1
|
Zeng Z, Lin C, Wang S, Wang P, Xu W, Ma W, Wang J, Xiang Q, Liu Y, Yang J, Ye F, Xie K, Xu J, Luo Y, Liu SL, Liu H. Suppressive activities of mangiferin on human epithelial ovarian cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153267. [PMID: 32570111 DOI: 10.1016/j.phymed.2020.153267] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/31/2020] [Accepted: 06/13/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Epithelial carcinoma is a subtype of ovarian cancers, with the highest lethality among all ovarian cancer subtypes. Hitherto surgical excision combined with chemotherapy has been the most extensively employed method in clinical treatment. However, the disease relapses very frequently, calling for more effective therapies. Mangiferin, a natural xanthone glucoside, has displayed promising anti-cancer activities by in vitro studies, but its therapeutic value in epithelial ovarian cancer treatment, either by in vivo or in vitro studies, remained to be known. PURPOSE This study aimed to determine the suppressive activities of mangiferin on human epithelial ovarian cancer and elucidate the underlying molecular mechanisms. STUDY DESIGN AND METHODS We employed the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the crystal violet assay to determine the half maximal inhibitory concentration (IC50) values of mangiferin with paclitaxel as a positive control and the inhibitory effects of mangiferin on the proliferation of two human epithelial ovarian cancer cell lines. Wound healing and Transwell assays were used to determine anti-metastastic activities of mangiferin. ES-2 xenograft nude mouse model was used for the in vivo experiments. Western blotting, enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) assays were carried out for evaluating the expression level of matrix metalloproteinase 2 (MMP2) and matrix metalloproteinase 9 (MMP9). RESULTS In the present study, we demonstrated by both in vitro and in vivo assays that mangiferin suppressed the progress of epithelial ovarian cancer in a dose-dependent manner. In the animals treated with mangiferin, the tumor volume and weight were reduced significantly. Analyses of involved molecular events demonstrated that mangiferin down-regulated the expression of metastasis-associated proteins MMP2 and MMP9. CONCLUSION Mangiferin strongly inhibited the progression of human epithelial ovarian cancer by down-regulating MMP2 and MMP9.
Collapse
Affiliation(s)
- Zheng Zeng
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada
| | - Caiji Lin
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Siwen Wang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Pengfei Wang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Wenwen Xu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Wenqing Ma
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Jiali Wang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Qian Xiang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Yiting Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Jiaming Yang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Fan Ye
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Kaihong Xie
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Jian Xu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Yao Luo
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Shu-Lin Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, T2N 4N1, Canada.
| | - Huidi Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada.
| |
Collapse
|
2
|
Zhang M, Hagan CT, Min Y, Foley H, Tian X, Yang F, Mi Y, Au KM, Medik Y, Roche K, Wagner K, Rodgers Z, Wang AZ. Nanoparticle co-delivery of wortmannin and cisplatin synergistically enhances chemoradiotherapy and reverses platinum resistance in ovarian cancer models. Biomaterials 2018; 169:1-10. [PMID: 29631163 DOI: 10.1016/j.biomaterials.2018.03.055] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 03/28/2018] [Accepted: 03/31/2018] [Indexed: 12/12/2022]
Abstract
Most ovarian cancer patients respond well to initial platinum-based chemotherapy. However, within a year, many patients experience disease recurrence with a platinum resistant phenotype that responds poorly to second line chemotherapies. As a result, new strategies to address platinum resistant ovarian cancer (PROC) are needed. Herein, we report that NP co-delivery of cisplatin (CP) and wortmannin (Wtmn), a DNA repair inhibitor, synergistically enhances chemoradiotherapy (CRT) and reverses CP resistance in PROC. We encapsulated this regimen in FDA approved poly(lactic-co-glycolic acid)-poly(ethylene glycol) (PLGA-PEG) NPs to reduce systemic side effects, enhance cellular CP uptake, improve Wtmn stability, and increase therapeutic efficacy. Treatment of platinum-sensitive ovarian cancer (PSOC) and PROC murine models with these dual-drug loaded NPs (DNPs) significantly reduced tumor burden versus treatment with combinations of free drugs or single-drug loaded NPs (SNPs). These results support further investigation of this NP-based, synergistic drug regimen as a means to combat PROC in the clinic.
