Elevated alpha1-acid glycoprotein in gastric cancer patients inhibits the anticancer effects of paclitaxel, effects restored by co-administration of erythromycin

Yoshinao Ohbatake, Sachio Fushida, Tomoya Tsukada, Jun Kinoshita, Katsunobu Oyama, Hironori Hayashi, Tomoharu Miyashita, Hidehiro Tajima, Hiroyuki Takamura, Itasu Ninomiya, Masakazu Yashiro, Kousei Hirakawa, Tetsuo Ohta, Yoshinao Ohbatake, Sachio Fushida, Tomoya Tsukada, Jun Kinoshita, Katsunobu Oyama, Hironori Hayashi, Tomoharu Miyashita, Hidehiro Tajima, Hiroyuki Takamura, Itasu Ninomiya, Masakazu Yashiro, Kousei Hirakawa, Tetsuo Ohta

Abstract

Paclitaxel (PTX) which easily elutes into ascites is widely used to treat gastric cancer patients with peritoneal carcinomatosis (PC), but clinical outcomes are suboptimal. Increased concentrations of α1-acid glycoprotein (AGP), an important drug-binding protein, have been reported in the plasma and ascites of cancer patients. This study sought to clarify whether AGP binds to PTX and alters its anticancer effects. AGP concentrations were measured in the serum and ascites of gastric cancer patients with PC and in the serum of healthy volunteers. The in vitro effects of AGP and AGP plus erythromycin (EM) on PTX were evaluated by MTT assays in the gastric cancer cell lines. We also measured AGP concentrations in the ascites of PC model mice and examined the effects of EM plus PTX on PC. The mean AGP concentrations in the serum and ascites of gastric cancer patients with PC were 1524 and 834 μg/mL, respectively, higher than the mean AGP concentration of 650 μg/mL observed in the sera of healthy volunteers. AGP > 400 μg/mL significantly suppressed the cell growth inhibitory effect of PTX in vitro, but the co-administration of EM restored it. Elevated AGP concentrations were observed in the ascites of PC model mice. Administration of PTX alone did not markedly diminish PC, whereas co-administration of PTX and EM significantly reduced PC (p = 0.011). AGP is an important regulatory factor modulating the anticancer activity of intraperitoneal PTX. The co-administration of PTX and EM may be effective in treating gastric cancer patients with PC.

Keywords: Erythromycin; Gastric cancer; Paclitaxel; Peritoneal carcinomatosis; α1-Acid glycoprotein.

Conflict of interest statement

The authors declare that they have no conflict of interest. Ethical approval All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. Human and animal rights All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional review board at Kanazawa University and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. All procedures performed in studies involving animals adhere to the Standard Guidelines for Animal Experiments at Kanazawa University. Informed consent Informed consent was obtained from all individual participants included in the study.

