Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Clin Rheumatol ; 37(2): 505-514, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28389989

ABSTRACT

A recent study in rheumatoid arthritis (RA) patients using electrical vagus nerve stimulation (VNS) to activate the inflammatory reflex has shown promising effects on disease activity. Innervation by the autonomic nerve system might be involved in the regulation of many endocrine and metabolic processes and could therefore theoretically lead to unwanted side effects. Possible effects of VNS on secretion of hormones are currently unknown. Therefore, we evaluated the effects of a single VNS on plasma levels of pituitary hormones and parameters of postprandial metabolism. Six female patients with RA were studied twice in balanced assignment (crossover design) to either VNS or no stimulation. The patients selected for this substudy had been on VNS therapy daily for at least 3 months and at maximum of 24 months. We compared 10-, 20-, and 30-min poststimulus levels to baseline levels, and a 4-h mixed meal test was performed 30 min after VNS. We also determined energy expenditure (EE) by indirect calorimetry before and after VNS. VNS did not affect pituitary hormones (growth hormone, thyroid stimulating hormone, adrenocorticotropic hormone, prolactin, follicle-stimulating hormone, and luteinizing hormone), postprandial metabolism, or EE. Of note, VNS reduced early postprandial insulin secretion, but not AUC of postprandial plasma insulin levels. Cortisol and catecholamine levels in serum did not change significantly. Short stimulation of vagal activity by VNS reduces early postprandial insulin secretion, but not other hormone levels and postprandial response. This suggests VNS as a safe treatment for RA patients.


Subject(s)
Arthritis, Rheumatoid/metabolism , C-Peptide/blood , Energy Metabolism/physiology , Postprandial Period/physiology , Vagus Nerve Stimulation , Adrenocorticotropic Hormone/blood , Adult , Calorimetry, Indirect , Cross-Over Studies , Female , Follicle Stimulating Hormone/blood , Human Growth Hormone/blood , Humans , Luteinizing Hormone/blood , Middle Aged , Prolactin/blood , Thyrotropin/blood
2.
J Intern Med ; 282(1): 64-75, 2017 07.
Article in English | MEDLINE | ID: mdl-28547815

ABSTRACT

Imbalance in the autonomic nervous system (ANS) has been observed in many established chronic autoimmune diseases, including rheumatoid arthritis (RA), which is a prototypic immune-mediated inflammatory disease (IMID). We recently discovered that autonomic dysfunction precedes and predicts arthritis development in subjects at risk of developing seropositive RA. In addition, RA patients with relatively high vagus nerve tone (higher parasympathetic parameters, measured by heart rate variability) respond better to antirheumatic therapies. Together, these data suggest that the ANS may control inflammation in humans. This notion is supported by experimental studies in animal models of RA. We have found that stimulation of the so-called cholinergic anti-inflammatory pathway by efferent electrical vagus nerve stimulation (VNS) or pharmacological activation of the alpha7 subunit of nicotinic acetylcholine receptors (α7nAChR) improves clinical signs and symptoms of arthritis, reduces cytokine production and protects against progressive joint destruction. Conversely, increased arthritis activity was observed in alpha7nAChR knockout mice. These studies together with previous work in animal models of sepsis and other forms of inflammation provided the rationale for an experimental clinical trial in patients with RA. We could for the first time show that an implantable vagus nerve stimulator inhibits peripheral blood cytokine production in humans. VNS significantly inhibited TNF and IL-6 production and improved RA disease severity, even in some patients with therapy-resistant disease. This work strongly supports further studies using a bioelectronic approach to treat RA and other IMIDs.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Arthritis, Rheumatoid/therapy , Autonomic Nervous System/physiopathology , Vagus Nerve Stimulation , Animals , Humans
3.
EBioMedicine ; 6: 231-237, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27211565

ABSTRACT

BACKGROUND: Heart rate variability (HRV) is a validated method to establish autonomic nervous system (ANS) activity. Rheumatoid arthritis (RA) is accompanied by ANS imbalance. We hypothesized that ANS dysfunction may precede the development of RA, which would suggest that it plays a role in its etiopathogenesis. METHODS: First, we assessed HRV parameters in supine (resting) and upright (active) position in healthy subjects (HS, n=20), individuals at risk of developing arthritis (AR subjects, n=50) and RA patients (RA, n=20). Next, we measured resting heart rate (RHR), a parasympathetic HRV parameter, in an independent prospective cohort of AR subjects (n=45). We also evaluated expression levels of the parasympathetic nicotinic acetylcholine receptor type 7 (α7nAChR) on circulating monocytes. FINDINGS: Both AR subjects (68 beats per minute (bpm), interquartile range (IQR) 68-73) and RA patients (68bpm, IQR 62-76) had a significantly higher RHR compared to HS (60bpm, IQR 56-63). RHR was significantly higher at baseline in individuals who subsequently developed arthritis. Expression levels of α7nAChR were lower in AR subjects with RHR ≥70bpm compared to those with RHR <70bpm, consistent with reduced activity of the parasympathetic cholinergic anti-inflammatory pathway. INTERPRETATION: These data support the notion that autonomic dysfunction precedes the development of RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/physiopathology , Autonomic Nervous System Diseases/diagnosis , alpha7 Nicotinic Acetylcholine Receptor/blood , Adult , Autonomic Nervous System Diseases/metabolism , Autonomic Nervous System Diseases/physiopathology , Female , Heart Rate , Humans , Male , Middle Aged , Prospective Studies
4.
Appl Environ Microbiol ; 81(18): 6253-67, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26150454

