1
|
Hussain A, Naughton DP, Barker J. Potential Effects of Ibuprofen, Remdesivir and Omeprazole on Dexamethasone Metabolism in Control Sprague Dawley Male Rat Liver Microsomes (Drugs Often Used Together Alongside COVID-19 Treatment). MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072238. [PMID: 35408639 PMCID: PMC9000592 DOI: 10.3390/molecules27072238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 01/08/2023]
Abstract
The role of individual cytochrome P450 (CYPs) responsible for the drug metabolism can be determined through their chemical inhibition. During the pandemic, dexamethasone and remdesivir with omeprazole were used for the treatment of COVID-19, while Ibuprofen was taken to treat the symptoms of fever and headache. This study aimed to examine the potency of ibuprofen remdesivir, and omeprazole as inhibitors of cytochrome P450s using rat liver microsomes in vitro. Dexamethasone a corticosteroid, sometimes used to reduce the body’s immune response in the treatment of COVID-19, was used as a probe substrate and the three inhibitors were added to the incubation system at different concentrations and analysed by a validated High Performance Liquid Chromatography (HPLC) method. The CYP3A2 isoenzyme is responsible for dexamethasone metabolism in vitro. The results showed that ibuprofen acts as a non-competitive inhibitor for CYP3A2 activity with Ki = 224.981 ± 1.854 µM and IC50 = 230.552 ± 2.020 µM, although remdesivir showed a mixed inhibition pattern with a Ki = 22.504 ± 0.008 µM and IC50 = 45.007 ± 0.016 µM. Additionally, omeprazole uncompetitively inhibits dexamethasone metabolism by the CYP3A2 enzyme activity with a Ki = 39.175 ± 0.230 µM and IC50 = 78.351 ± 0.460 µM. These results suggest that the tested inhibitors would not exert a significant effect on the CYP3A2 isoenzyme responsible for the co-administered dexamethasone drug’s metabolism in vivo.
Collapse
|
2
|
Rampp C, Eichelkraut A, Best J, Czamara D, Rex-Haffner M, Uhr M, Binder EB, Menke A. Sex-related differential response to dexamethasone in endocrine and immune measures in depressed in-patients and healthy controls. J Psychiatr Res 2018; 98:107-115. [PMID: 29331929 DOI: 10.1016/j.jpsychires.2017.12.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/21/2017] [Accepted: 12/31/2017] [Indexed: 01/26/2023]
Abstract
Although sex differences in major depression have been reported repeatedly, the underlying mechanisms are still disputed. The rapidly changing gonadal steroid concentrations of the postpartum period or during menopause have been shown to be associated with depressive symptoms and to modulate the hypothalamic-pituitary-adrenal (HPA)-axis, which is implicated in depression. The sample comprised of 128 depressed in-patients (36.7% women) and 166 healthy controls (30.0% women). Blood was collected at baseline (at 6pm) and then 3 h as well as 21 h after ingestion of 1.5 mg dexamethasone for measurement of cortisol, ACTH and blood count. To further assess the function of the HPA-axis the dexamethasone/corticotrophin releasing hormone (Dex-CRH) test was performed in a subsample of 115 patients and 116 controls the following day. A significant interaction effect between sex, disease and ACTH concentrations over time after dexamethasone stimulation was observed, with men showing increased ACTH concentrations at baseline and after 21 h, while there was no difference after 3 h (p = .007). After separating for disease status this significant interaction effect was only observed in controls (p = .005). The cortisol response in the dex-CRH test was enhanced in female compared to male controls (p = .002). Leucocytes showed a stronger increase upon dexamethasone administration only in female compared to male controls (p = .023). These findings suggest a higher glucocorticoid receptor sensitivity following in-vivo glucocorticoid stimulation in healthy women that was absent in depressed patients. The sex-related differences in HPA-axis regulation and immune system function may contribute to the vulnerability of female sex to the development of depression.