Figures

Fig. 1
Fig. 1
Concentrations of AGP in the serum of gastric cancer patients with PC were about 2.5-fold higher than those of healthy volunteers. The mean concentration of AGP was higher in ascites of gastric cancer patients with PC than in the serum of healthy volunteer, but this difference was not statistically significant. Data are presented as mean ± SD. *p < 0.05 versus serum of healthy volunteers
Fig. 2
Fig. 2
AGP concentrations in ascites of our xenograft model mouse increased with cancer progression, starting on day 10 and reaching a maximum of 400 μg/mL on day 21. Data are presented as mean ± SD. *p < 0.05 versus day 3
Fig. 3
Fig. 3
Effect of PTX was assessed by MTT assays. a OCUM-2MD3 cell viability, b NUGC-3 cell viabilities. Results reported as mean ± SD of three independent experiments
Fig. 4
Fig. 4
Effect of AGP (0–1200 μg/mL) on PTX-suppressed conditions was assessed by MTT assays. a OCUM-2MD3 cell viability, b NUGC-3 cell viability. Results reported as mean ± SD of three independent experiments. *p < 0.05 versus cells added culture medium alone
Fig. 5
Fig. 5
Effect of the co-administration of PTX and EM on OCUM-2MD3 and NUGC-3 cell viabilities in a high-AGP environment was determined using MTT assays. OCUM-2MD3 and NUGC-3 cells were incubated with EM (0.1, 1, 10, 100 μM), AGP (800 μg/mL), and PTX (10 nM), and cell viabilities were measured. Data are presented as mean ± SD of three independent experiments. *p < 0.05 versus cells treated with PTX and AGP
Fig. 6
Fig. 6
Effect of co-administration of PTX and EM without AGP on NUGC-3 cell viability was determined using MTT assays. NUGC-3 cells were incubated with EM (0.1, 1, 10, 100 μM), and PTX (10 nM), and cell viabilities were measured. The difference of cell viability between the group treated with PTX alone and the groups treated with PTX plus EM was not statistically significant. Data are presented as mean ± SD of three independent experiments
Fig. 7
Fig. 7
Xenograft model was made by incubating OCUM-2MD3 into nu/nu mice intraperitoneally. Effect of PTX plus EM on metastatic nodules was assessed. a Macroscopic views of peritoneal nodules (arrow head). b Relative number and weight of metastatic peritoneal nodules in the xenograft model. Data are shown as mean ± SD. *p < 0.05 versus untreated group