ABSTRACT

Lager brewing strains of Saccharomyces pastorianus are natural interspecific hybrids originating from the spontaneous hybridization of Saccharomyces cerevisiae and Saccharomyces eubayanus. Over the past 500 years, S. pastorianus has been domesticated to become one of the most important industrial microorganisms. Production of lager-type beers requires a set of essential phenotypes, including the ability to ferment maltose and maltotriose at low temperature, the production of flavors and aromas, and the ability to flocculate. Understanding of the molecular basis of complex brewing-related phenotypic traits is a prerequisite for rational strain improvement. While genome sequences have been reported, the variability and dynamics of S. pastorianus genomes have not been investigated in detail. Here, using deep sequencing and chromosome copy number analysis, we showed that S. pastorianus strain CBS1483 exhibited extensive aneuploidy. This was confirmed by quantitative PCR and by flow cytometry. As a direct consequence of this aneuploidy, a massive number of sequence variants was identified, leading to at least 1,800 additional protein variants in S. pastorianus CBS1483. Analysis of eight additional S. pastorianus strains revealed that the previously defined group I strains showed comparable karyotypes, while group II strains showed large interstrain karyotypic variability. Comparison of three strains with nearly identical genome sequences revealed substantial chromosome copy number variation, which may contribute to strain-specific phenotypic traits. The observed variability of lager yeast genomes demonstrates that systematic linking of genotype to phenotype requires a three-dimensional genome analysis encompassing physical chromosomal structures, the copy number of individual chromosomes or chromosomal regions, and the allelic variation of copies of individual genes.


Subject(s)
Adaptation, Biological , Aneuploidy , Beer/microbiology , Chromosomes, Fungal , Genome, Fungal , Industrial Microbiology , Saccharomyces/genetics , Fermentation , Flow Cytometry , High-Throughput Nucleotide Sequencing , Karyotype , Real-Time Polymerase Chain Reaction
5.
Oral Dis ; 21(7): 858-65, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26096224

ABSTRACT

OBJECTIVE: Activation of the cholinergic anti-inflammatory pathway (CAP) has been shown to reduce inflammation in animal models, while abrogation of the pathway increases inflammation. We investigated whether modulation of CAP influences inflammation in the non-obese diabetic (NOD) mouse model for Sjögren's syndrome and type 1 diabetes. METHODS: The alpha-7 nicotinic acetylcholine receptor (α7nAChR) was stimulated with AR-R17779 or nicotine in NOD mice. In a second study, unilateral cervical vagotomy was performed. α7nAChR expression, focus scores, and salivary flow were evaluated in salivary glands (SG) and insulitis score in the pancreas. Cytokines were measured in serum and SG. RESULTS: α7nAChR was expressed on myoepithelial cells in SG. Monocyte chemotactic protein-1 levels were reduced in SG after AR-R17779 treatment and tumor necrosis factor production was increased in the SG of the vagotomy group compared to controls. Focus score and salivary flow were unaffected. NOD mice developed diabetes more rapidly after vagotomy, but at completion of the study there were no statistically significant differences in number of mice that developed diabetes or in insulitis scores. CONCLUSION: Intervention of the CAP in NOD mice leads to minimal changes in inflammatory cytokines, but did not affect overall inflammation and function of SG or development of diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Pancreatitis/metabolism , Salivary Glands/metabolism , Sjogren's Syndrome/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Bridged-Ring Compounds/pharmacology , Chemokine CCL2/metabolism , Disease Models, Animal , Female , Inflammation , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Nicotine/pharmacology , Pancreatitis/pathology , Saliva/metabolism , Salivary Glands/drug effects , Salivation/drug effects , Spiro Compounds/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Vagotomy , alpha7 Nicotinic Acetylcholine Receptor/drug effects
6.
Best Pract Res Clin Rheumatol ; 28(4): 625-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25481554