Collapse
Affiliation(s)
- Carina Rampp
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, 80804, Germany
| | - Andreas Eichelkraut
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, 80804, Germany
| | - Johanna Best
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, 80804, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, 80804, Germany
| | - Monika Rex-Haffner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, 80804, Germany
| | - Manfred Uhr
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, 80804, Germany
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, Munich, 80804, Germany; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30307, USA
| | - Andreas Menke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Margarete-Hoeppel-Platz 1, Wuerzburg, 97080, Germany; Comprehensive Heart Failure Center, University Hospital of Wuerzburg, Am Schwarzenberg 15, Wuerzburg, 97080, Germany.
| |
Collapse
|
3
|
Allegra S, Fatiguso G, De Francia S, Favata F, Pirro E, Carcieri C, De Nicolò A, Cusato J, Di Perri G, D'Avolio A. Pharmacokinetic evaluation of oral itraconazole for antifungal prophylaxis in children. Clin Exp Pharmacol Physiol 2017; 44:1083-1088. [PMID: 28744925 DOI: 10.1111/1440-1681.12822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 01/02/2023]
Abstract
Itraconazole is a first-generation triazole agent with an extended spectrum of activity; it is licensed in adults for superficial and systemic fungal infections; no recommendation has been yet established for use in children patients. Its variable and unpredictable oral bioavailability make it difficult to determine the optimal dosing regimen. Hence, therapeutic drug monitoring, highly available in clinical practice, may improve itraconazole treatment success and safety. The aim of the study was to describe in paediatrics the oral itraconazole pharmacokinetics, used for prophylaxis. Moreover, we evaluated the utility of its therapeutic drug monitoring in this cohort. A fully validated chromatographic method was used to quantify itraconazole concentration in plasma collected from paediatric patients, at the end of dosing interval. Associations between variables were tested using the Pearson test. Mann-Whitney U test has been used to probe the influence of categorical variables on continuous ones. Any predictive power of the considered variables was finally evaluated through univariate and multivariate linear and logistic regression analyses. A high inter-individual variability was shown; ethnicity (beta coefficient, β -0.161 and interval of confidence at 95%, IC -395.035; -62.383) and gender (β 0.123 and IC 9.590; 349.395) remained in the final linear regression model with P value of .007 and .038, respectively. This study highlights that therapeutic drug monitoring is required to achieve an adequate target itraconazole serum exposure.
Collapse
Affiliation(s)
- Sarah Allegra
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| | - Giovanna Fatiguso
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| | - Silvia De Francia
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy
| | - Fabio Favata
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| | - Elisa Pirro
- Department of Biological and Clinical Sciences, University of Turin, S. Luigi Gonzaga Hospital, Orbassano (TO), Italy
| | - Chiara Carcieri
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| | - Amedeo De Nicolò
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| | - Jessica Cusato
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| | - Giovanni Di Perri
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| | - Antonio D'Avolio
- Laboratory of Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, Unit of Infectious Diseases, University of Torino, ASL Città di Torino, Amedeo di Savoia Hospital, Turin, Italy
| |
Collapse
|
4
|
Nieuweboer AJM, de Morrée ES, de Graan AJM, Sparreboom A, de Wit R, Mathijssen RHJ. Inter-patient variability in docetaxel pharmacokinetics: A review. Cancer Treat Rev 2015; 41:605-13. [PMID: 25980322 DOI: 10.1016/j.ctrv.2015.04.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 04/26/2015] [Accepted: 04/28/2015] [Indexed: 11/17/2022]
Abstract
Docetaxel is a frequently used chemotherapeutic agent in the treatment of solid cancers. Because of the large inter-individual variability (IIV) in the pharmacokinetics (PK) of docetaxel, it is challenging to determine the optimal dose in individual patients in order to achieve optimal efficacy and acceptable toxicity. Despite the established correlation between systemic docetaxel exposure and efficacy, the precise factors influencing docetaxel PK are not yet completely understood. This review article highlights currently known factors that influence docetaxel PK, and focusses on those that are clinically relevant. For example, liver impairment should be taken into account when calculating docetaxel dosages as this may decrease docetaxel clearance. In addition, drug-drug interactions may be of distinct clinical importance when using docetaxel. Particularly, drugs strongly inhibiting CYP3A4 such as ketoconazole should not be concurrently administered without dose modification, as they may decrease the clearance of docetaxel. Gender, castration status, and menopausal status might be of importance as potential factors influencing docetaxel PK. The role of pharmacogenetics in predicting docetaxel PK is still limited, since no polymorphisms of clinical importance have yet been established.
