1
|
Khattak MA, Iqbal Z, Fazli N, Khan FU, Khan S, Gohar S, Sakhi M, Iqbal M, Pervez S. Using the Design of Experiments for Development, Validation, and Optimization of HPLC-UV Assay of Tamoxifen Citrate in Polymeric Nanoparticles. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02789-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
2
|
Huang S, Wang H, Chen W, Zhan M, Xu S, Huang X, Lin R, Shen H, Wang J. Tamoxifen inhibits cell proliferation by impaired glucose metabolism in gallbladder cancer. J Cell Mol Med 2020; 24:1599-1613. [PMID: 31782270 PMCID: PMC6991689 DOI: 10.1111/jcmm.14851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/27/2019] [Accepted: 11/02/2019] [Indexed: 02/05/2023] Open
Abstract
Gallbladder cancer (GBC) is the leading malignancy of biliary system showing refractory chemoresistance to current first-line drugs. Growing epidemiological evidences have established that the incidence of GBC exhibits significant gender predominance with females two-threefold higher than males, suggesting oestrogen/oestrogen receptors (ERs) signalling might be a critical driver of tumorigenesis in gallbladder. This study aims to evaluate the antitumour activity of tamoxifen (TAM), a major agent of hormonal therapy for breast cancer, in preclinical GBC model. Quantitative real-time PCR was used to investigate mRNA levels. Protein expression was measured by immunohistochemistry and Western blot. Glycolytic levels were measured by glucose consumption and lactic acid measurement. The antitumour activity of TAM alone or with cisplatin was examined with CCK8 assay, colony formation, flow cytometry and in vivo models. The results revealed that ERɑ expression was higher in GBC tissues and predicted poor clinical outcomes. TAM was showed effective against a variety of GBC cell lines. Mechanical investigations revealed that TAM enabled potent reactive oxygen species (ROS) production by reduced nuclear factor Nrf2 expression and its target genes, leading to the activation of AMPK, which subsequently induced impaired glycolysis and survival advantages. Notably, TAM was demonstrated to sensitize GBC cells to cisplatin (CDDP) both in vitro and in vivo. In agreement with these findings, elimination of oestrogens by ovariectomy in nude mice prevented CDDP resistance. In summary, these results provide basis for TAM treatment for GBC and shed novel light on the potential application of endocrine therapy for patients with GBC.
Collapse
Affiliation(s)
- Shuai Huang
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Hui Wang
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Wei Chen
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Ming Zhan
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Sunwang Xu
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xince Huang
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Ruirong Lin
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Hui Shen
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jian Wang
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
8
|
Dalvie D, Obach RS, Kang P, Prakash C, Loi CM, Hurst S, Nedderman A, Goulet L, Smith E, Bu HZ, Smith DA. Assessment of Three Human in Vitro Systems in the Generation of Major Human Excretory and Circulating Metabolites. Chem Res Toxicol 2009; 22:357-68. [DOI: 10.1021/tx8004357] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Deepak Dalvie
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - R. Scott Obach
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Ping Kang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Chandra Prakash
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Cho-Ming Loi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Susan Hurst
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Angus Nedderman
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Lance Goulet
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Evan Smith
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Hai-Zhi Bu
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| | - Dennis A. Smith
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego California 92121, Pfizer Global Research and Development, Groton Connecticut 06340, and Pfizer Global Research and Development, Sandwich, Kent, United Kingdom
| |
Collapse
|
9
|
Abstract
Recent clinical data on selective estrogen receptor modulators (SERMs) have provided the basis for reassessment of the SERM concept. The molecular basis of SERM activity involves binding of the ligand SERM to the estrogen receptor (ER), causing conformational changes which facilitate interactions with coactivator or corepressor proteins, and subsequently initiate or suppress transcription of target genes. SERM activity is intrinsic to each ER ligand, which accomplishes its unique profile by specific interactions in the target cell, leading to tissue selective actions. We discuss the estrogenic and anti-estrogenic effects of early SERMs, such as clomiphene citrate, used for treatment of ovulation induction, and the triphenylethylene, tamoxifen, which has ER antagonist activity in the breast, and is used for prevention and treatment of ER-positive breast cancer. Since the development of tamoxifen, other triphenylethylene SERMs have been studied for breast cancer prevention, including droloxifene, idoxifene, toremifene, and ospemifene. Other SERMs have entered clinical development more recently, including benzothiophenes (raloxifene and arzoxifene), benzopyrans (ormeloxifene, levormeloxifene, and EM-800), lasofoxifene, pipendoxifene, bazedoxifene, HMR-3339, and fulvestrant, an anti-estrogen which is approved for breast cancer treatment. SERMs have effects on tissues containing ER, such as the breast, bone, uterine and genitourinary tissues, and brain, and on markers of cardiovascular risk. Current evidence indicates that each SERM has a unique array of clinical activities. Differences in the patterns of action of SERMs suggest that each clinical end point must be evaluated individually, and conclusions about any particular SERM can only be established through appropriate clinical trials.
