Ocoxin oral solution® as a complement to irinotecan chemotherapy in the metastatic progression of colorectal cancer to the liver

Iera Hernandez-Unzueta, Aitor Benedicto, Elvira Olaso, Eduardo Sanz, Cristina Viera, Beatriz Arteta, Joana Márquez, Iera Hernandez-Unzueta, Aitor Benedicto, Elvira Olaso, Eduardo Sanz, Cristina Viera, Beatriz Arteta, Joana Márquez

Abstract

Colorectal cancer (CRC) is an aggressive disease in which patients usually die due to its metastatic progression to the liver. Up to date, irinotecan is one of the most used chemotherapeutic agents to treat CRC metastasis with demonstrated efficacy. However, the severity of the side effects constitute the main limitation to its use in the treatment. Consequently, new complementary therapies are being developed to avoid these adverse effects while maintaining the efficacy of the antitumoral drugs. Ocoxin oral solution (OOS®) is a nutritional mixture containing biologically active compounds with demonstrated antitumoral and immunomodulatory effects. Thus, we aimed to analyze the effect of OOS® as a suitable complement to irinotecan therapy in the treatment of CRC metastasis to the liver. First, the effect of OOS®, irinotecan and the combination of both on the viability of C26 cells was tested in vitro and in vivo. Second, the expression of caspase-3, Ki67 and the macrophage infiltration by F4/80 marker was quantified in liver tissue sections by immunohistochemistry. Finally, mRNA microarray study was carried out on tumor cells isolated from tumor-bearing livers collected from mice subjected to the above treatments. Our results show that OOS® administered as a complementary therapy to irinotecan reduced tumor cell viability in vitro. Moreover, irinotecan administered either alone or in combination with 100 µl OOS® from the 7th day after tumor cell inoculation decreased the metastatic growth in the liver. Besides, several genes with binding and catalytic activities showed to be deregulated by RNA microarray analysis. In conclusion, OOS®, when administered as a complement to irinotecan, reduced the metastatic development of colorectal cancer to the liver. Additionally, the overall health state of the animals improved. These results point out OOS® as a potential supplement to the anti-tumoral treatments used in clinical settings in patients suffering from disseminated colorectal cancer.

Keywords: colorectal cancer; liver; metastasis; nutrient mixture; ocoxin oral solution.

Figures

Figure 1.
Figure 1.
Experimental animal groups treatment pattern. Seven days after tumor cell inoculation, at least 6 mice per group were divided into 5 experimental groups as follows: group I, untreated mice; group II, mice treated with a daily oral administration of 100 µl of OOS®; group III, mice treated with a i.p. dose of 20 µg/ml of irinotecan every two days; group IV, mice treated with both, a daily oral dose of 100 µl of OOS® and 20 µg/ml of irinotecan every two days; group V, untreated mice used to control the metastatic development. All mice were treated for a period of 14 days.
Figure 2.
Figure 2.
Effect of the combined treatment of irinotecan and OOS® in the viability of C26 cells. The viability of C26 cells was tested in the presence of OOS® (1:100), irinotecan (50 µM) or the combination of OOS® and irinotecan (1:100 and 50 µM) for 48 h. Then, the viability was quantified in untreated (black) and treated (grey) cells. Data are mean values ± SD from three different experiments. Differences in the viability of treated cells vs. untreated cells (**) and vs. initially cultured cells (*) were considered to be statistically significant at P<0.05.
Figure 3.
Figure 3.
Effect of the combined treatment of irinotecan and OOS® in the development of CRC metastasis to the liver. C26 cells were i.s. inoculated into mice and seven days later they were divided into the five groups shown in Fig. 1 with at least 6 mice per group. Mice were either untreated or treated with OOS® (100 µl), irinotecan (20 mg/kg) or with the combined therapy (100 µl OOS® and 20 mg/kg irinotecan) as described in Material and Methods. (A) Images showing tumor foci grown in the liver of untreated and treated mice. Image original magnification was ×4. (B) The total tumoral burden was quantified and represented as the percentage of liver area occupied by the tumor. Differences were considered statistically significant at *P<0.05.
Figure 4.
Figure 4.
Effect of the combined treatment of irinotecan and OOS® in the proliferation and apoptotic markers. The expression of Ki67 and caspase-3 in livers collected from tumor-bearing mice either untreated or treated with OOS® (100 µl), irinotecan (20 mg/kg) or the complementary therapy (100 µl OOS® and 20 mg/kg irinotecan) was analyzed by immunohistochemistry with at least 6 mice per group. (A) Ki67 expression was quantified in livers collected from untreated and treated C26-bearing mice as the percentage of the area positive for Ki67 expression within tumor foci respect to the total metastatic tumor area. (B) Caspase-3 expression was quantified in livers collected from untreated and treated C26-bearing mice as the percentage of the area positive for caspase-3 expression respect to the total metastatic tumor area. (C) The ratio between Ki67 and caspase-3 in livers was calculated from results shown in A and B. Differences were considered statistically significant at *P<0.05.
Figure 5.
Figure 5.
Effect of the combined treatment of irinotecan and OOS® on the tumor infiltration of macrophages in vivo. Expression level of F4/80 was analyzed in liver tissue by immunohistochemistry. (A) Images showing F4/80 expression (brown) and hematoxylin (purple) in liver tissue collected from untreated and treated mice. Image magnification was ×20. (B) F4/80 expression was quantified in livers collected from untreated and treated C26-bearing mice. Data are calculated as % of F4/80 expression per tumor foci area. At least 6 mice per group were used and differences were considered statistically significant at *P<0.05.
Figure 6.
Figure 6.
Metastatic tumor explant based mRNA microarray study. A microarray assay was carried out to detect differences in gene expression levels between tumor cells collected from mice treated under different protocols. Four tumor explants per experimental group were collected, each of them from one tumor bearing mouse. (A) A molecular functional gene analysis was carried out with the PANTHER (v.10.0) (9,10) analysis system for the untreated Control vs. irinotecan treatments. (B) A molecular functional gene analysis was carried out with the PANTHER (v.10.0) (9,10) analysis system for the OOS®+irinotecan vs. irinotecan treatments.

