Molecular iodine/doxorubicin neoadjuvant treatment impair invasive capacity and attenuate side effect in canine mammary cancer

Xóchitl Zambrano-Estrada, Brianda Landaverde-Quiroz, Andrés A Dueñas-Bocanegra, Marco A De Paz-Campos, Gerardo Hernández-Alberto, Benjamín Solorio-Perusquia, Manuel Trejo-Mandujano, Laura Pérez-Guerrero, Evangelina Delgado-González, Brenda Anguiano, Carmen Aceves, Xóchitl Zambrano-Estrada, Brianda Landaverde-Quiroz, Andrés A Dueñas-Bocanegra, Marco A De Paz-Campos, Gerardo Hernández-Alberto, Benjamín Solorio-Perusquia, Manuel Trejo-Mandujano, Laura Pérez-Guerrero, Evangelina Delgado-González, Brenda Anguiano, Carmen Aceves

Abstract

Background: Mammary cancer has a high incidence in canines and is an excellent model of spontaneous carcinogenesis. Molecular iodine (I2) exerts antineoplastic effects on different cancer cells activating re-differentiation pathways. In co-administration with anthracyclines, I2 impairs chemoresistance installation and prevents the severity of side effects generated by these antineoplastic drugs. This study is a random and double-blind protocol that analyzes the impact of I2 (10 mg/day) in two administration schemes of Doxorubicin (DOX; 30 mg/m2) in 27 canine patients with cancer of the mammary gland. The standard scheme (sDOX) includes four cycles of DOX administered intravenously for 20 min every 21 days, while the modified scheme (mDOX) consists of more frequent chemotherapy (four cycles every 15 days) with slow infusion (60 min). In both schemes, I2 or placebo (colored water) was supplemented daily throughout the treatment.

Results: mDOX attenuated the severity of adverse events (VCOG-CTCAE) in comparison with the sDOX group. The overall tumor response rate (RECIST criteria) for all dogs was 18% (interval of reduction 48-125%), and no significant difference was found between groups. I2 supplementation enhances the antineoplastic effect in mDOX, exhibiting a significant decrease in the tumor epithelial fraction, diminished expression of chemoresistance (MDR1 and Survivin) and invasion (uPA) markers and enhanced expression of the differentiation factor known as peroxisome proliferator-activated receptors type gamma (PPARγ). Significant tumor lymphocytic infiltration was also observed in both I2-supplemented groups. The ten-month survival analysis showed that the entire I2 supplementation (before and after surgery) induced 67-73% of disease-free survival, whereas supplementation in the last period (only after surgery) produced 50% in both schemes.

Conclusions: The mDOX+I2 scheme improves the therapeutic outcome, diminishes the invasive capacity, attenuates the adverse events and increases disease-free survival. These data led us to propose mDOX+I2 as an effective treatment for canine mammary cancer.

Keywords: Animal welfare; Canine mammary cancer; Doxorubicin; Molecular iodine; Neoadjuvant chemotherapy.