Collapse
|
10
|
Parte P, Kupfer D. Oxidation of tamoxifen by human flavin-containing monooxygenase (FMO) 1 and FMO3 to tamoxifen-N-oxide and its novel reduction back to tamoxifen by human cytochromes P450 and hemoglobin. Drug Metab Dispos 2005; 33:1446-52. [PMID: 15987777 DOI: 10.1124/dmd.104.000802] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tamoxifen (TAM), used as the endocrine therapy of choice for breast cancer, undergoes metabolism primarily forming N-desmethyltamoxifen, 4-hydroxytamoxifen, alpha-hydroxytamoxifen, and tamoxifen-N-oxide (TNO). Our earlier studies demonstrated that flavin-containing monooxygenases (FMOs) catalyze the formation of TNO. The current study demonstrates that human FMO1 and FMO3 catalyze TAM N-oxidation to TNO and that cytochromes P450 (P450s), but not FMOs, reduce TNO to TAM. CYP1A1, CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 all reduced TNO, with CYP2A6, CYP1A1, and CYP3A4 producing the greatest reduction. A portion of TAM formed by CYP3A4-mediated reduction of TNO was further metabolized, but not TAM formed by the other P450s. TNO reduction by P450s is extremely rapid with considerable TAM formation detected at the earliest time point that products could be measured. TAM formation exhibited a lack of linearity with incubation time but increased linearly as a function of TNO and P450 concentration. TNO was converted into TAM by reduced hemoglobin (Hb) and NADPH-P450 oxidoreductase, suggesting involvement of the same heme-Fe(2+) complex in both Hb and P450s. The findings raise the question of whether the reductive activity may be nonenzymatic. Results of this in vitro study demonstrate the potential of TAM and TNO to be interconverted metabolically. FMO seems to be the major enzymatic oxidant, whereas several P450 enzymes and even reduced hemoglobin are capable of reducing TNO back to TAM. The possibility that these processes may comprise a metabolic cycle in vivo is discussed in this article.
Collapse
Affiliation(s)
- Priyanka Parte
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| | | |
Collapse
|
11
|
Abstract
Despite the established impact of highly active antiretroviral therapy (HAART) in reducing HIV-related morbidity and mortality, malignancy remains an important cause of death. Patients who receive the combination of cancer chemotherapy and HAART may achieve better response rates and higher rates of survival than patients who receive antineoplastic therapy alone. However, the likelihood of drug interactions with combined therapy is high, since protease inhibitors (PIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) are substrates and potent inhibitors or inducers of the cytochrome P450 (CYP) system. Since many antineoplastic drugs are also metabolised by the CYP system, coadministration with HAART could result in either drug accumulation and possible toxicity, or decreased efficacy of one or both classes of drugs. Although formal, prospective pharmacokinetic interaction studies are not available in most instances, it is possible to infer the nature of drug interactions based on the metabolic fates of these agents. Paclitaxel and docetaxel are both metabolised by the CYP system, although differences exist in the nature of the isoenzymes involved. Case reports describing adverse consequences of concomitant taxane-antiretroviral therapy exist. Although other confounding factors may have been present, these cases serve as reminders of the vigilant monitoring necessary when taxanes and HAART are coadministered. Similarly, vinca alkaloids are substrates of CYP3A4 and are, thus, vulnerable to PI- or NNRTI-mediated changes in their pharmacokinetics. Interactions with the alkylating agents cyclophosphamide and ifosfamide are complicated as a result of the involvement of the CYP3A4 and CYP2B6 isoenzymes in both the metabolic activation of these drugs and the generation of potentially neurotoxic metabolites. Existing data regarding the metabolic fate of the anthracyclines doxorubicin and daunorubicin suggest that clinically detrimental interactions would not be expected with coadministered HAART. Commonly used endocrine therapies are largely substrates of the CYP system and may, therefore, be amenable to modulation by concomitant HAART. In addition, tamoxifen itself has been associated with reduced concentrations of both anastrozole and letrozole, raising the concern that similar inducing properties may adversely affect the outcome of PI- or NNRTI-based therapy. Similarly, dexamethasone is both a substrate and concentration-dependent inducer of CYP3A4; enhanced corticosteroid pharmacodynamics may result with CYP3A4 inhibitors, while the efficacy of concomitant HAART may be compromised with prolonged dexamethasone coadministration. Since PIs and NNRTIs may also induce or inhibit the expression of P-glycoprotein, the potential for additional interactions to arise via modulation of this transporter also exists. Further research delineating the combined safety and pharmacokinetics of antiretrovirals and antineoplastic therapy is necessary.
Collapse
|