ABSTRACT

There has been a marked improvement in the treatment of rheumatoid arthritis (RA), but most patients do not achieve disease remission. Therefore, there is still a need for new treatments. By screening an adenoviral short hairpin RNA library, we discovered that knockdown of the nicotinic acetylcholine receptor type 7 (α7nAChR) in RA fibroblast-like synoviocytes results in an increased production of mediators of inflammation and degradation. The α7nAChR is intimately involved in the cholinergic anti-inflammatory pathway (CAP). This led us to study the effects of α7nAChR activation in an animal model of RA, and we could show that this resulted in reduced arthritis activity. Accordingly, stimulation of the CAP by vagus nerve stimulation improved experimental arthritis. Conversely, we found aggravation of arthritis activity after unilateral cervical vagotomy as well as in α7nAChR-knockout mice. Together, these data provided the basis for exploration of vagus nerve stimulation in RA patients as a novel anti-inflammatory approach.


Subject(s)
Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/therapy , Inflammation/metabolism , Vagus Nerve Stimulation/methods , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Humans
7.
Br J Cancer ; 85(10): 1472-7, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11720431

ABSTRACT

The purpose of this study was to investigate the effect of the co-solvents Cremophor EL and polysorbate 80 on the absorption of orally administered paclitaxel. 6 patients received in a randomized setting, one week apart oral paclitaxel 60 mg m(-2) dissolved in polysorbate 80 or Cremophor EL. For 3 patients the amount of Cremophor EL was 5 ml m(-2), for the other three 15 ml m(-2). Prior to paclitaxel administration patients received 15 mg kg(-1) oral cyclosporin A to enhance the oral absorption of the drug. Paclitaxel formulated in polysorbate 80 resulted in a significant increase in the maximal concentration (C(max)) and area under the concentration-time curve (AUC) of paclitaxel in comparison with the Cremophor EL formulations (P = 0.046 for both parameters). When formulated in Cremophor EL 15 ml m(-2), paclitaxel C(max) and AUC values were 0.10 +/- 0.06 microM and 1.29 +/- 0.99 microM h(-1), respectively, whereas these values were 0.31 +/- 0.06 microM and 2.61 +/- 1.54 microM h(-1), respectively, when formulated in polysorbate 80. Faecal data revealed a decrease in excretion of unchanged paclitaxel for the polysorbate 80 formulation compared to the Cremophor EL formulations. The amount of paclitaxel excreted in faeces was significantly correlated with the amount of Cremophor EL excreted in faeces (P = 0.019). When formulated in Cremophor EL 15 ml m(-2), paclitaxel excretion in faeces was 38.8 +/- 13.0% of the administered dose, whereas this value was 18.3 +/-15.5% for the polysorbate 80 formulation. The results show that the co-solvent Cremophor EL is an important factor limiting the absorption of orally administered paclitaxel from the intestinal lumen. They highlight the need for designing a better drug formulation in order to increase the usefulness of the oral route of paclitaxel


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Glycerol/analogs & derivatives , Glycerol/pharmacology , Intestinal Absorption/drug effects , Neoplasms/metabolism , Paclitaxel/pharmacokinetics , Administration, Oral , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Area Under Curve , Cyclosporine/administration & dosage , Cyclosporine/pharmacokinetics , Cyclosporine/pharmacology , Drug Combinations , Feces/chemistry , Humans , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Pharmaceutical Vehicles , Polysorbates/pharmacology , Solvents/pharmacology
8.
Cancer Chemother Pharmacol ; 47(4): 347-54, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11345652

