Elsevier

Gynecologic Oncology

Volume 135, Issue 2, November 2014, Pages 318-324
Gynecologic Oncology

Inhibition of epithelial ovarian cancer by Minnelide, a water-soluble pro-drug

https://doi.org/10.1016/j.ygyno.2014.08.031Get rights and content

Highlights

  • Minnelide is an effective agent against epithelial ovarian cancer in preclinical models.

  • Given the promising in vitro and in vivo results, Minnelide should progress to further studies in epithelial ovarian cancer.

Abstract

Objective

Minnelide is a water-soluble pro-drug of triptolide, a natural product. The goal of this study was to evaluate the effectiveness of Minnelide on ovarian cancer growth in vitro and in vivo.

Methods

The effect of Minnelide on ovarian cancer cell proliferation was determined by real time electrical impedance measurements. Multiple mouse models with C200 and A2780 epithelial ovarian cancer cell lines were used to assess the efficacy of Minnelide in inhibiting ovarian cancer growth.

Results

Minnelide decreased cell viability of both platinum sensitive and resistant epithelial ovarian cancer cells in vitro. Minnelide with carboplatin showed additive effects in vitro. Minnelide monotherapy increased the survival of mice bearing established ovarian tumors. Minnelide, in combination with carboplatin and paclitaxel, improved overall survival of mice.

Conclusions

Minnelide is a promising pro-drug for the treatment of ovarian cancer, especially when combined with standard chemotherapy.

Introduction

Ovarian cancer has the highest mortality rate of all of the malignancies of the female genital tract. In the United States 21,980 patients were diagnosed with ovarian cancer in 2014 and 14,270 patients died of their disease [1]. The 5 year survival rate for ovarian cancer is 25% for advanced stage disease, which is when 85% of patients are diagnosed. The poor survival is due to a combination of factors including a lack of effective screening and prevention techniques, late stage at diagnosis and lack of treatment options once a patient has failed initial therapies. The current standard first line treatment for ovarian cancer is a combination of a taxane and platinum agent. While 80% of patients will respond to first line treatment, 20% of patients will be platinum resistant and not respond to standard therapy. There are a number of second line agents approved for the treatment of recurrent ovarian cancer including liposomal doxorubicin, topotecan and gemcitabine; however progression free survival times are generally < 12 months [2]. Given these relatively low response rates there is a need for new drug discovery and more effective ways to treat ovarian cancer.

Triptolide, a diterpenoid triepoxide isolated from the plant Tripterygium wilfordii Hook F, has shown efficacy against several different cancer types including pancreatic, melanoma, neuroblastoma and gastric [3], [4], [5]. Because triptolide is not water soluble, the physical properties of this natural product have limited its clinical development. To overcome solubility problems, triptolide was chemically modified to prepare a prodrug which was named Minnelide [6], [7]. Minnelide or 14-O-phosphonooxymethyltriptolide disodium salt is a white powder which is converted into the parent compound triptolide in the presence of alkaline phosphatase which is common in all tissues in the body [8]. The conversion of triptolide to Minnelide has been shown previously both in vitro and in vivo [6]. This compound has been found to be effective against pancreatic cancer as well as osteosarcoma and non-small cell lung cancer both in vitro and in vivo [6], [9], [10]. Since both pancreatic and ovarian cancers metastasize in the peritoneum, we sought to determine the efficacy of Minnelide in inhibiting ovarian cancer growth.

Section snippets

Reagents

Minnelide was synthesized in the Department of Medicinal Chemistry, University of Minnesota as described by Chugh et al. [6]. Carboplatin was purchased from Calbiochem and Paclitaxel from Sigma Aldrich.

