Potentiation of flutamide-induced hepatotoxicity in mice by Xian-Ling-Gu-Bao through induction of CYP1A2

https://doi.org/10.1016/j.jep.2021.114299Get rights and content

Highlights

  • Xian-Ling-Gu-Bao (XLGB) is an inducer of CYP1A2 enzyme activity.

  • XLGB increases the formation of toxic flutamide metabolite.

  • The combination of XLGB and flutamide increases the hepatotoxicity in mice.

  • This study suggests potential side effects of the drug-drug interaction between flutamide and XLGB in clinical practice.

Abstract

Ethnopharmacological relevance

Xian-Ling-Gu-Bao (XLGB) Fufang is herbal formula widely used to treat osteoporosis and other bone disorders. Because of its commonality in the clinical use, there is a safety concern over the use of XLGB combined with other androgen deprivation therapy (ADT) drugs such as flutamide (FLU) that is associated with reduced bone density. To date, there have been no evaluations on the side effects of the drug-drug interaction between XLGB and FLU.

Aim of the study

The present study was designed to investigate the hepatotoxicity in the context of the combined treatment of XLGB and FLU in a mouse model, and to determine whether the metabolic activation of FLU through induction of CYP1A2 plays a role in the increased hepatoxicity caused by the combination of XLGB and FLU.

Materials and methods

C57 mice were administered with either XLGB (6,160 mg/kg), FLU (300 mg/kg), or with the combination of the two drugs. Animals were treated with XLGB for 5 days before the combined administration of XLGB and FLU for another 4 days. The serum of mice from single or the combined administration groups was collected for biochemical analysis. The mouse liver was collected to examine liver morphological changes, evaluate liver coefficient, as well as determine the mRNA expression of P450 isozymes (Cyp1a2, Cyp3a11 and Cyp2c37). For metabolism analysis, mice were treated with XLGB, FLU, or the combination of XLGB and FLU for 24 h. The urine samples were collected for the analysis of FLU-NAC conjugate by UPLC-Q-Orbitrap MS. The liver microsomes were prepared from fresh livers to determine the activity of metabolizing enzyme CYP1A2.

Results

The combined treatment of XLGB and FLU caused loss of mice body weight and elicited significant liver toxicity as evidenced by an increased liver coefficient and serum lactate dehydrogenase (LDH) activity as well as pathological changes of fatty lesion of liver tissue. FLU increased hepatic expression of Cyp1a2 mRNA that was further elevated in the liver of mice when administered with both FLU and XLGB. Treatment of FLU resulted in an increase in the expression of Cyp3a11 mRNA that was negated when mice were co-treated with FLU and XLGB. No significant difference in Cyp2c37 mRNA expression was observed among the different treatment groups as compared to the control. Analysis of metabolic activity showed that the combined administration caused a synergic effect in elevating the activity of the CYP1A2 enzyme. Mass spectrometry analysis identified the presence of FLU reactive metabolite derived FLU-NAC conjugate in the urine of mice treated with FLU. Strikingly, about a two-fold increase of the FLU-NAC conjugate was detected when treated with both FLU and XLGB, indicating an elevated amount of toxic metabolite produced from FLU in the present of XLGB.

Conclusion

FLU and XLGB co-treatment potentiated FLU-induced hepatoxicity. This increased hepatoxicity was mediated through the induction of CYP1A2 activity which in turn enhanced bioactivation of FLU leading to over production of FLU-NAC conjugate and oxidative stress. These results offer warnings about serious side effects of the FLU-XLGB interaction in the clinical practice.

Introduction

Prostate cancer is a common malignant tumor in elderly men, and its occurrence and development are closely related to androgen. Anti-androgen deprivation therapy (ADT) has been the most widely used systemic treatment for prostate cancer in clinical practice. Although it is necessary, the long-term use of ADT introduces a variety of adverse events, including osteoporosis with decreased bone mineral density (Chen et al., 2019; Kim et al., 2019). A study of 390 prostate cancer patients followed up for 10 years showed that the incidence rate of osteoporosis in patients who received ADT was 80%, while the rate in patients who did not receive ADT was 35.4% (Morote et al., 2007). Flutamide (FLU) is a first-line drug for ADT, which has been used primarily for the treatment of prostate cancer (Bahnson, 2007). It is also prescribed for other androgen-dependent conditions such as in the treatment of women with polycystic ovary syndrome (PCOS) (Vincenzo et al., 1998). Though highly effective, the treatment of FLU interferes with bone metabolism and causes reduced bone mineral density. Animal studies have shown that FLU-mediated androgen blockade induces osteopenia in rats primarily due to reduced bone formation (Goulding and Gold, 1993). In human studies of patients with PCOS, a 6-month course of therapy with FLU revealed a significant reduction in bone mineral density along with elevated urinary calcium excretion (Moghetti et al., 1999). Considering the serious effects of ADT on bone mass density and the increased risk of fracture, it has been essential to precisely evaluate bone quantity and manage the adverse effects of ADT therapy on bone health. In this regard, ADT therapy has often been used in combination with osteoporosis treatment drugs in clinical practice.