ABSTRACT

PURPOSE: To investigate dose escalation of bi-daily (b.i.d.) oral paclitaxel in combination with cyclosporin A in order to improve and prolong the systemic exposure to paclitaxel and to explore the maximum tolerated dose and dose limiting toxicity (DLT) of this combination. PATIENTS AND METHODS: A total of 15 patients received during course 1 two doses of oral paclitaxel (2 x 60, 2 x 90, 2 x 120, or 2 x 160 mg/m2) 7 h apart in combination with 15 mg/kg of cyclosporin A, co-administered to enhance the absorption of paclitaxel. During subsequent courses, patients received 3-weekly intravenous paclitaxel at a dose of 175 mg/m2 as a 3-h infusion. RESULTS: Toxicities observed following b.i.d. dosing of oral paclitaxel were generally mild and included toxicities common to paclitaxel administration and mild gastrointestinal toxicities such as nausea, vomiting, and diarrhea, which occurred more often at the higher dose levels. Dose escalation of b.i.d. oral paclitaxel from 2 x 60 to 2 x 160 mg/m2 did not result in a significant increase in the area under the plasma concentration-time curve (AUC) of paclitaxel. The AUC after doses of 2 x 60, 90, 120, and 160 mg/m2 were 3.77 +/- 2.70, 4.57 +/- 2.43, 3.62 +/- 1.58, and 8.58 +/- 7.87 microM.h, respectively. The AUC achieved after intravenous administration of paclitaxel 175 mg/m2 was 17.95 +/- 3.94 microM.h. CONCLUSION: Dose increment of paclitaxel did not result in a significant additional increase in the AUC values of the drug. Dose escalation of the b.i.d. dosing regimen was therefore not continued up to DLT. As b.i.d. dosing appeared to result in higher AUC values compared with single-dose administration (data which we have published previously), we recommend b.i.d. dosing of oral paclitaxel for future studies. Although pharmacokinetic data are difficult to interpret, due to the limited number of patients at each dose level and the large interpatient variability, we recommend the dose level of 2 x 90 mg/m2 for further investigation, as this dose level showed the highest systemic exposure to paclitaxel combined with good safety.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cyclosporine/administration & dosage , Cyclosporine/pharmacokinetics , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Administration, Oral , Adult , Aged , Antineoplastic Agents, Phytogenic/adverse effects , Area Under Curve , Cyclosporine/adverse effects , Drug Combinations , Female , Humans , Immunosuppressive Agents/adverse effects , Injections, Intravenous , Male , Middle Aged , Neoplasms/metabolism , Paclitaxel/adverse effects
9.
Anticancer Drugs ; 12(4): 315-23, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11335787

ABSTRACT

Intravenous administration of paclitaxel is hindered by poor water solubility of the drug. Currently, paclitaxel is dissolved in a mixture of ethanol and Cremophor EL; however, this formulation (Taxol) is associated with significant side effects, which are considered to be related to the pharmaceutical vehicle. A new polymer-conjugated derivative of paclitaxel, PNU166945, was investigated in a dose-finding phase I study to document toxicity and pharmacokinetics. A clinical phase I study was initiated in patients with refractory solid tumors. PNU16645 was administered as a 1-h infusion every 3 weeks at a starting dose of 80 mg/m(2), as paclitaxel equivalents. Pharmacokinetics of polymer-bound and released paclitaxel were determined during the first course. Twelve patients in total were enrolled in the study. The highest dose level was 196 mg/m(2), at which we did not observe any dose-limiting toxicities. Hematologic toxicity of PNU166945 was mild and dose independent. One patient developed a grade 3 neurotoxicity. A partial response was observed in one patient with advanced breast cancer. PNU166945 displayed a linear pharmacokinetic behavior for the bound fraction as well as for released paclitaxel. The study was discontinued prematurely due to severe neurotoxicity observed in additional rat studies. The presented phase I study with PNU166945, a water-soluble polymeric drug conjugate of paclitaxel, shows an alteration in pharmacokinetic behavior when paclitaxel is administered as a polymer-bound drug. Consequently, the safety profile may differ significantly from standard paclitaxel.


Subject(s)
Anemia/chemically induced , Nervous System Diseases/chemically induced , Paclitaxel/pharmacokinetics , Paclitaxel/toxicity , Polymers/pharmacokinetics , Polymers/toxicity , Adult , Aged , Area Under Curve , Breast Neoplasms/drug therapy , Carcinoma, Small Cell/drug therapy , Colonic Neoplasms/drug therapy , Dose-Response Relationship, Drug , Drug Carriers , Female , Gastrointestinal Diseases/chemically induced , Humans , Infusions, Intravenous , Lung Neoplasms/drug therapy , Male , Maximum Tolerated Dose , Methacrylates/chemistry , Middle Aged , Ovarian Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/chemical synthesis , Paclitaxel/chemistry , Polymers/chemical synthesis , Polymers/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/toxicity , Remission Induction , Skin Neoplasms/secondary , Solubility , Taxoids/analogs & derivatives
10.
Anticancer Drugs ; 12(4): 351-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11335792