Collapse
Affiliation(s)
- Maofan Zhang
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, PR China; Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - C Tilden Hagan
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuangzeng Min
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hayley Foley
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xi Tian
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Feifei Yang
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, PR China
| | - Yu Mi
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kin Man Au
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yusra Medik
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kyle Roche
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kyle Wagner
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zachary Rodgers
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Chemistry, Westminster College, New Wilmington, PA 16172, USA
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
Akiyama M, Sowa Y, Taniguchi T, Watanabe M, Yogosawa S, Kitawaki J, Sakai T. Three Combined Treatments, a Novel HDAC Inhibitor OBP-801/YM753, 5-Fluorouracil, and Paclitaxel, Induce G₂ Phase Arrest Through the p38 Pathway in Human Ovarian Cancer Cells. Oncol Res 2017; 25:1245-1252. [PMID: 28117030 PMCID: PMC7841106 DOI: 10.3727/096504017x14850164661097] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Ovarian cancer is the most lethal disease among gynecological malignancies. More effective therapy is required to counter high recurrence rates and chemotherapy resistance. We investigated the efficacy and molecular mechanisms of three combined treatments (TCTs)—a novel histone deacetylase (HDAC) inhibitor OBP-801/YM753, 5-fluorouracil (5-FU), and paclitaxel (PTX)—in human ovarian cancer SKOV-3 and OVCAR-3 cells. The inhibition of cell growth was stronger with TCTs than with each single agent and with two combined treatments. The TCTs significantly induce G2 phase arrest in both cell lines. We then analyzed the molecular mechanisms and found that the TCTs increased the phosphorylation of p38 (Thr180/Tyr182), decreased the expression of CDC25C, and increased the phosphorylation of CDC2 (Tyr15), an inactive form of CDC2. To examine the responsibilities of the p38 pathway for G2 phase arrest induced by the TCTs, we employed the p38 inhibitor SB203580. SB203580 inhibited G2 phase arrest, suppression of CDC25C, and phosphorylation of CDC2 (Tyr15) induced by the TCTs. These results suggest that the TCTs can induce G2 phase arrest through activation of the p38 signaling pathway. We therefore believe that this combination is promising as a novel therapeutic strategy against ovarian cancer.
Collapse
|
4
|
Asami Y, Jang JH, Soung NK, He L, Moon DO, Kim JW, Oh H, Muroi M, Osada H, Kim BY, Ahn JS. Protuboxepin A, a marine fungal metabolite, inducing metaphase arrest and chromosomal misalignment in tumor cells. Bioorg Med Chem 2012; 20:3799-806. [DOI: 10.1016/j.bmc.2012.04.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 04/19/2012] [Accepted: 04/19/2012] [Indexed: 11/30/2022]
|
5
|
|
6
|
Rudin D, Li L, Niu N, Kalari KR, Gilbert JA, Ames MM, Wang L. Gemcitabine Cytotoxicity: Interaction of Efflux and Deamination. ACTA ACUST UNITED AC 2011; 2:1-10. [PMID: 21804948 DOI: 10.4172/2157-7609.1000107] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gemcitabine is a cytidine analogue used in the treatment of various solid tumors. Little is known about how gemcitabine and its metabolites are transported out of cells. We set out to study the efflux of gemcitabine and the possible consequences of that process in cancer cells. We observed the efflux of gemcitabine and its deaminated metabolite, 2',2'-difluorodeoxyuridine (dFdU) using high performance liquid chromatography and tandem mass spectrometry (LC-MS/MS) after gemcitabine treatment. Non-selective ABCC-transport inhibition with probenecid significantly increased intracellular dFdU concentrations, with a similar trend observed with verapamil, a non-selective ABCB1 and ABCG2 transport inhibitor. Neither probenecid nor verapamil altered intracellular gemcitabine levels after the inhibition of deamination with tetrahydrourudine, suggesting that efflux of dFdU, but not gemcitabine, was mediated by ABC transporters. MTS assays showed that probenecid increased sensitivity to gemcitabine. While dFdU displayed little cytotoxicity, intracellular dFdU accumulation inhibited cytidine deaminase, resulting in increased gemcitabine levels and enhanced cytotoxicity. Knockdown of ABCC3, ABCC5 or ABCC10 individually did not significantly increase gemcitabine sensitivity, suggesting the involvement of multiple transporters. In summary, ABCC-mediated efflux may contribute to gemcitabine resistance through increased dFdU efflux that allows for the continuation of gemcitabine deamination. Reversing efflux-mediated gemcitabine resistance may require broad-based efflux inhibition.