Conflict of interest statement

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Functional value according to the VCOG-CTCAE scale. Each point represents the mean and SD for each clinical report during all treatments. Arrows represent the day of chemotherapy (DOX) application
Fig. 2
Fig. 2
Iodine ingestion, thyroid status and cardiac damage during treatments. Values were recorded on the day of the patient’s admission to the protocol (initial) and on the day of mastectomy (final). Total iodine was determined in urine. Triiodothyronine, thyrotropin and creatine kinase type MB (CK-MB) were quantified in serum. Data are expressed as mean ± SD, and the asterisk indicates a significant difference with respect to the initial condition (unpaired Student t test; P < 0.05)
Fig. 3
Fig. 3
Electrocardiogram profile. Values represent the day of the patient’s admission to the protocol (initial) and 1 week before mastectomy (final). One-way ANOVA was performed for each variable and no significant differences were observed
Fig. 4
Fig. 4
Residual tumor size (%), histopathological classification and grade of malignity were analyzed by RECIST scale. Each point represents an individual tumor. One-way ANOVA was performed, and no significant differences were observed
Fig. 5
Fig. 5
Epithelial and connective tissue proportion (%) in the final tumor mass. Micrograph staining with Masson’s trichrome method (red, epithelium; blue, connective tissue). Quantitative analysis was performed as the average of three random regions (40X) using the ImageJ 1.47 program. Data are expressed as mean ± SD, and the asterisk indicates a significant difference between groups. Student´s t-test (P < 0.05)
Fig. 6
Fig. 6
Effect of treatments on the expression of chemoresistant, invasive and differentiation markers. RT-qPCR amplification were performed in residual tumors. Bax/Bcl2 index as apoptotic induction. Multidrug resistance protein 1 (MDR1); urokinase plasminogen activator (uPA), Survivin protein (Surv). Peroxisome proliferator-activated receptors type gamma (PPARγ). Gene expression was calculated using the D cycle threshold method and normalized to β-actin content. Data are expressed as median and the asterisks indicate significant differences between DOX and DOX + I2 groups in each treatment (Mann-Whitney U; P < 0.05)
Fig. 7
Fig. 7
Effect of treatments on lymphocytic infiltration. Micrographs stained with H&E (20X). Quantitative analysis was performed as the average of three random regions using the ImageJ 1.47 program. Linear regression between residual tumor size (%) and lymphocyte number from DOX and DOX + I2 groups (* Pearson coefficient, p > 0.04)