References

    1. Jemal A, Bray F, Center MM, Ferlay J, Ward EFD. Global cancer statistics. A Cancer J Clin. 2011;61:69–90. doi: 10.3322/caac.20107.
    1. Bertuccio P, Chatenoud L, Levi F, et al. Recent patterns in gastric cancer : a global overview. Int J Cancer. 2009;125:666–673. doi: 10.1002/ijc.24290.
    1. Okabe H, Ueda S, Obama K, Hosogi H, Sakai Y. Induction chemotherapy with S-1 plus cisplatin followed by surgery for treatment of gastric cancer with peritoneal dissemination. Ann Surg Oncol. 2009;16:3227–3236. doi: 10.1245/s10434-009-0706-z.
    1. Koizumi W, Narahara H, Hara T, et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase III trial. Lancet Oncol. 2008;9:215–221. doi: 10.1016/S1470-2045(08)70035-4.
    1. Yamao T, Shimada Y, Shirao K, et al. Phase II study of sequential methotrexate and 5-fluorouracil chemotherapy against peritoneally disseminated gastric cancer with malignant ascites: a report from the Gastrointestinal Oncology Study Group of the Japan Clinical Oncology Group, JCOG 9603 Trial. Jpn J Clin Oncol. 2004;34:316–322. doi: 10.1093/jjco/hyh063.
    1. Kobayashi M, Sakamoto J, Namikawa T, et al. Pharmacokinetic study of paclitaxel in malignant ascites from advanced gastric cancer patients. World J Gastroenterol. 2006;12:1412–1415. doi: 10.3748/wjg.v12.i9.1412.
    1. Tamura S, Miki H, Nakata K, et al. Intraperitoneal administration of paclitaxel and oral S-1 for a patient with peritoneal dissemination and hydronephrosis due to advanced gastric cancer. Gastric Cancer. 2007;10:251–255. doi: 10.1007/s10120-007-0431-x.
    1. Ishigami H, Kitayama J, Kaisaki S, et al. Phase II study of weekly intravenous and intraperitoneal paclitaxel combined with S-1 for advanced gastric cancer with peritoneal metastasis. Ann Oncol. 2010;21:67–70. doi: 10.1093/annonc/mdp260.
    1. Takiuchi H, Fukuda H, Boku N, et al. Randomized phase II study of best-available 5-fluorouracil (5-FU) versus weekly paclitaxel in gastric cancer (GC) with peritoneal metastasis (PM) refractory to 5-FU-containing regimens (JCOG0407). J Clin Oncol 2010;28:15(suppl; abstr 4052).
    1. Tsukada T, Fushida S, Harada S, et al. Low-dose paclitaxel modulates tumour fibrosis in gastric cancer. Int J Oncol. 2013;42:1167–1174.
    1. Lima JJ, Boudoulas H, Blanford M. Concentration-dependence protein and its influence of disopyramide binding to on kinetics and dynamics 1. J Pharmacol Exp Ther. 1981;219:741–747.
    1. Stewart CF, Arbuck SG, Fleming R, Evans WE. Relation of systemic exposure to unbound etoposide and hematologic toxicity. Clin Pharmacol Ther. 1991;50:385–393. doi: 10.1038/clpt.1991.155.
    1. Finlay GJ, Baguley BC. Effects of protein binding on the in vitro activity of antitumour acridine derivatives and related anticancer drugs. Cancer Chemother Pharmacol. 2000;45(5):417–422. doi: 10.1007/s002800051011.
    1. Routledge PA. The plasma protein binding of basic drugs. Br J Clin Pharmacol. 1986;22:499–506. doi: 10.1111/j.1365-2125.1986.tb02927.x.
    1. Piver MS, Moyer M, Diakun K, Lele SB, Chu TM. Serum alpha1-acid glycoprotein in epithelial ovarian cancer. Gynecol Oncol. 1988;29:305–308. doi: 10.1016/0090-8258(88)90229-6.
    1. Elg SA, Mayer AR, Carson LF, Twiggs LB, Hill RB, Ramakrishnan S. Alpha-1 acid glycoprotein is an immunosuppressive factor found in ascites from ovaria carcinoma. Cancer. 1997;80:1448–1456. doi: 10.1002/(SICI)1097-0142(19971015)80:8<1448::AID-CNCR12>;2-5.
    1. Azuma M, Nishioka Y, Aono Y, et al. Role of alpha1-acid glycoprotein in therapeutic antifibrotic effects of imatinib with macrolides in mice. Am J Respir Crit Care Med. 2007;176:1243–1250. doi: 10.1164/rccm.200702-178OC.
    1. Filip Z, Jan K, Vendula S, Jana KZ, Kamil M, Kamil K. Albumin and α1-acid glycoprotein: old acquaintances. Expert Opin Drug Metab Toxicol. 2013;9:943–954. doi: 10.1517/17425255.2013.790364.
    1. Otagiri M, Miyoshi T, Yamamichi R, Maruyama T, Perrin JH. Effects of tricyclic drug on induced circular dichroism spectra of dicumarol bound to α 1-acid glycoprotein. Biochem Pharmacol. 1991;42:729–733. doi: 10.1016/0006-2952(91)90029-5.