Collapse
Affiliation(s)
- Dan Rudin
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905
| | | | | | | | | | | | | |
Collapse
|
7
|
Engineering of self-assembled nanoparticle platform for precisely controlled combination drug therapy. Proc Natl Acad Sci U S A 2010; 107:17939-44. [PMID: 20921363 DOI: 10.1073/pnas.1011368107] [Citation(s) in RCA: 422] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The genomic revolution has identified therapeutic targets for a plethora of diseases, creating a need to develop robust technologies for combination drug therapy. In the present work, we describe a self-assembled polymeric nanoparticle (NP) platform to target and control precisely the codelivery of drugs with varying physicochemical properties to cancer cells. As proof of concept, we codelivered cisplatin and docetaxel (Dtxl) to prostate cancer cells with synergistic cytotoxicity. A polylactide (PLA) derivative with pendant hydroxyl groups was prepared and conjugated to a platinum(IV) [Pt(IV)] prodrug, c,t,c-[Pt(NH(3))(2)(O(2)CCH(2)CH(2)COOH)(OH)Cl(2)] [PLA-Pt(IV)]. A blend of PLA-Pt(IV) functionalized polymer and carboxyl-terminated poly(D,L-lactic-co-glycolic acid)-block-poly(ethylene glycol) copolymer in the presence or absence of Dtxl, was converted, in microfluidic channels, to NPs with a diameter of ∼100 nm. This process resulted in excellent encapsulation efficiency (EE) and high loading of both hydrophilic platinum prodrug and hydrophobic Dtxl with reproducible EEs and loadings. The surface of the NPs was derivatized with the A10 aptamer, which binds to the prostate-specific membrane antigen (PSMA) on prostate cancer cells. These NPs undergo controlled release of both drugs over a period of 48-72 h. Targeted NPs were internalized by the PSMA-expressing LNCaP cells via endocytosis, and formation of cisplatin 1,2-d(GpG) intrastrand cross-links on nuclear DNA was verified. In vitro toxicities demonstrated superiority of the targeted dual-drug combination NPs over NPs with single drug or nontargeted NPs. This work reveals the potential of a single, programmable nanoparticle to blend and deliver a combination of drugs for cancer treatment.
Collapse
|
8
|
Seliger G, Mueller LP, Kegel T, Kantelhardt EJ, Grothey A, Groe R, Strauss HG, Koelbl H, Thomssen C, Schmoll HJ. Phase 2 trial of docetaxel, gemcitabine, and oxaliplatin combination chemotherapy in platinum- and paclitaxel-pretreated epithelial ovarian cancer. Int J Gynecol Cancer 2009; 19:1446-53. [PMID: 20009905 DOI: 10.1111/igc.0b013e3181b62f38] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND This phase 2 trial was designed to evaluate the efficacy and toxicity of a combination of docetaxel, gemcitabine, and oxaliplatin for platinum- and paclitaxel-pretreated epithelial ovarian cancer. PATIENTS AND METHODS Heavily pretreated patients (N = 30; median age, 61 years) received docetaxel, 55 mg/m2; gemcitabine, 500 mg/m2 (day 1); and oxaliplatin, 70 mg/m2 (day 2) biweekly. Twelve patients had platinum-sensitive disease, and 18 patients had platinum-resistant disease. RESULTS Median follow-up was 18.6 months. No differences in patient characteristics were observed between patients with carboplatinum-sensitive and carboplatinum-resistant disease. In patients with carboplatin-sensitive disease, an overall response (OR) of 83.3%, a progression-free survival of 10.6 months, and an overall survival of 18.9 months were observed. In patients with carboplatinum-resistant disease, an OR was seen in 38.9% with a progression-free survival of 5.3 months and an overall survival of 16.3 months. Patients with platinum-refractory disease (progression under previous carboplatinum therapy, n = 13) had an OR of 23%, whereas patients with objective response but relapse less than 6 months after carboplatinum therapy had an OR of 80.0%. Grade 3 and 4 toxicities were only observed for anemia (6.7%), neutropenia (20.0%), thrombopenia, peripheral neuropathy, and diarrhea (16.7%). No neutropenic fever or treatment-related death occurred. CONCLUSIONS In comparison with current standard protocols, a combination of docetaxel, gemcitabine, and oxaliplatin showed considerably higher efficacy without remarkable increased toxicity; particularly for patients with early relapse after a platinum-containing therapy.
