686 research outputs found

    Irinotecan: from clinical pharmacokinetics to pharmacogenetics

    Get PDF
    Four decades ago, a plant alkaloid was isolated from the Chinese tree Camptotheca acuminata (Nyssaceae family), which showed promising antitumor activity in vitro (1 ). At that moment, a relatively new class of anticancer agents was born. Unfortunately, during clinical trials, this alkaloid, called camptothecin (CPT), showed severe and unpredictable side effects (myelosuppression, diarrhea, and hemorrhagic cystitis), which were unmanageble during this early chemotherapeutic era (2-4). Later on, the drug's relative lack of hydrophility was found to be the main reason for these adverse events. Despite this ''false starf', renewed interest in this compound was found after the elucidation of its mechanism of action during the mid-1980s (i.e the inhibition of topoisomerase I; ref. 5-7). Topoisomerase I is an essential enzyme in DNA replication and RNA transcription and is present in all eukaryotic cells (8). In the physiologic state of the cell nucleus, DNA is double-stranded, super-coiled and fits tightly into a chromosome. To synthesize new DNA, this topological constrained DNA should unwind. To this end, topoisomerase I binds to the DNA, forms a covalent reversible adduct (which is called the cleavable complex), cleaves a phosphodiester bond and a single-strand break occurs (6). Next, the DNA molecule is able to rotate freely around its intact single strand, and relaxation of DNA takes place. After the religation of the cleavage, the enzyme dissociates from DNA. The cleavable complex usually binds for a short period, just to allow the single strand to unwind (9, 1 0). CPT induces the stabilization of the cleavable complexes (11, 12), what will lead to a disruption of the DNA-strand when the replication fork meets the cleavable complex (7). This doublestrand break will lead to the activation of apoptosis pathways and ultimately result in cell-death (7). In subsequent years, various derivatives of CPT with improved water solubility have been developed, of which irinotecan (CPT-11) was considered to be among the most promising ones (13). It is a semisynthetic water-soluble prodrug, with minor intrinsic cy1otoxic activity. After administration, the drug is partially converted into the 1 00- to 1 ,000-times more active metabolite SN-38 by the enzyme carboxyl esterase (14-17). In the liver, SN-38 is de-toxified by undine-diphosphate glucuronosyltransferase 1 A isoforms, whereby SN-38-glucuronide is formed (18). All these compounds can be secreted into the bile by specific transporters (19). In the intestines, several bacteria produce ~-glucuronidase, which enables re-activatation of the glucuronidated form of SN-38 (20). Relatively high concentrations of SN-38 will lead to damage of the bowel mucosa, which results in one of the drug's most prominent side effects: diarrhea (21). In addition, irinotecan can also be oxidated by cy1ochrome P450 isozymes (22, 23), which will lead to the formation of several inactive metabolites, including APC and NPC

    Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics

    Get PDF
    Since its clinical introduction in 1998, the topoisomerase I inhibitor irinotecan has been widely used in the treatment of solid tumors, including colorectal, pancreatic, and lung cancer. Irinotecan therapy is characterized by several dose-limiting toxicities and large interindividual pharmacokinetic variability. Irinotecan has a highly complex metabolism, including hydrolyzation by carboxylesterases to its active metabolite SN-38, which is 100- to 1000-fold more active compared with irinotecan itself. Several phase I and II enzymes, including cytochrome P450 (CYP) 3A4 and uridine diphosphate glucuronosyltransferase (UGT) 1A, are involved in the formation of inactive metabolites, making its metabolism prone to environmental and genetic influences. Genetic variants in the DNA of these enzymes and transporters coul

    Pharmacological and clinical aspects of immediate release fentanyl preparations: Criteria for selection

    Get PDF
    In palliative care, pain management is often hampered by episodes of breakthrough pain, characterised by a rapid onset and, on average, duration less than 1 h. Until recently, only immediate release morphine and oxycodon preparations were available for the treatment of these episodes but time until effect is too long for both of these drugs. Recently, immediate release fentanyl products have become available for the treatment of breakthrough pain. These products can be classified as oromucosal and nasal products. Both are absorbed rapidly by the mucosa, although the oromucosally delivered products are partly absorbed by the gastrointestinal tract and therefore reach maximum plasma levels somewhat slower (after 30-90 min) than the nasally delivered products (after ~15 min). In clinical placebo controlled studies, all new immediate release fentanyl products were proven to be effective from 15 min after administration in the treatment of breakthrough pain. The first studies comparing immediate release fentanyl with immediate release morphine or oxycodon also found superiority for the new fentanyl products

    Quantification of afatinib, alectinib, crizotinib and osimertinib in human plasma by liquid chromatography/triple-quadrupole mass spectrometry; focusing on the stability of osimertinib

    Get PDF
    The development and full validation of a sensitive and selective ultra-performance liquid chromatography/ tandem mass spectrometry (UPLC–MS/MS) method are described for the simultaneous analysis of afatinib, alectinib, crizotinib and osimertinib in human lithium heparinized plasma. Afatinib-d6, crizotinib-d5 and erlotinib-d6 were used as internal standards. Given osimertinib's instability in plasma and whole blood at ambient temperature, samples should be solely processed on ice (T = 0 °C). Chromatographic separation was obtained on an Acquity UPLC ® BEH C18; 2.1 × 50 mm, 1.7 μm column, which was eluted with 0.400 mL/minute flow on a linear gradient, consisting of 10 mM ammonium formate (pH 4.5) and acetonitrile. Calibration curves for all compounds were linear for concentration ranges of 1.00 to 100 ng/mL for afatinib and 10.0 to 1000 ng/mL for alectinib, crizotinib and osimertinib, herewith validating the lower limits of quantification at 1.00 ng/mL for afatinib and 10.0 ng/mL for alectinib, crizotinib and osimertinib. Within-run and between-run precision measurements fell within 10.2%, with accuracy ranging from 89.2 to 110%