References

    1. Fujita K, Kubota Y, Ishida H, Sasaki Y. Irinotecan, a key chemotherapeutic drug for metastatic colorectal cancer. World J Gastroenterol. 2015;21:12234–12248. doi: 10.3748/wjg.v21.i43.12234.
    1. Van den Eynden GG, Majeed AW, Illemann M, Vermeulen PB, Bird NC, Høyer-Hansen G, Eefsen RL, Reynolds AR, Brodt P. The multifaceted role of the microenvironment in liver metastasis: Biology and clinical implications. Cancer Res. 2013;73:2031–2043. doi: 10.1158/0008-5472.CAN-12-3931.
    1. Gol'dberg ED, Amosova EN, Zueva EP, Razina TG, Krylova SG, Zorikov PS. Licorice preparations improve efficiency of chemotherapy and surgical treatment of transplanted tumors. Bull Exp Biol Med. 2008;145:252–255. doi: 10.1007/s10517-008-0063-0.
    1. Kontek R, Drozda R, Sliwiński M, Grzegorczyk K. Genotoxicity of irinotecan and its modulation by vitamins AC and E in human lymphocytes from healthy individuals and cancer patients. Toxicol In Vitro. 2010;24:417–424. doi: 10.1016/j.tiv.2009.10.013.
    1. Arabski M, Kazmierczak P, Wisniewska-Jarosinska M, Poplawski T, Klupinska G, Chojnacki J, Drzewoski J, Blasiak J. Interaction of amoxicillin with DNA in human lymphocytes and H. pylori-infected and non-infected gastric mucosa cells. Chem Biol Interact. 2005;152:13–24. doi: 10.1016/j.cbi.2005.01.004.
    1. Hernández-Garcia S, González V, Sanz E, Pandiella A. Effect of oncoxin oral solution in HER2-overexpressing breast cancer. Nutr Cancer. 2015;67:1159–1169. doi: 10.1080/01635581.2015.1068819.
    1. Márquez J, Mena J, Hernandez-Unzueta I, Benedicto A, Sanz E, Arteta B, Olaso E. Ocoxin® oral solution slows down tumor growth in an experimental model of colorectal cancer metastasis to the liver in balb/c mice. Oncol Rep. 2016;35:1265–1272.
    1. Diaz-Rodriguez E, Hernández-Garcia S, Sanz E, Pandiella A. Antitumoral effect of ocoxin on acute myeloid leukemia. Oncotarget. 2016;7:6231–6242.
    1. Mi H, Muruganujan A, Casagrande JT, Thomas PD. Large-scale gene function analysis with the PANTHER classification system. Nat Protoc. 2013;8:1551–1566. doi: 10.1038/nprot.2013.092.
    1. Mi H, Poudel S, Muruganujan A, Casagrande JT, Thomas PD. PANTHER version 10: Expanded protein families and functions, and analysis tools. Nucleic Acids Res. 2016;44(D1):D336–D342. doi: 10.1093/nar/gkv1194.
    1. Langford DJ, Bailey AL, Chanda ML, Clarke SE, Drummond TE, Echols S, Glick S, Ingrao J, Klassen-Ross T, Lacroix-Fralish ML, et al. Coding of facial expressions of pain in the laboratory mouse. Nat Methods. 2010;7:447–449. doi: 10.1038/nmeth.1455.
    1. Mego M, Chovanec J, Vochyanova-Andrezalova I, Konkolovsky P, Mikulova M, Reckova M, Miskovska V, Bystricky B, Beniak J, Medvecova L, et al. Prevention of irinotecan induced diarrhea by probiotics: A randomized double blind, placebo controlled pilot study. Complement Ther Med. 2015;23:356–362. doi: 10.1016/j.ctim.2015.03.008.
    1. Mikalauskas S, Mikalauskiene L, Bruns H, Nickkholgh A, Hoffmann K, Longerich T, Strupas K, Büchler MW, Schemmer P. Dietary glycine protects from chemotherapy-induced hepatotoxicity. Amino Acids. 2011;40:1139–1150. doi: 10.1007/s00726-010-0737-6.