Collapse
Affiliation(s)
- Gregor Seliger
- Department of Gynecology, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Tsimberidou AM, Braiteh F, Stewart DJ, Kurzrock R. Ultimate fate of oncology drugs approved by the us food and drug administration without a randomized Trial. J Clin Oncol 2009; 27:6243-50. [PMID: 19826112 DOI: 10.1200/jco.2009.23.6018] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To approve a new anticancer drug, the US Food and Drug Administration often requires randomized trials. However, several oncology drugs have been approved on the basis of objective end points without a randomized trial. We reviewed the long-term safety and efficacy of such agents. METHODS We searched the Web site of the US Food and Drug Administration's Center for Drug Evaluation and Research and MEDLINE for initial applications of investigational anticancer drugs from 1973 through 2006. RESULTS Overall, 68 oncology drugs, excluding hormone therapy and supportive care, were approved, including 31 without a randomized trial. For these 31 drugs, a median of two clinical trials (range, one to seven) and 79 patients (range, 40 to 413) were used per approval. Objective response was the most common end point used for approval; median response rate was 33% (range, 11% to 90%). Thirty drugs are still fully approved. United States marketing authorization for one drug, gefitinib (an epidermal growth factor receptor [EGFR] inhibitor), was rescinded after a randomized trial showed no survival improvement; however, this trial was performed in unselected patients, and it was subsequently demonstrated that patients with EGFR mutation are more likely to respond. Nineteen of the 31 drugs have additional uses (per National Comprehensive Cancer Network or National Cancer Institute Physician Data Query guidelines), and subsequent formal US Food and Drug Administration approvals were obtained for 11 of these (range, one to 18 new indications). No drug has demonstrated safety concerns. CONCLUSION Nonrandomized clinical trials with definitive end points can yield US Food and Drug Administration approvals, and these drugs have a reassuring record of long-term safety and efficacy.
Collapse
Affiliation(s)
- Apostolia-Maria Tsimberidou
- The University of Texas M. D. Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Unit 455, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
10
|
Frederick PJ, Kendrick JE, Straughn JM, Della Manna DL, Oliver PG, Lin HY, Grizzle WE, Stockard CR, Alvarez RD, Zhou T, LoBuglio AF, Buchsbaum DJ. Effect of TRA-8 anti-death receptor 5 antibody in combination with chemotherapy in an ex vivo human ovarian cancer model. Int J Gynecol Cancer 2009; 19:814-9. [PMID: 19574765 DOI: 10.1111/igc.0b013e3181a2a003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVES To investigate the cytotoxicity of TRA-8, an antibody that specifically binds death receptor 5, alone and in combination with chemotherapy, using an ex vivo human ovarian cancer model. MATERIALS AND METHODS Twenty-six ovarian cancer specimens were obtained during ovarian cancer debulking, and tumor slices were prepared with the Krumdieck tissue slicer. The tumor slices were exposed to varying concentrations of TRA-8, carboplatin/paclitaxel, or the combination of TRA-8 and chemotherapy. Using nonlinear modeling, dose-response curves and IC50 values were generated for specimens treated with TRA-8. The additive and synergistic cytotoxic effects of chemotherapy combination with TRA-8 were evaluated in specimens. In addition to adenosine triphosphate viability assays, the treated and untreated slices were assessed by immunohistochemistry to confirm apoptosis induction. RESULTS Specimens from 13 patients yielded TRA-8-induced IC50 values. Of these specimens, 15% were found to be sensitive to TRA-8-induced cytotoxicity at IC50 doses less than 500 ng/mL. Specimens from 13 patients underwent combination treatment with TRA-8 and carboplatin/paclitaxel. Of these specimens, 77% exhibited additive cytotoxicity in comparison with those treated with either agent alone, whereas 15% exhibited synergistic cytotoxicity. Immunohistochemical analysis of terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling and cleaved caspase 3 staining demonstrated a dose-dependent increase in apoptosis with the combination treatment. CONCLUSIONS This study demonstrates the efficacy of the death receptor monoclonal antibody TRA-8 in combination with conventional chemotherapy in an ex vivo human ovarian cancer model. This model can be used to assess cytotoxicity of novel agents in combination with chemotherapy in ovarian cancer.