ABSTRACT

The objective of this study was to define the minimally effective dose of cyclosporin A (CsA) that would result in a maximal increase of the systemic exposure to oral paclitaxel. Six evaluable patients participated in this randomized cross-over study in which they received at two occasions two doses of 90 mg/m(2) oral paclitaxel 7 h apart in combination with 10 or 5 mg/kg CsA. Dose reduction of CsA from 10 to 5 mg/kg resulted in a statistically significant decrease in the area under the plasma concentration-time curve (AUC) and time above the threshold concentrations of 0.1 microM (T>0.1 microM) of oral paclitaxel. The mean (+/-SD) AUC and T>0.1 microM values of oral paclitaxel with CsA 10 mg/kg were 4.29+/-0.88 microM x h and 12.0+/-2.1 h, respectively. With CsA 5 mg/kg these values were 2.75+/-0.63 microM x h and 7.0+/-2.1 h, respectively (p=0.028 for both parameters). In conclusion, dose reduction of CsA from 10 to 5 mg/kg resulted in a significant decrease in the AUC and T>0.1 microM values of oral paclitaxel. Because CsA 10 mg/kg resulted in similar paclitaxel AUC and T>0.1 microM values compared to CsA 15 mg/kg (data which we have published previously), the minimally effective dose of CsA is determined at 10 mg/kg.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cyclosporine/administration & dosage , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Adenocarcinoma/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/toxicity , Area Under Curve , Breast Neoplasms/drug therapy , Carcinoma, Small Cell/drug therapy , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Gastrointestinal Diseases/chemically induced , Hematologic Diseases/chemically induced , Humans , Infusions, Intravenous , Lung Neoplasms/drug therapy , Male , Middle Aged , Neoplasms/pathology , Premedication , Stomach Neoplasms/drug therapy , Uterine Cervical Neoplasms/drug therapy , Uterine Neoplasms/drug therapy
11.
J Clin Oncol ; 19(4): 1160-6, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11181682

ABSTRACT

PURPOSE: Oral bioavailability of docetaxel is very low, which is, at least in part, due to its affinity for the intestinal drug efflux pump P-glycoprotein (P-gp). In addition, metabolism of docetaxel by cytochrome P450 (CYP) 3A4 in gut and liver may also contribute. The purpose of this study was to enhance the systemic exposure to oral docetaxel on coadministration of cyclosporine (CsA), an efficacious inhibitor of P-gp and substrate for CYP 3A4. PATIENTS AND METHODS: A proof-of-concept study was carried out in 14 patients with solid tumors. Patients received one course of oral docetaxel 75 mg/m(2) with or without a single oral dose of CsA 15 mg/kg. CsA preceded oral docetaxel by 30 minutes. During subsequent courses, patients received intravenous (IV) docetaxel 100 mg/m(2). RESULTS: The mean (+/- SD) area under the concentration-time curve (AUC) in patients who received oral docetaxel 75 mg/m(2) without CsA was 0.37 +/- 0.33 mg.h/L and 2.71 +/- 1.81 mg.h/L for the same oral docetaxel dose with CsA. The mean AUC of IV docetaxel 100 mg/m(2) was 4.41 +/- 2.10 mg.h/L. The absolute bioavailability of oral docetaxel was 8% +/- 6% without and 90% +/- 44% with CsA. The oral combination of docetaxel and CsA was well tolerated. CONCLUSION: Coadministration of oral CsA strongly enhanced the oral bioavailability of docetaxel. Interpatient variability in the systemic exposure after oral drug administration was of the same order as after IV administration. These data are promising and form the basis for the further development of a clinically useful oral formulation of docetaxel.


Subject(s)
Cyclosporine/administration & dosage , Paclitaxel/analogs & derivatives , Paclitaxel/metabolism , Taxoids , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Availability , Docetaxel , Female , Humans , Male , Middle Aged , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Paclitaxel/adverse effects
12.
Br J Cancer ; 84(1): 42-7, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11139311

ABSTRACT

Oral bioavailability of paclitaxel is very low, which is due to efficient transport of the drug by the intestinal drug efflux pump P-glycoprotein (P-gp). We have recently demonstrated that the oral bioavailability of paclitaxel can be increased at least 7-fold by co-administration of the P-gp blocker cyclosporin A (CsA). Now we tested the potent alternative orally applicable non-immunosuppressive P-gp blocker GF120918. Six patients received one course of oral paclitaxel of 120 mg/m(2)in combination with 1000 mg oral GF120918 (GG918, GW0918). Patients received intravenous (i.v.) paclitaxel 175 mg/m(2)as a 3-hour infusion during subsequent courses. The mean area under the plasma concentration-time curve (AUC) of paclitaxel after oral drug administration in combination with GF120918 was 3.27 +/- 1.67 microM x h. In our previously performed study of 120 mg/m(2)oral paclitaxel in combination with CsA the mean AUC of paclitaxel was 2.55 +/- 2.29 microM x h. After i.v. administration of paclitaxel the mean AUC was 15.92( )+/- 2.46 microM x h. The oral combination of paclitaxel with GF120918 was well tolerated. The increase in systemic exposure to paclitaxel in combination with GF120918 is of the same magnitude as in combination with CsA. GF120918 is a good and safe alternative for CsA and may enable chronic oral therapy with paclitaxel.