    Review – The impact of pharmacogenetics on the outcome of immune checkpoint inhibitors

    Get PDF
    The development of immune checkpoint inhibitors (ICIs) has a tremendous effect on the treatment options for multiple types of cancer. Nonetheless, there is a large interpatient variability in response, survival, and the development of immune-related adverse events (irAEs). Pharmacogenetics is the general term for germline genetic variations, which may cause the observed interindividual differences in response or toxicity to treatment. These genetic variations can either be single-nucleotide polymorphisms (SNPs) or structural variants, such as gene deletions, amplifications or rearrangements. For ICIs, pharmacogenetic variation in the human leukocyte antigen molecules has also been studied with regard to treatment outcome. This review presents a summary of the literature regarding the pharmacogenetics of ICI treatment, discusses the most important known genetic variations and offers recommendations on the application of pharmacogenetics for ICI treatment.</p

    Inhibition of OATP1B1 by tyrosine kinase inhibitors: In vitro-in vivo correlations

    Get PDF
    Background:Several tyrosine kinase inhibitors (TKIs) can decrease docetaxel clearance in patients by an unknown mechanism. We hypothesised that these interactions are mediated by the hepatic uptake transporter OATP1B1.Methods:The influence of 16 approved TKIs on transport was studied in vitro using HEK293 cells expressing OATP1B1 or its mouse equivalent Oatp1b2. Pharmacokinetic studies were performed with Oatp1b2-knockout and OATP1B1-transgenic mice.Results:All docetaxel-interacting TKIs, including sorafenib, were identified as potent inhibitors of OATP1B1 in vitro. Although Oatp1b2 deficiency in vivo was associated with increased docetaxel exposure, single- or multiple-dose sorafenib did not influence docetaxel pharmacokinetics.Conclusion: These findings highlight the importance of identifying proper preclinical models for verifying and predicting TKI-chemotherapy interactions involving transporters

    Recent Clinical Developments of Nanomediated Drug Delivery Systems of Taxanes for the Treatment of Cancer

    Get PDF
    Conventional taxanes are used as cornerstone of the chemotherapeutical treatment for a variety of malignancies. Nevertheless, a large proportion of patients do not benefit from their treatment while they do suffer from severe adverse events related to the solvent or to the active compound. Cremophor EL and polysorbate 80 free formulations, conjugates, oral formulations and different types of drug delivery systems are some examples of the several attempts to improve the treatment with taxanes. In this review article, we discuss recent clinical developments of nanomediated drug delivery systems of taxanes for the treatment of cancer. Targeting mechanisms of drug delivery systems and characteristics of the most commonly used taxane-containing drug delivery systems in the clinical setting will be discussed in this review

    Effects of St. John's wort on irinotecan metabolism

    Get PDF
    St. John's wort (SJW), a widely used herbal product, has been implicated in drug interactions resulting from the induced expression of the cytochrome P450 CYP3A4 isoform. In this study, we determined the effect of SJW on the metabolism of irinotecan, a pro-drug of SN-38 and a known substrate for CYP3A4. Five cancer patients were treated with irinotecan (350 mg/m(2), intravenously) in the presence and absence of SJW (900 mg daily, orally for 18 days) in an unblinded, randomized crossover study design. The plasma levels of the active metabolite SN-38 decreased by 42% (95% confidence interval [CI] = 14% to 70%) following SJW cotreatment with 1.0 micro M x h (95% CI = 0.34 micro M x h to 1.7 micro M x h) versus 1.7 micro M x h (95% CI = 0.83 micro M x h to 2.6 micro M x h) (P =.033, two-sided paired Student's t test). Consequently, the degree of myelosuppression was substantially worse in the absence of SJW. These findings indicate that patients on irinotecan treatment should refrain from taking SJW because plasma levels of SN-38 were dramatically reduced, which may have a deleterious impact on treatment outcome

    Drug-drug interactions with tyrosine-kinase inhibitors:A clinical perspective

    Get PDF
    In the past decade, many tyrosine-kinase inhibitors have been introduced in oncology and haemato-oncology. Because this new class of drugs is extensively used, serious drug-drug interactions are an increasing risk. In this Review, we give a comprehensive overview of known or suspected drug-drug interactions between tyrosine-kinase inhibitors and other drugs. We discuss all haemato-oncological and oncological tyrosine-kinase inhibitors that had been approved by Aug 1, 2013, by the US Food and Drug Administration or the European Medicines Agency. Various clinically relevant drug interactions with tyrosine-kinase inhibitors have been identified. Most interactions concern altered bioavailability due to altered stomach pH, metabolism by cytochrome P450 isoenzymes, and prolongation of the QTc interval. To guarantee the safe use of tyrosine-kinase inhibitors, a drugs review for each patient is needed. This Review provides specific recommendations to guide haemato-oncologists, oncologists, and clinical pharmacists, through the process of managing drug-drug interactions during treatment with tyrosine-kinase inhibitors in daily clinical practice
    • …
    corecore