TJ, Keshamouni VG. Peroxisome proliferator-activated receptor-gamma activation inhibits tumor metastasis by antagonizing Smad3-mediated epithelial-mesenchymal transition. Mol Cancer Ther. 2010;12:3221–3232.
    1. Kenkel J. Analytical chemistry for technicians. 2. Boca Raton, Florida: CRC press LLC; 1994. pp. 164–166.
    1. Thompson HJ. In: Methods in Mammary Gland Biology and Breast Cancer Research. 8. Ip M, Asch BB, editor. New York: Kluwer Academic/Plenum Publisher; 2000. Methods for the induction of mammary carcinogenesis in the rat using either 7,12-dimethylbenz(a)antracene or 1-methyl-1-nitrosourea; pp. 19–29.
    1. Fleige S, Pfaffl MW. RNA integrity and the effect on realtime qRT-PCR performance. Mol Aspects Med. 2006;27:126–139.
    1. Aebi H. Catalase in vitro. Methods Enzymol. 1984;105:121–126.
    1. Ottolenghi A. Interaction of ascorbic acid and mitocondrial lipids. Arch Biochem Biophys. 1959;79:355–363.
    1. Benzie IF, Strain JJ. The Ferric Reducing Ability of Plasma (FRAP) as a Measure of “Antioxidant Power”: The FRAP Assay. Analyt Biochem. 1996;239:70–76.
    1. Carabez-T A, Sandoval F, Palma-T L. Ultraestructural changes in tissues produced by inhalation of thinner in rats. Microsc Res Thech. 1998;40:56–62.

Source: PubMed

3
구독하다