Subject(s)
Acridines/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Breast Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Isoquinolines/administration & dosage , Lung Neoplasms/metabolism , Neoplasms, Unknown Primary/metabolism , Paclitaxel/pharmacokinetics , Tetrahydroisoquinolines , Administration, Oral , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Area Under Curve , Biological Availability , Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Female , Humans , Infusions, Intravenous , Lung Neoplasms/drug therapy , Male , Middle Aged , Neoplasms, Unknown Primary/drug therapy , Paclitaxel/administration & dosage , Paclitaxel/adverse effects
13.
J Clin Oncol ; 18(12): 2468-75, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10856107

ABSTRACT

PURPOSE: To investigate dose escalation of oral paclitaxel in combination with dose increment and scheduling of cyclosporine (CsA) to improve the systemic exposure to paclitaxel and to explore the maximum-tolerated dose (MTD) and dose-limiting toxicity (DLT). PATIENTS AND METHODS: A total of 53 patients received, on one occasion, oral paclitaxel in combination with CsA, coadministered to enhance the absorption of paclitaxel, and, on another occasion, intravenous paclitaxel at a dose of 175 mg/m(2) as a 3-hour infusion. RESULTS: The main toxicities observed after oral intake of paclitaxel were acute nausea and vomiting, which reached DLT at the dose level of 360 mg/m(2). Dose escalation of oral paclitaxel from 60 to 300 mg/m(2) resulted in significant but less than proportional increases in the plasma area under the concentration-time curve (AUC) of paclitaxel. The mean AUC values +/- SD after 60, 180, and 300 mg/m(2) of oral paclitaxel were 1.65 +/- 0.93, 3.33 +/- 2.39, and 3.46 +/- 1.37 micromol/L.h, respectively. Dose increment and scheduling of CsA did not result in a further increase in the AUC of paclitaxel. The AUC of intravenous paclitaxel was 15.39 +/- 3.26 micromol/L.h. CONCLUSION: The MTD of oral paclitaxel was 300 mg/m(2). However, because the pharmacokinetic data of oral paclitaxel, in particular at the highest doses applied, revealed nonlinear pharmacokinetics with only a moderate further increase of the AUC with doses up to 300 mg/m(2), the oral paclitaxel dose of 180 mg/m(2) in combination with 15 mg/kg oral CsA is considered most appropriate for further investigation. The safety of the oral combination at this dose level was good.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Cyclosporine/administration & dosage , Enzyme Inhibitors/administration & dosage , Paclitaxel/administration & dosage , Administration, Oral , Adult , Aged , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Area Under Curve , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Middle Aged , Neoplasms/drug therapy , Paclitaxel/adverse effects , Paclitaxel/pharmacokinetics
14.
Anticancer Drugs ; 11(10): 813-20, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11142688

ABSTRACT

The objective of this study was to compare the quantitative excretion of paclitaxel and metabolites after i.v. and oral drug administration. Four patients received 300 mg/m2 paclitaxel orally 30 min after 15 mg/kg oral cyclosporin A, co-administered to enhance the uptake of paclitaxel. Three weeks later these and three other patients received 175 mg/m2 paclitaxel by i.v. infusion. Blood samples, urine and feces were collected up to 48-96 h after administration, and analyzed for paclitaxel and metabolites. The area under the plasma concentration-time curve of paclitaxel after i.v. administration (175 mg/m2) was 16.2 +/- 1.7 microM x h and after oral administration (300 mg/m2) 3.8 +/- 1.5 microM x h. Following i.v. infusion of paclitaxel, total fecal excretion was 56 +/- 25%, with the metabolite 6alpha-hydroxypaclitaxel being the main excretory product (37 +/- 18%). After oral administration of paclitaxel, total fecal excretion was 76 +/- 21%, of which paclitaxel accounted for 61 +/- 14%. In conclusion, after i.v. administration of paclitaxel, excretion occurs mainly in the feces with the metabolites as the major excretory products. Orally administered paclitaxel is also mainly excreted in feces but with the parent drug in highest amounts. We assume that this high amount of parent drug is due to incomplete absorption of orally administered paclitaxel from the gastrointestinal tract.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/metabolism , Paclitaxel/pharmacokinetics , Administration, Oral , Adult , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biological Availability , Cyclosporine/administration & dosage , Drug Synergism , Enzyme Inhibitors/administration & dosage , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Paclitaxel/metabolism
15.
Clin Cancer Res ; 5(11): 3379-84, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589748