References

    1. Pinho SS, Carvalho S, Cabral J, Reis CA, Gärtner F. Canine tumors: a spontaneous animal model of human carcinogenesis. Transl Res. 2012;159:165–172. doi: 10.1016/j.trsl.2011.11.005.
    1. Karayannopoulou M, Kaldrymidou E, Constantinidis TC, Dessiris A. Adjuvant post-operative chemotherapy in bitches with mammary cancer. J Vet Med A Physiol Pathol Clin Med. 2001;48:85–96. doi: 10.1046/j.1439-0442.2001.00336.x.
    1. Lavalle GE, De Campos CB, Bertagnolli AC, Cassali GD. Canine malignant mammary gland neoplasm with advanced clinical staging treated with carboplatin and cyclooxygenase innibitors. In Vivo. 2012;26:375–379.
    1. Arenas C, Peña L, Granados-Soler JL, Pérez-Alenza MD. Adjuvant therapy for highly malignant canine mammary tumours: Cox-2 inhibitor versus chemotherapy: a case-control prospective study. Vet Rec. 2016; 10.1136/vr.103398.
    1. Karayannopoulou M, Lafioniatis S. Recent advances on canine mammary cancer chemotherapy : a review of studies from 2000 to date. Revue Med Vet. 2016;167:192–200.
    1. Mauri D, Pavlidis N, Ioannidis JP. Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis. J Ntl Cancer Inst. 2005;97:188–194. doi: 10.1093/jnci/dji021.
    1. Wiley JL, Rook KA, Clifford CA, Gregor TP, Sorenmo KU. Efficacy of doxorubicin-based chemotherapy for non-resectable canine subcutaneous haemangiosarcoma. Vet Comp Oncol. 2010;8:221–233.
    1. Mestrinho LA, Bernardo E, Niza MM, Lloret A, Buracco P. Neoadjuvant chemoradiotherapy and surgery as treatment for oral maxillary squamous cell carcinoma in a dog. Aust Vet J. 2012;90:264–268. doi: 10.1111/j.1751-0813.2012.00928.x.
    1. Wouda RM, Hocker SE, Higginbotham ML. Safety evaluation of combination carboplatin and toceranib phosphate (palladia) in tumour-bearing dogs: a phase I dose finding study. Vet Comp Oncol. 2017; 10.1111/vco.12332.
    1. van Dalen EC, van der Pal HJ, Kremer LC. Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy. Cochrane Database Syst Rev. 2016:CD005008. 10.1002/14651858.
    1. Tien CC, Peng YC, Yang FL, Subeq YM, Lee RP. Slow infusion rate of doxorubicin induces higher pro-inflammatory cytokine production. Regul Toxicol Pharmacol. 2016;81:69–76. doi: 10.1016/j.yrtph.2016.08.002.
    1. Polton G. Novel drug approaches in veterinary cancer therapy. Vet Ireland J. 2014;4:27–32.
    1. Leach TN, Childress MO, Greene SN, Mohamed AS, Moore GE, Schrempp DR, Lahrman SR, Knapp DW. Prospective trial of metronomic chlorambucil chemotherapy in dogs with naturally occurring cancer. Vet Comp Oncol. 2012;10:102–112. doi: 10.1111/j.1476-5829.2011.00280.x.
    1. Elmslie RE, Glawe P, Dow SW. Metronomic therapy with cyclophosphamide and piroxicam effectively delays tumor recurrence in dogs with incompletely resected soft tissue sarcomas. J Vet Intern Med. 2008;22:1373–1379. doi: 10.1111/j.1939-1676.2008.0179.x.
    1. Vail DM, von Euler H, Rusk AW, Barber L, Clifford C, Elmslie R, Fulton L, Hirschberger J, Klein M, London C, Martano M, McNiel EA, Morris JS, Northrup N, Phillips B, Polton G, Post G, Rosenberg M, Ruslander D, Sahora A, Siegel S, Thamm D, Westberg S, Winter J, Khanna C. A randomized trial investigating the efficacy and safety of water soluble micellar paclitaxel (Paccal vet) for treatment of nonresectable grade 2 or 3 mast cell tumors in dogs. J Vet Intern Med. 2012;26:598–607. doi: 10.1111/j.1939-1676.2012.00897.x.
    1. London CA, Malpas PB, Wood-Follis SL, Boucher JF, Rusk AW, Rosenberg MP, Henry CJ, Mitchener KL, Klein MK, Hintermeister JG, Bergman PJ, Couto GC, Mauldin GN, Michels GM. Multi-center, placebo-controlled, double-blind, randomized study of oral toceranib phosphate (SU11654), a receptor tyrosine kinase inhibitor, for the treatment of dogs with recurrent (either local or distant) mast cell tumor following surgical excision. Clin Cancer Res. 2009;15:3856–3865. doi: 10.1158/1078-0432.CCR-08-1860.