Xian-Ling-Gu-Bao (XLGB) is a well-known patent herbal medicine formula, which is composed of six herbal medicines as follows: foliage of Epimedium brevicornu Maxim (Berberidaceae, Yin Yang Huo in Chinese, 70% g/g), stem of Dipsacus asper Wall. ex C.B. Clarke (Caprifoliaceae, Xu Duan in Chinese, 10% g/g), fructus of Cullen corylifolium (L.) Medik (Fabaceae, Bu Gu Zhi in Chinese, 5% g/g), root and rhizome of Salvia miltiorrhiza Bunge (Lamiaceae, Dan Shen in Chinese, 5% g/g), rhizome of Anemarrhena asphodeloides Bunge (Asparagaceae, Zhi Mu in Chinese, 5% g/g), and root and rhizome of Rehmannia glutinosa (Gaertn.) DC. (Orobanchaceae, Di Huang in Chinese, 5% g/g) (Wu et al., 2019). XLGB is herbal formulation approved by the China Food and Drug Administration (CFDA; China, Z20025337). It has been widely used in the clinic to treat osteoporosis, osteoarthritis, fractures and other diseases. The efficacy and quality control of XLGB have been widely described previously (Cheng et al., 2013; Guan et al., 2011; Yao et al., 2017; Zhu et al., 2012). Chinese herbal medicine has a long history of medical applications both in prescribed and self-prescribed medications for various acute and chronic conditions. Patients generally have a high acceptance of Chinese patent medicines in the belief that Chinese herbal medicines are associated with lower side effects. Combinations of Chinese patent medicines with other drugs are therefore quite common. However, herbal medicine presents a greater risk of adverse effects compared to other complementary therapies, especially in the context of potential drug interactions (Tachjian et al., 2010; Vickers et al., 2001). XLGB as a preventive drug for osteoporosis is often chosen to be used in combination with FLU in the treatment of prostate cancer patients, yet the safety of this combination has not been evaluated.

The combined use of drugs can increase the possibility of pharmacokinetic or pharmacodynamic interactions. Clinical studies have demonstrated that the combination of Chinese patent medicines and other drugs can enhance or reduce the efficacy and toxicity of drugs (Liu et al., 2011; Xu et al., 2019). Hepatotoxicity of FLU or XLGB has been reported in a number of clinical applications (Giorgetti et al., 2017; Manso et al., 2006; Tavakkoli et al., 2011; Yang and Peng, 2013; Yang and Zhou, 2007; Zheng et al., 2014). Will the combination of two hepatotoxic drugs further increase the risk? At present, the safety risks associated with the combination of FLU and XLGB in clinical practice remain unknown. Drug interactions are complex in clinical practice and often difficult to identify and predict. Given the commonality of the clinical usage of FLU and XLGB, it is necessary to evaluate the toxicity of the interaction of the combination of FLU and XLGB in an animal model.

The toxicity of FLU is mainly caused by its metabolites, and the key metabolic enzymes of FLU are cytochrome P450 1A2 (CYP1A2) and cytochrome P450 3A4 (CYP3A4) (Bahnson, 2007; Ohbuchi et al., 2009). In our previous studies, XLGB was found to induce the expression of Cyp1a2 mRNA in mouse liver (Ding et al., 2019). On the basis of these findings, it is likely that there may exist a high possibility of drug interaction between XLGB and FLU when used in combination. Therefore, in the present study, we evaluated the hepatotoxicity of XLGB combined with FLU in mice. Using mass spectrometry analysis, we further explored the underlying mechanism by dissecting the reactive metabolites of FLU in combination with XLGB.

Section snippets

Reagents and materials

XLGB (Batch number: 1704001, specification: 0.5 g × 50 capsules) was obtained from Tongjitang Guizhou Pharmaceutical Co. Ltd., Guizhou, China. The preparation process of the manufacturer is as follows: foliage of E. brevicornu Maxim, root and rhizome of R. glutinosa (Gaertn.) DC., and rhizome of A. asphodeloides Bunge were decocted in water for three times, and all decoctions were combined. Decoctions were concentrated to a thick paste with a relative density of 1.35–1.38 g/mL. Root and rhizome

Hepatotoxicity of XLGB combined with FLU in mice

Treatment of mice with FLU resulted in the loss of body weight, while the combination of FLU with XLGB further worsened the effect of FLU on body weight (Fig. 1A). The examination of liver tissues found that the mice in XLGB + FLU group had significantly enlarged livers as compared with the control group. The evaluation of liver coefficient revealed that there was a marked increase in the liver coefficient in mice treated with the combination of FLU and XLGB, while no noticeable change was