Collapse
MESH Headings
- Adenocarcinoma/immunology
- Adenocarcinoma/pathology
- Adenocarcinoma/therapy
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Apoptosis/drug effects
- Carboplatin/administration & dosage
- Carcinoma, Papillary/immunology
- Carcinoma, Papillary/pathology
- Carcinoma, Papillary/therapy
- Combined Modality Therapy
- Cystadenocarcinoma, Serous/immunology
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/therapy
- Dose-Response Relationship, Drug
- Drug Synergism
- Endometrial Neoplasms/immunology
- Endometrial Neoplasms/pathology
- Endometrial Neoplasms/therapy
- Female
- Humans
- Immunoenzyme Techniques
- In Situ Nick-End Labeling
- Inhibitory Concentration 50
- Middle Aged
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/therapy
- Paclitaxel/administration & dosage
- Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
- Treatment Outcome
Collapse
Affiliation(s)
- Peter J Frederick
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zhang N, Zhang H, Xia L, Zheng Y, Yu Y, Zhu Y, Chen G, Di W. NSC606985 induces apoptosis, exerts synergistic effects with cisplatin, and inhibits hypoxia-stabilized HIF-1alpha protein in human ovarian cancer cells. Cancer Lett 2009; 278:139-144. [PMID: 19339107 DOI: 10.1016/j.canlet.2008.12.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Revised: 12/15/2008] [Accepted: 12/16/2008] [Indexed: 11/19/2022]
Abstract
The camptothecins, which target the intranuclear enzyme topoisomerase I, have advanced to the forefront of several areas of developmental chemotherapy of cancers. In the present study, we investigated the potential anti-human ovarian cancer effects of NSC606985, a novel and rarely studied camptothecin analog, and its combination with cisplatin (CDDP). Human ovarian cancer cell line COC1 cells were treated with different nanomolar of NSC606985 with or without CDDP, and cell growth and apoptosis were evaluated, respectively, by MTT assay and annexin-V assay on flow cytometry. Chou-Talalay analysis was used to evaluate combined effect of NSC606985 and CDDP. Western blot was used to detect protein kinase Cdelta (PKCdelta), caspase-3 and hypoxia-inducible factor-1alpha (HIF-1alpha) proteins. Our results showed that NSC606985 at nanomolar concentration induced apoptosis with the activation of PKCdelta in COC1 cells. Especially, NSC606985 presented the significant combined effects on COC1 cells in terms of growth inhibition and apoptosis induction. In addition, NSC606985 significantly antagonized the accumulation of HIF-1alpha stabilized by hypoxia or hypoxia-mimetic agent. These results suggest that NSC606985 and its combination with CDDP present the therapeutic potential on ovarian cancer, and deserve further preclinical and clinical studies.
Collapse
Affiliation(s)
- Ning Zhang
- Dept. of Obstetrics and Gynecology, Institute of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Hanwen Zhang
- Dept. of Obstetrics and Gynecology, Institute of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Li Xia
- Dept. of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Zheng
- Dept. of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yun Yu
- Dept. of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuanshan Zhu
- Dept. of Medicine/Endocrinology, Weill Medical College of Cornell University, New York, NY, 10021, USA
| | - Guoqiang Chen
- Dept. of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Wen Di
- Dept. of Obstetrics and Gynecology, Institute of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China.
| |
Collapse
|
12
|
Gemici G, Cinçin A, Değertekin M, Oktay A. Paclitaxel-induced ST-segment elevations. Clin Cardiol 2009; 32:E94-6. [PMID: 19382279 PMCID: PMC6653473 DOI: 10.1002/clc.20291] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 08/24/2007] [Indexed: 11/08/2022] Open
Abstract
A 51-year-old woman presented with severe chest pain minutes after starting intravenous paclitaxel as a part of the systemic chemotherapy due to ovarian carcinoma. The electrocardiogram (ECG) revealed sinus rhythm with ST-segment elevations in inferior and anterior leads. The ST-segment elevations resolved immediately after sublingual nitroglycerine. Cardiac troponin T and CPK MB levels remained in the normal range at repeat measurements. It was presumed that in spite of standard premedication, paclitaxel had induced acute coronary syndrome with ST-segment elevations in this patient.