ABSTRACT

i.v. paclitaxel is inconvenient and associated with significant and poorly predictable side effects largely due to the pharmaceutical vehicle Cremophor EL. Oral administration may be attractive because it may circumvent the use of Cremophor EL. However, paclitaxel, as well as many other commonly applied drugs, has poor bioavailability caused by high affinity for the mdrl P-glycoprotein drug efflux pump, which is abundantly present in the gastrointestinal tract. Consequently, inhibition of P-glycoprotein by oral cyclosporin A (CsA) should increase systemic exposure of oral paclitaxel to therapeutic levels. A proof-of-concept study was carried out in 14 patients with solid tumors. Patients received one course of oral paclitaxel of 60 mg/m2 with or without 15 mg/kg CsA and with i.v. paclitaxel in subsequent courses. The pharmacokinetics of paclitaxel and its major metabolites were determined during the first two courses. In addition, levels of CsA, Cremophor EL, and ethanol were measured. Bioavailability of oral paclitaxel in combination with CsA was 8-fold higher than after oral paclitaxel alone (P<0.001). Therapeutic concentrations were achieved on average during 7.4 h, which is comparable with an equivalent i.v. dose. The oral combination was well tolerated and did not induce gastrointestinal toxicity or myelosuppression. Cremophor EL plasma levels after oral drug administration were undetectable. In conclusion, coadministration of oral CsA increased the systemic exposure of oral paclitaxel from negligible to therapeutic levels. The combination enables treatment with oral paclitaxel. Undetectable Cremophor EL levels after oral administration may have a very beneficial influence on the safety of the treatment with oral paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Cyclosporine/therapeutic use , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Administration, Oral , Adult , Aged , Antineoplastic Agents, Phytogenic/pharmacokinetics , Area Under Curve , Biological Availability , Biotransformation , Female , Humans , Male , Middle Aged , Paclitaxel/pharmacokinetics
16.
Pharmacol Res ; 40(1): 67-74, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10378993

ABSTRACT

AIM: To investigate the pharmacokinetics of paclitaxel (Paxene) administered to patients with advanced breast or ovarian cancer and to document safety and anti-tumour activity in this study population. PATIENTS AND METHODS: Patients with advanced breast or ovarian cancer were accrued to two clinical studies. Paclitaxel (Paxene) was administered as a 3-h 175 mg m-2 or as a 96-h 140 mg m-2(105 mg m-2 in the presence of liver metastases) infusion. Patients not responding to the 3-h schedule were permitted to cross-over to the 96-h schedule. The data were compared to those of five patients who were previously treated with paclitaxel administered as Taxol (140 mg m-296-h infusion) at our Institute. RESULTS: Fourteen patients with breast cancer and five ovarian cancer patients were entered into this study. Seven patients received the 3-h regimen, and 12 were assigned to the 96-h schedule. Five patients originally treated with the 3-h schedule, crossed over to the 96-h arm. For the 3-h 175 mg m-2 dose, the area under the plasma concentration vs time curve (AUC) was (mean+/-SD) 16.9+/-4.8 h x micromol x l-1, whereas the AUCs were 5.5+/-1.2 and 4.3+/-0.9 h x micromol x l-1 for the 96-h 140 mg m-2 and 105 mg m-2 doses, respectively. The clearance of paclitaxel was independent of the dose in the 96-h group, indicating linear pharmacokinetics. Pharmacokinetics of Paxene (96-h 140 mg m-2) were not significantly different from the kinetics after Taxol (96-h 140 mg m-2) administration.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Breast Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacokinetics , Adult , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Breast Neoplasms/metabolism , Female , Humans , Infusions, Intravenous , Middle Aged , Ovarian Neoplasms/metabolism , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Treatment Outcome
17.
Anticancer Drugs ; 9(8): 665-73, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9823424

ABSTRACT

Doxorubicin and paclitaxel both display strong antitumor activity in the treatment of breast cancer. The optimal schedule of this combination, however, remains undefined. In this phase I and pharmacologic study, we administered weekly 12 mg/m2 doxorubicin as a bolus infusion immediately followed by a 1 h 80 mg/m2 paclitaxel infusion to patients with metastatic breast cancer. A total of 119 weekly courses were delivered to seven patients. Grade IV neutropenia was observed in two patients at the first dose level, thus already defining the maximum tolerated dose. Pronounced non-hematologic toxicities were mild neuropathy (grade I: 39%) and stomatitis (grade I: 19%, grade II: 8%). No signs of cardiac toxicity were observed with this dose schedule. Three partial responses were achieved in this group of heavily pretreated patients. The pharmacokinetics of paclitaxel, doxorubicin and Cremophor EL with this schedule were analyzed. Overall, the schedule was well tolerated and combined with its preliminary response rate justifies further evaluation in phase II studies.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Adult , Alopecia/chemically induced , Alopecia/complications , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dose-Response Relationship, Drug , Doxorubicin/toxicity , Drug Administration Schedule , Female , Glycerol/analogs & derivatives , Glycerol/pharmacokinetics , Humans , Infusions, Intravenous , Middle Aged , Neutropenia/chemically induced , Neutropenia/complications , Paclitaxel/toxicity
18.
J Chromatogr B Biomed Sci Appl ; 696(1): 89-98, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9300913