    1. Dayem-Uddin M, Islam M, Mahmood I, Gosh A, Khatun R, Kundu S. Findings of the 3-month supportive treatment with oncoxin solution beside the standard modalities of patients with different neoplastic diseases. TAJ. 2009;22:172–175.
    1. Kim JA, Kim DH, Hossain MA, Kim MY, Sung B, Yoon JH, Suh H, Jeong TC, Chung HY, Kim ND. HS-1793, a resveratrol analogue, induces cell cycle arrest and apoptotic cell death in human breast cancer cells. Int J Oncol. 2014;44:473–480.
    1. Gullett NP, Amin AR Ruhul, Bayraktar S, Pezzuto JM, Shin DM, Khuri FR, Aggarwal BB, Surh YJ, Kucuk O. Cancer prevention with natural compounds. Semin Oncol. 2010;37:258–281. doi: 10.1053/j.seminoncol.2010.06.014.
    1. Mehta RG, Murillo G, Naithani R, Peng X. Cancer chemoprevention by natural products: How far have we come? Pharm Res. 2010;27:950–961. doi: 10.1007/s11095-010-0085-y.
    1. Cho YH, Yoon SY, Kim SN. Irinotecan monotherapy versus irinotecan-based combination as second-line chemotherapy in advanced gastric cancer: A meta-analysis. Cancer Res Treat. 2017;49:255–262. doi: 10.4143/crt.2015.452.
    1. Kachalaki S, Ebrahimi M, Khosroshahi L Mohamed, Mohammadinejad S, Baradaran B. Cancer chemoresistance; biochemical and molecular aspects: A brief overview. Eur J Pharm Sci. 2016;89:20–30. doi: 10.1016/j.ejps.2016.03.025.
    1. Stringer AM, Gibson RJ, Logan RM, Bowen JM, Yeoh AS, Laurence J, Keefe DM. Irinotecan-induced mucositis is associated with changes in intestinal mucins. Cancer Chemother Pharmacol. 2009;64:123–132. doi: 10.1007/s00280-008-0855-y.
    1. Stringer AM, Gibson RJ, Bowen JM, Logan RM, Ashton K, Yeoh AS, Al-Dasooqi N, Keefe DM. Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile. Int J Exp Pathol. 2009;90:489–499. doi: 10.1111/j.1365-2613.2009.00671.x.
    1. Kung HN, Weng TY, Liu YL, Lu KS, Chau YP. Sulindac compounds facilitate the cytotoxicity of β-lapachone by up-regulation of NAD(P)H quinone oxidoreductase in human lung cancer cells. PLoS One. 2014;9:e88122. doi: 10.1371/journal.pone.0088122.
    1. Wei D, Zhang X, Zou H, Wang L, Fu B, Wu X, Luo Z, Li X, Ge J, Li Y, et al. WW domain containing oxidoreductase induces apoptosis in gallbladder-derived malignant cell by upregulating expression of P73 and PUMA. Tumour Biol. 2014;35:1539–1550. doi: 10.1007/s13277-013-1213-1.
    1. Pinho SS, Reis CA. Glycosylation in cancer: Mechanisms and clinical implications. Nat Rev Cancer. 2015;15:540–555. doi: 10.1038/nrc3982.
    1. Sheta R, Woo CM, Roux-Dalvai F, Fournier F, Bourassa S, Droit A, Bertozzi CR, Bachvarov D. A metabolic labeling approach for glycoproteomic analysis reveals altered glycoprotein expression upon GALNT3 knockdown in ovarian cancer cells. J Proteomics. 2016;145:91–102. doi: 10.1016/j.jprot.2016.04.009.
    1. Feferman L, Bhattacharyya S, Deaton R, Gann P, Guzman G, Kajdacsy-Balla A, Tobacman JK. Arylsulfatase B (N-acetylgalactosamine-4-sulfatase): Potential role as a biomarker in prostate cancer. Prostate Cancer Prostatic Dis. 2013;16:277–284. doi: 10.1038/pcan.2013.18.
    1. Brinkmann K, Zigrino P, Witt A, Schell M, Ackermann L, Broxtermann P, Schüll S, Andree M, Coutelle O, Yazdanpanah B, et al. Ubiquitin C-terminal hydrolase-L1 potentiates cancer chemosensitivity by stabilizing NOXA. Cell Rep. 2013;3:881–891. doi: 10.1016/j.celrep.2013.02.014.

Source: PubMed

3
Iratkozz fel