Collapse
Affiliation(s)
| | - Ellen S de Morrée
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Anne-Joy M de Graan
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Alex Sparreboom
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands; Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
5
|
Younis IR, George DJ, McManus TJ, Hurwitz H, Creel P, Armstrong AJ, Yu JJ, Bacon K, Hobbs G, Peer CJ, Petros WP. Clinical pharmacology of an atrasentan and docetaxel regimen in men with hormone-refractory prostate cancer. Cancer Chemother Pharmacol 2014; 73:991-7. [PMID: 24619498 DOI: 10.1007/s00280-014-2432-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 02/27/2014] [Indexed: 11/29/2022]
Abstract
PURPOSE This study was conducted to evaluate potential pharmacokinetic interactions between docetaxel and atrasentan as part of a phase I/II clinical trial. METHODS Patients with prostate cancer were treated with intravenous docetaxel (60-75 mg/m(2)) every 3 weeks and oral atrasentan (10 mg) daily starting on day 3 of cycle 1 and then given continuously. The pharmacokinetics of both drugs were evaluated individually (cycle 1, day 1 for docetaxel; day 21 for atrasentan) and in combination (cycle 2, day 1 for both drugs). Pharmacogenomics of alpha-1-acid glycoprotein (AAG) were also explored. RESULTS Paired pharmacokinetic data sets for both drugs were evaluable in 21 patients. Atrasentan was rapidly absorbed and plasma concentrations varied over a fourfold range at steady state within a typical patient. The median apparent oral clearance of atrasentan was 17.4 L/h in cycle 1 and was not affected by docetaxel administration (p = 0.9). Median systemic clearance of docetaxel was 51.1 L/h on the first cycle and significantly slower (p = 0.01) compared with that obtained during co-administration of atrasentan, 61.6 L/h. Docetaxel systemic clearance in cycle 1 was 70.0 L/h in patients homozygous for a variant allele in AAG compared with 44.5 L/h in those with at least one wild-type allele (p = 0.03). CONCLUSION Genetic polymorphism in AAG may explain some inter-patient variability in docetaxel pharmacokinetics. The systemic clearance of docetaxel is increased by approximately 21 % when given concomitantly with atrasentan; however, atrasentan pharmacokinetics does not appear to be influenced by docetaxel administration.
Collapse
Affiliation(s)
- Islam R Younis
- Mary Babb Randolph (MBR) Cancer Center, West Virginia University, PO Box 9300, Morgantown, WV, 26506, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Krens SD, McLeod HL, Hertz DL. Pharmacogenetics, enzyme probes and therapeutic drug monitoring as potential tools for individualizing taxane therapy. Pharmacogenomics 2013; 14:555-74. [PMID: 23556452 DOI: 10.2217/pgs.13.33] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The taxanes are a class of chemotherapeutic agents that are widely used in the treatment of various solid tumors. Although taxanes are highly effective in cancer treatment, their use is associated with serious complications attributable to large interindividual variability in pharmacokinetics and a narrow therapeutic window. Unpredictable toxicity occurrence necessitates close patient monitoring while on therapy and adverse effects frequently require decreasing, delaying or even discontinuing taxane treatment. Currently, taxane dosing is based primarily on body surface area, ignoring other factors that are known to dictate variability in pharmacokinetics or outcome. This article discusses three potential strategies for individualizing taxane treatment based on patient information that can be collected before or during care. The clinical implementation of pharmacogenetics, enzyme probes or therapeutic drug monitoring could enable clinicians to personalize taxane treatment to enhance efficacy and/or limit toxicity.
Collapse
Affiliation(s)
- Stefanie D Krens
- UNC Institute for Pharmacogenomics & Individualized Therapy, University of North Carolina at Chapel Hill, 120 Mason Farm Road, CB 7361, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
7
|
Abstract
There are clear gender-dependent differences in response rates and the probability of side effects in patients treated with chemotherapy. Sex-biased expression levels of metabolic enzymes and transporters in liver and kidney leading to different pharmacokinetics have been described for most common anti-cancer drugs. In women, half-life is often longer, which is associated with improved survival, but also increased toxicity.Some chemotherapy protocols lead to a better response rate in women without increasing toxicity (e.g., cisplatin and irinotecan), while others only increase toxicity, but do not improve response rates in women (e.g., 5-fluorouracil). The increased toxicity often correlates with different pharmacokinetics, but women also show a higher sensitivity to some agents with shorter half-life (e.g., steroids). Organ-specific toxicities like cardiac toxicity after doxorubicin treatment or neurotoxicity associated with ifosfamide are more severe in women due to gender-specific changes in gene expression. Novel therapies like tyrosine kinase inhibitors or monoclonal antibodies show very complex, but clinical significant differences depending on gender. Antibodies often have a longer half-life in women, which is associated with an improved response to therapy.Side effects appear to be highly dependent on different tissue properties, as women have a higher incidence of oral mucositis, but lower rates of gut toxicity. Nausea and vomiting is a greater problem in females during therapy due to the lower activity of anti-emetic drugs. Nausea and vomiting pose a bigger challenge in female patients, as anti-emetic drugs seem to be less effective.