    1. London C, Mathie T, Stingle N, Clifford C, Haney S, Klein MK, Beaver L, Vickery K, Vail DM, Hershey B, Ettinger S, Vaughan A, Alvarez F, Hillman L, Kiselow M, Thamm D, Higginbotham ML, Gauthier M, Krick E, Phillips B, Ladue T, Jones P, Bryan J, Gill V, Novasad A, Fulton L, Carreras J, McNeill C, Henry C, Gillings S. Preliminary evidence for biologic activity of toceranib phosphate (palladia ®) in solid tumours. Vet Comp Oncol. 2012;10:194–205. doi: 10.1111/j.1476-5829.2011.00275.x.
    1. Fazekas J, Fürdös I, Singer J, Jensen-Jarolim E. Why man’s best friend, the dog, could also benefit from an anti-HER-2 vaccine. Oncol Lett. 2016;12:2271–2276. doi: 10.3892/ol.2016.5001.
    1. Thamm DH, Kurzman ID, Clark MA, Ehrhart EJ, Kraft SL, Gustafson DL, Vail DM. Preclinical investigation of PEGylated tumor necrosis factor α in dogs with spontaneous tumors: phase I evaluation. Clin Cancer Res. 2010;16:1498–1508. doi: 10.1158/1078-0432.CCR-09-2804.
    1. Bolger GT, Licollari A, Tan A, Greil R, Vcelar B, Majeed M, Helson L. Distribution and metabolism of lipocurc™ (liposomal curcumin) in dog and human blood cells: species selectivity and pharmacokinetic relevance. Anticancer Res. 2017;37:3483–3492.
    1. Aceves C, Anguiano B, Delgado G. The extrathyronine actions of iodine as antioxidant, apoptotic, and differentiation factor in various tissues. Thyroid. 2013;23:938–946. doi: 10.1089/thy.2012.0579.
    1. Rösner H, Möller W, Groebner S, Torremante P. Antiproliferative/cytotoxic effects of molecular iodine, povidone-iodine and Lugol's solution in different human carcinoma cell lines. Oncol Lett. 2016;12:2159–2162. doi: 10.3892/ol.2016.4811.
    1. Bontempo A, Ugalde-Villanueva B, Delgado-González E, Rodríguez AL, Aceves C. Molecular iodine impairs chemoresistance mechanisms, enhances doxorubicin retention and induces downregulation of the CD44+/CD24+ and E-cadherin+/vimentin+ subpopulations in MCF-7 cells resistant to low doses of doxorubicin. Oncol Rep. 2017; 10.3892/or.2017.5934.
    1. Alfaro Y, Delgado G, Cárabez A, Anguiano B, Aceves C. Iodine and doxorubicin, a good combination for mammary cancer treatment: antineoplastic adjuvancy, chemoresistance inhibition, and cardioprotection. Mol Cancer. 2013;12:45–56. doi: 10.1186/1476-4598-12-45.
    1. Aceves C, Peralta G, Torres J, Delgado G, Domínguez A, De Obaldía R, Duarte LF, Paredes E, Avecilla C. AACR special conference: Advances in Breast Cancer Research. 2011. Iodine-supplemented diets prevents the development of resistance in breast cancer chemotherapy: participation of proliferative peroxisome-activated receptor gamma (PPARg)
    1. Nava-Villalba M, Nuñez-Anita RE, Bontempo A, Aceves C. Activation of peroxisome proliferator-activated receptor gamma is crucial for antitumoral effects of 6-iodolactone. Mol Cancer. 2015;14:168–172. doi: 10.1186/s12943-015-0436-8.
    1. Yousefi B, Zarghami N, Samadi N, Majidinia M. Peroxisome proliferator-activated receptors and their ligands in cancer drug- resistance: opportunity or challenge. Anti Cancer Agents Med Chem. 2016;16:1541–1548. doi: 10.2174/1871520616666160204112941.
    1. Goldschmidt M, Peña L, Rasotto R, Zappulli V. Classification and grading of canine mammary tumors. Vet Pathol. 2011;48:117–131. doi: 10.1177/0300985810393258.
    1. Veterinary cooperative oncology group - common terminology criteria for adverse events (VCOG-CTCAE) following chemotherapy or biological antineoplastic therapy in dogs and cats v1.1. Vet Comp Oncol. 2016;14:417–46.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Arroyo-Helguera O, Rojas E, Delgado G, Aceves C. Signaling pathways involved in the antiproliferative effect of molecular iodine in normal and tumoral breast cells: evidence that 6-iodolactone mediates apoptotic effects. Endocr Relat Cancer. 2008;15:1003–1011. doi: 10.1677/ERC-08-0125.