Collapse
|
13
|
|
14
|
Brito NMB, Brito MVH, Carvalho RDKV, Matos LTDMB, Veloso TS, Vasconcelos DMD, Brito RB. Experimental inoculation model of Walker 256 carcinoma into vagina and cervix uteri of female rats. Acta Cir Bras 2009; 22:495-8. [PMID: 18235940 DOI: 10.1590/s0102-86502007000600014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Accepted: 07/26/2007] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To establish an inoculation model of Walker 256 carcinoma on cervix uteri and vagina of rats. METHODS Fifteen female rats were used, and assigned to three groups each one with five rats: group A - rats with 4 x 10(6) cells of Walker 256 carcinoma without acid acetic inoculation; group B - rats with 2 x 10(6) cells of Walker 256 carcinoma with acid acetic inoculation and group C: rats with 4 x 10(6) cells of Walker 256 carcinoma with acid acetic inoculation. The day before tumor cells inoculation the rats from groups B and C were anaesthetized with diethylether and 0,3 ml of acetic acid was inoculated into their vaginas. Tumor cell inoculation into the vagina and cervix was done under general anesthesia with diethylether. Then a endocervical brush was used to scrape the vaginal wall and after that 0,3 ml of the liquid containing tumor cells was inoculated on the vagina and cervix. For the tumor analysis, animals were euthanized at day 12 following tumor cell implantation by an excessive inhalation of diethylether. Tumor was resected entirely and weighed and the tumors were then sectioned and counter stained with hematoxylin and eosin for histopathologic evaluation. It was also calculated the percentage of tumor equivalent to the body weight by the formula: P= tumor weight / body weight x 100. Data were analyzed by one-way analysis of variance - ANOVA. P values < 0.05 were taken to indicate statistical significance. RESULTS Implantation and growth on GB and GC was 100% and on GA 20%. There was no statistical difference between GB and GC averages. CONCLUSION According to the methods used, the Walker 256 carcinoma inoculation model into vagina and cervix have an implantation and growth rate of 100% when associated with previous acid acetic inoculation and there is no behavioral difference between using 2 x 10(6) or 4 x 10(6) cells on its inoculation.
Collapse
|
15
|
Frederick PJ, Straughn JM, Alvarez RD, Buchsbaum DJ. Preclinical studies and clinical utilization of monoclonal antibodies in epithelial ovarian cancer. Gynecol Oncol 2009; 113:384-90. [PMID: 19232697 DOI: 10.1016/j.ygyno.2009.01.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 01/15/2009] [Accepted: 01/17/2009] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To present an overview of selected monoclonal antibodies (mAbs) that have been studied in epithelial ovarian cancer with a focus on combination treatment with conventional chemotherapy. METHODS The authors perform a narrative review of the literature. Preclinical studies that provided rationale for mAb use are examined, and selected clinical trials that evaluated efficacy and tolerability are reviewed. RESULTS Numerous mAbs have been utilized in epithelial ovarian cancer, including bevacizumab (anti-vascular endothelial growth factor), trastuzumab (anti-human epidermal growth factor-2), cetuximab (anti-epidermal growth factor receptor), and oregovomab (anti-CA125). Favorable preclinical results have lead to the development of a number of clinical trials. Side-effects have been minimal and combination therapy has been well-tolerated. Efficacy has been variable in the clinical trials. CONCLUSIONS Targeted treatment with mAbs in conjunction with cytotoxic chemotherapy has been an important research area during the last decade. This therapeutic approach holds promise for improved outcomes in patients with ovarian cancer.