Discussion

FLU hepatoxicity has been demonstrated in both in vitro and in vivo models (Audrey et al., 2014; Maruf and O'Brien, 2014). Although the specific mechanism underlying the FLU-induced hepatoxicity remains to be fully elucidated, the bioactivation of FLU leading to redox cycling and GSH depletion is proposed to be responsible for FLU toxicity (Kang et al., 2008; Wen et al., 2008). In the present study, we demonstrated that the combined administration of XLGB and FLU aggravates FLU-elicited

Conclusion

In the present study we demonstrated that the combination of XLGB and FLU increased the hepatotoxicity in mice. The underlying mechanism may be related to the increased activation of FLU through induction of CYP1A2 enzyme activity to produce more reactive metabolites in the context of combined treatment of FLU and XLGB. This study offers warnings about serious side effects of the drug-drug interaction between FLU and XLGB in the clinical practice. The combined usage of these two drugs needs to

Declaration of competing interest

The authors declare that they have no conflict of interest.

Acknowledgments

The work was supported by the National Natural Science Foundation of China (Grant No. 81760678), and the First-Class Disciplines Fund of Education Department of Guizhou Province (Grant No. GNYL [2017]006 YLXKJS-YS-05).

We would like to thank Associate Professor Weian Deng from the Pathology Department of First Affiliated Hospital of Zunyi Medical University for the help on the histopathological evaluation of the liver tissues.

References (55)

  • W. Wu et al.

    Xian-Ling-Gu-Bao induced inflammatory stress rat liver injury: inflammatory and oxidative stress playing important roles

    J. Ethnopharmacol.

    (2019)
  • L.Q. Yang et al.

    Severe liver damage caused by Xian-ling-gu-bao capsule: a case

    Chin. J. Pharmacovigilance

    (2013)
  • G.M. Amaya et al.

    Cytochromes P450 1A2 and 3A4 catalyze the metabolic activation of sunitinib

    Chem. Res. Toxicol.

    (2018)
  • Legendre Audrey et al.

    Investigation of the hepatotoxicity of flutamide: pro-survival/apoptotic and necrotic switch in primary rat hepatocytes characterized by metabolic and transcriptomic profiles in microfluidic liver biochips

    Toxicol. Vitro

    (2014)
  • A.L. Ball et al.

    Identification of the additional mitochondrial liabilities of 2-hydroxyflutamide when compared with its parent compound, flutamide in HepG2 cells

    Toxicol. Sci.

    (2016)
  • A. Berson et al.

    Metabolic activation of the nitroaromatic antiandrogen flutamide by rat and human cytochromes P-450, including forms belonging to the 3A and 1A subfamilies

    J. Pharmacol. Exp. Therapeut.

    (1993)
  • Z. Bibi

    Role of cytochrome P450 in drug interactions

    Nutr. Metabol.

    (2008)
  • J.F. Chen et al.

    Androgens and androgen receptor actions on bone health and disease: from androgen deficiency to androgen therapy

    Cells

    (2019)
  • Y.N. Ding et al.

    The hepatotoxicity of Xian-Ling-Gu-Bao and its combination with Omeprazole in mice

    J. Zunyi Med. Univ.

    (2019)
  • D. Fau et al.

    Toxicity of the antiandrogen flutamide in isolated rat hepatocytes

    J. Pharmacol. Exp. Therapeut.

    (1994)
  • R. Giorgetti et al.

    Flutamide-induced hepatotoxicity: ethical and scientific issues

    Eur. Rev. Med. Pharmacol. Sci.

    (2017)
  • A. Goulding et al.

    Flutamide-mediated androgen blockade evokes osteopenia in the female rat

    J. Bone Miner. Res.

    (1993)
  • L.I. Jia-Nan et al.

    Inhibitory effect of shikonin on cytochrome P450 1A2 and carboxylesterase 2 mediated metabolism of flutamide in vitro

    Chin. J. Pharmacol. Toxicol.

    (2018)
  • W. Jian et al.

    Rapid detection and characterization of in vitro and urinary N-acetyl-L-cysteine conjugates using quadrupole-linear ion trap mass spectrometry and polarity switching

    Chem. Res. Toxicol.

    (2009)
  • P. Kang et al.

    Identification of a novel glutathione conjugate of flutamide in incubations with human liver microsomes

    Drug Metab. Dispos.

    (2007)
  • P. Kang et al.

    Bioactivation of flutamide metabolites by human liver microsomes

    Drug Metabol. Disposition Biol. Fate Chem

    (2008)
  • Wen Kexin et al.

    Effect of flutamide exposed during pregnancy on reproductive organs development and oxidative stress responsein in male mice offspring

    J. Hyg. Res.

    (2018)
  • Cited by (0)

    View full text