    1. Miyoshi T, Yamamichi R, Maruyama T, Otagiri M. Reversal of signs of induced cotton effects of dicumarol-alpha 1-acid glycoprotein systems by phenothiazine neuroleptics through ternary complexation. Pharm Res. 1992;9:845–849. doi: 10.1023/A:1015880327911.
    1. Miyoshi T, Yamamichi R, Maruyama T, Takadate A, Otagiri M. Further characterization of reversal of signs of induced cotton effects of dicumarol derivatives-alpha 1-acid glycoprotein systems by protriptyline. Biochem Pharmacol. 1992;43:2161–2167. doi: 10.1016/0006-2952(92)90175-I.
    1. Otagiri M. A molecular functional study on the interactions of drugs with plasma proteins. Drug Metab Pharmacokinet. 2005;20:309–323. doi: 10.2133/dmpk.20.309.
    1. Tamura K, Shibata Y, Matsuda Y, Ishida N. Isolation and characterization of an immunosuppressive acidic protein from ascitic fluids of cancer patients isolation and characterization of an immunosuppressive. Cancer Res. 1981;41:3244–3252.
    1. Shibata Y, Tamura K, Ishida N. In vivo analysis of the suppressive effects of immunosuppressive acidic protein, a type of alpha 1-acid glycoprotein, in connection with its high level in tumor-bearing mice. Cancer Res. 1983;43:2889–2896.
    1. Shimizu N, Yamane T, Karino T, et al. Immunosuppressive acidic protein (IAP) in gastric cancer patients. Jpn J Surg. 1983;13:312–316. doi: 10.1007/BF02469512.
    1. Harris JW, Rahman A, Kim BR, Guengerich FP, Collins JM. Metabolism of taxol by human hepatic microsomes and liver slices: participation of cytochrome P450 3A4 and an unknown P450 enzyme. Cancer Res. 1994;54(15):4026–4035.
    1. Rahman A, Korzekwa KR, Grogan J, Gonzalez FJ, Harris JW. Selective biotransformation of taxol to 6 alpha-hydroxytaxol by human cytochrome P450 2C8. Cancer Res. 1994;54(21):5543–5546.
    1. Rahman A, Korzekwa KR, Grogan J, Gonzalez FJ, Harris JW. Selective biotransformation of taxol to 6 alpha-hydroxytaxol by human cytochrome P450 2C8. Cancer Res. 1994;54(21):5543–5546.
    1. Sparreboom A A, van Asperen J, Mayer U, et al. Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci USA. 1997;94(5):2031–2035. doi: 10.1073/pnas.94.5.2031.
    1. Huisman MT, Chhatta AA, van Tellingen O, Beijnen JH, Schinkel AH. MRP2 (ABCC2) transports taxanes and confers paclitaxel resistance and both processes are stimulated by probenecid. Int J Cancer. 2005;116(5):824–829. doi: 10.1002/ijc.21013.
    1. Franke RM, Lancaster CS, Peer CJ, et al. Effect of ABCC2 (MRP2) transport function on erythromycin metabolism. Clin Pharmacol Ther. 2011;89(5):693–701. doi: 10.1038/clpt.2011.25.
    1. Bruno R, Olivares R, Berille J, et al. Alpha-1-acid glycoprotein as an independent predictor for treatment effects and a prognostic factor of survival in patients with non-small cell lung cancer treated with docetaxel. Clin Cancer Res. 2003;9:1077–1082.
    1. Finlay GJ, Baguley BC. Effects of protein binding on the in vitro activity of antitumour acridine derivatives and related anticancer drugs. Cancer Chemother Pharmacol. 2000;45:417–422. doi: 10.1007/s002800051011.
    1. Conte JE, Golden JA, Duncan S, McKenna E, Zurlinden E. Intrapulmonary pharmacokinetics of clarithromycin and of erythromycin. Antimicrob Agents Chemother. 1995;39:334–338. doi: 10.1128/AAC.39.2.334.
    1. Männistö PT, Hanhijärvi H, Havas A, Vuorela A, Komulainen H, Rauramaa V. Efficacy of erythromycin acistrate (2′-acetyl erythromycin stearate) and erythromycin stearate in experimental infections in mice. J Pharmacol Exp Ther. 1989;250:1028–1033.
    1. Pai SR, Singh KV, Murray BE. In vivo efficacy of the ketolide ABT-773 (cethromycin) against enterococci in a mouse peritonitis model. Antimicrob Agents Chemother. 2003;47:2706–2709. doi: 10.1128/AAC.47.8.2706-2709.2003.
    1. Fruscio R, Lissoni AA, Frapolli R, et al. Clindamycin–paclitaxel pharmacokinetic interaction in ovarian cancer patients. Cancer Chemother Pharmacol. 2006;58(3):319–325. doi: 10.1007/s00280-005-0160-y.
    1. Fournier T, Medjoubi-N N, Porquet D. Alpha-1-acid glycoprotein. Biochim Biophys Acta. 2000;1482:157–171. doi: 10.1016/S0167-4838(00)00153-9.
    1. Twining SS, Brecher AS. Identification of alpha 1-acid glycoprotein, alpha2-macroglobulin and antithrombin III as components of normal and malignant human tissues. Clin Chim Acta. 1977;75:143–148. doi: 10.1016/0009-8981(77)90510-1.

Source: PubMed

3
Subscribe