Collapse
Affiliation(s)
- Peter J Frederick
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL 35249, USA.
| | | | | | | |
Collapse
|
16
|
Royer B, Jullien V, Guardiola E, Heyd B, Chauffert B, Kantelip JP, Pivot X. Population Pharmacokinetics and Dosing Recommendations for Cisplatin during Intraperitoneal Peroperative Administration. Clin Pharmacokinet 2009; 48:169-80. [DOI: 10.2165/00003088-200948030-00003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
Sehouli J, Stengel D, Oskay-Oezcelik G, Zeimet AG, Sommer H, Klare P, Stauch M, Paulenz A, Camara O, Keil E, Lichtenegger W. Nonplatinum Topotecan Combinations Versus Topotecan Alone for Recurrent Ovarian Cancer: Results of a Phase III Study of the North-Eastern German Society of Gynecological Oncology Ovarian Cancer Study Group. J Clin Oncol 2008; 26:3176-82. [PMID: 18591555 DOI: 10.1200/jco.2007.15.1258] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PurposeThe management of recurrent ovarian cancer remains controversial. Single-agent topotecan is an established treatment option, and preliminary evidence suggests improved tumor control by combining topotecan with etoposide or gemcitabine.Patients and MethodsWomen with relapsed ovarian cancer after primary surgery and platinum-based chemotherapy were randomly assigned to topotecan monotherapy 1.25 mg/m2/d, topotecan 1.0 mg/m2plus oral etoposide 50 mg/d, or topotecan 0.5 mg/m2/d plus gemcitabine 800 mg/m2on day 1 and 600 mg/m2on day 8 every 3 weeks. Patients were stratified for platinum-refractory and platinum-sensitive disease according to a recurrence-free interval of less or more than 12 months, respectively. The primary end point was overall survival. Secondary end points included progression-free survival, objective response rates, toxicity, and quality of life (as measured by the European Organisation for Research and Treatment of Cancer [EORTC] 30-item Quality-of-Life Questionnaire).ResultsThe trial enrolled 502 patients with a mean age of 60.5 years (± 10.2 years), 208 of whom were platinum resistant. Median overall survival was 17.2 months (95% CI, 13.5 to 21.9 months) with topotecan, 17.8 months (95% CI, 13.7 to 20.0 months) with topotecan plus etoposide (log-rank P = .7647), and 15.2 months (95% CI, 11.3 to 20.9 months) with topotecan plus gemcitabine (log-rank P = .2344). Platinum-sensitive patients lived significantly longer than platinum-refractory patients (21.9 v 10.6 months). The median progression-free survival was 7.0, 7.8, and 6.3 months, respectively. Objective response rates were 27.8%, 36.1%, and 31.6%, respectively. Patients under combined treatment were at higher risk of severe thrombocytopenia.ConclusionNonplatinum topotecan combinations do not provide a survival advantage over topotecan alone in women with relapsed ovarian cancer.
Collapse
Affiliation(s)
- Jalid Sehouli
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Dirk Stengel
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Guelten Oskay-Oezcelik
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Alain G. Zeimet
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Harald Sommer
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Peter Klare
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Martina Stauch
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Axel Paulenz
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Oumar Camara
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Elke Keil
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| | - Werner Lichtenegger
- From the Department of Gynecology, Charité University Medical Center; The Center for Clinical Research, Unfallkrankenhaus; Helios-Klinikum Berlin, Germany; Praxis for Gynecologic Oncology, Berlin; Ernst-Moritz-Arndt University of Greifswald, Greifswald; University of Jena, Jena; Ludwig-Maximilian-Universität of Munich, Munich; Klinikum Ernst-von-Bergmann Potsdam, Potsdam; Practice for Oncology and Hematology, Kronach, Germany; and the University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
18
|
Abstract
Irinotecan, a chemotherapeutic agent against various solid tumors, is a prodrug requiring activation to SN-38. Irinotecan's complex pharmacokinetics potentially allow for many genetic sources of variability. We explored relationships between pharmacokinetic pathways and polymorphisms in genes associated with irinotecan's metabolism and transport. We fitted a seven-compartment pharmacokinetic model with enterohepatic recirculation (EHR) to concentrations of irinotecan and metabolites SN-38, SN-38 glucuronide (SN-38G), and aminopentanoic acid (APC). Principal component analysis (PCA) of patient-specific parameter estimates produced measures interpretable along pathways. Nine principal components provided good characterization of the overall variation. Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. The component characterizing irinotecan's compartments was associated with HNF1alpha and ABCC2 polymorphisms. The exploratory analysis with PCA in this pharmacogenetic analysis was able to identify known associations and may have allowed identification of previously uncharacterized functional polymorphisms.
Collapse
|