    1. Upadhyay G, Singh R, Sharma R, Balapure AK, Godbole MM. Differential action of iodine on mitochondria from human tumoral- and extra-tumoral tissue in inducing the release of apoptogenic proteins. Mitochondrion. 2002;2:199–210. doi: 10.1016/S1567-7249(02)00068-5.
    1. Rillema JA, Mulder JA. Arachidonic acid distribution in lipids of mammary glands and DMBA-induced tumors of rats. Prostaglandins Med. 1978;1:31–38. doi: 10.1016/0161-4630(78)90074-5.
    1. Razanamahefa L, Prouff S, Bardon S. Stimulatory effect of arachidonic acid on T-47D human breast cancer cell growth is associated with enhancement of cyclin D1 mRNA expression. Nutr Cancer. 2000;38:274–280. doi: 10.1207/S15327914NC382_17.
    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. doi: 10.1186/1476-4598-8-33.
    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. Prostaglandins Other Lipid Mediat. 2009;89:34–42. doi: 10.1016/j.prostaglandins.2009.04.001.
    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;9:3221–3232. doi: 10.1158/1535-7163.MCT-10-0570.
    1. Wahl GM, Spike BT. Cell state plasticity, stem cells, EMT, and the generation of intra-tumoral heterogeneity. NPJ Breast Cancer. 2017; 10.1038/s41523-017-0012-z.
    1. Asano Y, Kashiwagi S, Goto W, Kurata K, Noda S, Takashima T, Onoda N, Tanaka S, Ohsawa M, Hirakawa K. Tumour-infiltrating CD8 to FOXP3 lymphocyte ratio in predicting treatment responses to neoadjuvant chemotherapy of aggressive breast cancer. Br J Surg. 2016;103:845–854. doi: 10.1002/bjs.10127.
    1. Denkert C, Loibl S, Noske A, Roller M, Müller BM, Komor M, Budczies J, Darb-Esfahani S, Kronenwett R, Hanusch C, von Törne C, Weichert W, Engels K, Solbach C, Schrader I, Dietel M, von Minckwitz G. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clinl Oncol. 2010;28:105–113. doi: 10.1200/JCO.2009.23.7370.
    1. Beukelman CJ, van den Berg AJ, Hoekstra MJ, Uhl R, Reimer K, Mueller S. Anti-inflammatory properties of a liposomal hydrogel with povidone-iodine (Repithel) for wound healing in vitro. Burns. 2008;34:845–855. doi: 10.1016/j.burns.2007.11.014.
    1. Soriguer F, Gutiérrez-Repiso C, Rubio-Martin E, Linares F, Cardona I, López-Ojeda J, Pacheco M, González-Romero S, Garriga MJ, Velasco I, Santiago P, García-Fuentes E. Iodine intakes of 100–300 μg/d do not modify thyroid function and have modest anti-inflammatory effects. Br J Nutr. 2011;105:1783–1790. doi: 10.1017/S0007114510005568.
    1. Miyachi Y, Niwa Y. Effects of potassium iodide, colchicine and dapsone on the generation of polymorphonuclear leukocyte-derived oxygen intermediates. Br J Dermatol. 1982:209–14.
    1. Honma K, Saga K, Onodera H, Takahashi M. Potassium iodide inhibits neutrophil chemotaxis. Acta Derm Venereol. 1990;70:247–249.
    1. Costa RO, Macedo PM, Carvalhal A, Bernardes-Engemann AR. Use of potassium iodide in dermatology: updates on an old drug. An Bras Dermatol. 2013;88:396–402. doi: 10.1590/abd1806-4841.20132377.
    1. Moore K, Thomas A, Harding KG. Iodine released from the wound dressing iodosorb modulates the secretion of cytokines by human macrophages responding to bacterial lipopolysaccharide. Int J Biochem Cell Biol. 1997;29:163–171. doi: 10.1016/S1357-2725(96)00128-8.
    1. McCarty MF, Barroso-Aranda J, Contreras F. PPAR gamma agonists can be expected to potentiate the efficacy of metronomic chemotherapy through CD36 up-regulation. Med Hypotheses. 2008;70:419–423. doi: 10.1016/j.mehy.2006.12.064.
    1. Aceves C, Anguiano B. Is iodine an antioxidant and Antiproliferative agent for the mammary and prostate glands? In: Preedy VR, Burrow GN, Watson RR, editors. Comprehensive Handbook of Iodine. Oxford: Academic Press, Elsevier; 2009. pp. 249–257.
    1. Zava TT, Zava DT. Assessment of Japanese iodine intake based on seaweed consumption in Japan: a literature-based analysis. Thyroid Res. 2011; 10.1186/1756-6614-4-14.

Source: PubMed

3
구독하다