Cell lines and cell culture

Platinum sensitive (A2780) and platinum resistant (C200) ovarian cancer cell lines were obtained from Thomas Hamilton, Fox Chase Cancer Center, Philadelphia. Both cell lines were cultured at 37 °C in the presence of 5% CO2 in Roswell Park Memorial Institute (RPMI)-1640 medium supplemented with 10%

Minnelide decreases cell viability of ovarian cancer cells in vitro

In the initial experiment, cell viability was determined following treatment with Minnelide. Both A2780 and C200 cells were inhibited in a concentration dependent manner between 10 and 250 nM (Fig. 1). To assess the efficacy of Minnelide on proliferation of ovarian cancer cell lines, we treated A2780 with 50 nM, 100 nM, 200 nM and 500 nM of Minnelide and C200 cells with 200 nM, 500 nM and 1000 nM of Minnelide. Preliminary studies had shown that C200 cells display a multi-drug resistance phenotype and

Discussion

Previous studies have shown that Minnelide, a water soluble analog of triptolide, is effective in pancreatic, gastric and osteosarcoma [9], [10], [3], [5], [13]. We have found that Minnelide is an active agent against ovarian cancer both in vitro and in vivo.

Chugh et al. have shown Minnelide to be highly active and even curative for early pancreatic tumors [6]. Others have shown that Minnelide exhibits anti-proliferative effects in non-small cell lung cancer by promoting apoptosis in vitro and

Conflict of interest statement

Dr. Saluja is the only author with a conflict of interest in that he has Minneamrita, stock options and acts as a consultant for Minnelide.

References (20)

There are more references available in the full text version of this article.

Cited by (24)

  • Analysis of the role of geranylgeranyl diphosphate synthase 8 from Tripterygium wilfordii in diterpenoids biosynthesis

    2019, Plant Science
    Citation Excerpt :

    The genus Tripterygium is known to produce many types of terpenoids, including diterpenoids, sesquiterpenoids, and triterpenoids, of which triptolide, a labdane-type diterpenoid, shows impressive and effective anti-inflammatory, immunosuppressive and antitumor activities, and potential medicinal value for central nervous system diseases (e.g., Parkinson’s and Alzheimer’s diseases) [2–7]. Moreover, several derivatives of triptolide have undergone clinical trials [8–10]. Due to the rich pharmaceutical potential but low content of triptolide in planta, many studies have been targeted toward the elucidation of the triptolide biosynthetic pathway in order to achieve sustainable and reliable triptolide production via synthetic biology strategies [11–13].

  • Triptolide and Its Derivatives as Cancer Therapies

    2019, Trends in Pharmacological Sciences
    Citation Excerpt :

    TPL has attracted considerable attention among researchers due to its efficacy against various diseases, although its clinical potential remains to be fully explored. TPL inhibits cancer cell growth and exhibits preclinical antitumor activity in a variety of cancers including acute myeloid leukemia (AML) [6], breast cancer [7], osteosarcoma [8], ovarian cancer [9], lung cancer [10], prostate cancer [11], neuroblastoma [12], and multiple gastrointestinal cancers (e.g., colon, liver, stomach, and pancreas cancers) [13–16]. In this review, we focus on the mechanism of action and current state of preclinical and clinical development of TPL and its analogs as anticancer agents.

  • Biochemical and computational evaluation of Triptolide-induced cytotoxicity against NSCLC

    2018, Biomedicine and Pharmacotherapy
    Citation Excerpt :

    Chemotherapeutic agents derived from Traditional Chinese Medicine represent a valuable tool for cancer treatment. Triptolide, a natural compound isolated from the Chinese plant Tripterygium wilfordii, has been reported as being effective against various types of cancers including leukemia [26], pancreatic cancer [27,28], breast cancer [29–31], cholangiocarcinoma [11,32],and ovarian cancer [33]. Although it has been studied extensively, the molecular mechanism underlying Triptolide’s activity is still poorly understood and appears to involve multiple signaling pathways.

View all citing articles on Scopus

The research reported in this publication was supported by the NIH grant P30 CA77598 utilizing the Biostatistics and Bioinformatics Core shared resource of the Masonic Cancer Center, University of Minnesota and by the National Center for Advancing Translational Sciences of the National Institutes of Health Award Number UL1TR000114.

This research was also supported in part by the Minnesota Ovarian Cancer Alliance (MOCA).

View full text