ABSTRACT

A semi-automated reversed-phase high-performance liquid chromatography (HPLC) method has been developed and validated for the quantification of the novel anticancer drug docetaxel in human plasma. The chromatographic system also separated putative hydroxylated metabolites. A limited validation was performed for the assay of the metabolites while these reference compounds were not available in large quantities. The sample pretreatment of the plasma samples involves a solid-phase extraction (SPE) on Cyano end-capped columns. 2'-Methylpaclitaxel was used as internal standard. An APEX-octyl column (150 x 4.6 mm I.D.; particle size 5 microns) was used with acetonitrile-0.02 M ammonium acetate buffer pH 5 mixture as the mobile phase. UV detection was performed at 227 nm. In patient samples hydroxylated docetaxel metabolites were detected and quantified by using the docetaxel calibration curve. The accuracies and precisions of the assay fall within +/- 15% for all quality control samples and within +/- 20% for the lower limit of quantitation, which was 10 ng/ml using 1.00 ml of sample for both the parent drug and its metabolites. The overall recovery of the sample pretreatment procedure for docetaxel was 78.0% +/- 5.8% anc 84.8% +/- 3.1% for the internal standard 2'-methylpaclitaxel. Docetaxel was found to be stable in human plasma at -30 degrees C for at least 6 months. At ambient temperature docetaxel was stable for at most 15 h in human plasma.


Subject(s)
Antineoplastic Agents, Phytogenic/blood , Chromatography, High Pressure Liquid/methods , Paclitaxel/analogs & derivatives , Taxoids , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Circadian Rhythm , Docetaxel , Female , Humans , Hydroxylation , Linear Models , Methanol/chemistry , Middle Aged , Osmolar Concentration , Paclitaxel/administration & dosage , Paclitaxel/blood , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Reproducibility of Results , Spectrophotometry, Ultraviolet
19.
J Chromatogr B Biomed Appl ; 664(2): 373-82, 1995 Feb 17.
Article in English | MEDLINE | ID: mdl-7780590

ABSTRACT

A reversed-phase high-performance liquid chromatographic (RP-HPLC) method has been developed and validated for the quantitative determination of paclitaxel in human urine. A comparison is made between solid-phase extraction (SPE) and liquid-liquid extraction (LLE) as sample pretreatment. The HPLC system consists of an APEX octyl analytical column and acetonitrile-methanol-0.2 microM ammonium acetate buffer pH 5 (4:1:5, v/v) as the mobile phase. Detection is performed by UV absorbance measurement at 227 nm. The SPE procedure involves extraction on Cyano Bond Elut columns. n-Butylchloride is the organic extraction fluid used for the LLE. The recoveries of paclitaxel in human urine are 79 and 75% for SPE and LLE, respectively. The accuracy for the LLE and SPE sample pretreatment procedures is 100.4 and 104.9%, respectively, at a 5 micrograms/ml drug concentration. The lower limit of quantitation is 0.01 microgram/ml for SPE and 0.25 microgram/ml for LLE. Stability data of paclitaxel in human urine are also presented.


Subject(s)
Paclitaxel/urine , Calibration , Chromatography, High Pressure Liquid , Humans , Paclitaxel/chemistry , Quality Control , Spectrophotometry, Ultraviolet
20.
J Chromatogr ; 622(2): 235-42, 1993 Dec 22.
Article in English | MEDLINE | ID: mdl-8150871

ABSTRACT

A sensitive high-performance liquid chromatographic assay has been developed to determine the levels of 3'-amino-3'-deoxythymidine (AMT), a cytotoxic metabolite of 3'-azido-3'-deoxy-thymidine (AZT, zidovudine), in human plasma. The sample pretreatment involved solid-phase extraction using cation-exchange extraction columns. Chromatography was carried out on a C8 column, using a mobile phase of methanol-0.01 M ammonium acetate (pH 5)-0.25 M sodium dioctylsulfosuccinate (60:40:4, v/v/v) and ultraviolet detection at 265 nm. The method has been validated, and stability tests under various conditions have been performed. The lower limit of quantitation is 5 ng/ml (using 500-microliters human plasma samples). The bioanalytical assay has been used for the determination of AMT in patients with AIDS who used AZT.


Subject(s)
Dideoxynucleosides/blood , Zidovudine/blood , Acquired Immunodeficiency Syndrome/blood , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Humans , Spectrophotometry, Ultraviolet , Zidovudine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...