Collapse
Affiliation(s)
- Oliver Schmetzer
- Max-Delbrück-Centrum for Molecular Medicine Molecular Immunotherapy, Berlin, Germany.
| | | |
Collapse
|
8
|
Engels FK, Loos WJ, van der Bol JM, de Bruijn P, Mathijssen RHJ, Verweij J, Mathot RAA. Therapeutic drug monitoring for the individualization of docetaxel dosing: a randomized pharmacokinetic study. Clin Cancer Res 2011; 17:353-62. [PMID: 21224369 DOI: 10.1158/1078-0432.ccr-10-1636] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Docetaxel pharmacokinetic (PK) parameters, notably clearance and exposure (AUC), are characterized by large interindividual variability. The purpose of this study was to evaluate the effect of PK-guided [area under the plasma concentration versus time curve (AUC) targeted], individualized docetaxel dosing on interindividual variability in exposure. EXPERIMENTAL DESIGN A limited sampling strategy in combination with a validated population PK model, Bayesian analysis, and a predefined target AUC was used. Fifteen patients were treated for at least 2 courses with body surface area-based docetaxel and 15 with at least 1 course of PK-guided docetaxel dosing. RESULTS Interindividual variability (SD of ln AUC) was decreased by 35% (N = 15) after 1 PK-guided course; when all courses were evaluated, variability was decreased by 39% (P = 0.055). PK-guided dosing also decreased the interindividual variability of percentage decrease in white blood cell and absolute neutrophil counts by approximately 50%. CONCLUSIONS Further research is required to determine whether the decrease in PK variability can contribute to a reduction in interindividual variability in efficacy and toxicity.
Collapse
Affiliation(s)
- Frederike K Engels
- Department of Medical Oncology, Erasmus MC-Daniel den Hoed Cancer Center, Rotterdam, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
The liver is fundamentally important in drug metabolism. In oncology, the astute clinician must not only understand the meaning and limitations of commonly ordered liver biochemical tests, but also be aware of which anticancer agents might induce liver dysfunction, and of the strategies for appropriate dosing of patients with pre-existing liver dysfunction. In part I of our Review, we highlighted both the importance and inadequacies of identifying serum biochemical liver abnormalities in oncology; we also discussed a lack of routine formal investigation of liver function. We summarised chemotherapy-related hepatotoxicity and other causes of liver toxic effects in patients with cancer. Here in part II, we discuss trials that have specifically assessed chemotherapy dosing strategies in the setting of overt biochemical liver dysfunction and we note their recommendations. Furthermore, we review other assessments of liver metabolic and excretory function, particularly in the setting of chemotherapy drug handling. We discuss the potential use of these metabolic probes in practice.
Collapse
Affiliation(s)
- Kathryn M Field
- Division of Haematology and Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | | |
Collapse
|
10
|
van Schaik RH. CYP450 pharmacogenetics for personalizing cancer therapy. Drug Resist Updat 2008; 11:77-98. [DOI: 10.1016/j.drup.2008.03.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Revised: 03/25/2008] [Accepted: 03/26/2008] [Indexed: 01/11/2023]
|
11
|
Puisset F, Alexandre J, Treluyer JM, Raoul V, Roché H, Goldwasser F, Chatelut E. Clinical pharmacodynamic factors in docetaxel toxicity. Br J Cancer 2007; 97:290-6. [PMID: 17595656 PMCID: PMC2360335 DOI: 10.1038/sj.bjc.6603872] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neutropenia is the main dose-limiting toxicity occurring in docetaxel treatment. The objective of this study was to identify pharmacodynamic (PD) factors responsible for the neutropaenia caused by docetaxel. Data were obtained from 92 patients treated with docetaxel as a monochemotherapy in two different treatment centres. A semiphysiological population pharmacokinetic–pharmacodynamic (PK/PD) model was applied to describe the time course of neutrophils and the neutropaenic effect of docetaxel. The plasma docetaxel concentration was assumed to inhibit the proliferation of neutrophil precursors through a linear model: Drug effect=Slope × Conc. Slope corresponds to the patients’ sensitivity to the neutropaenic effect of docetaxel. Covariate analysis was performed by testing the relationship between the patients’ characteristics and Slope using the program NONMEM. The neutropaenic effect of docetaxel showed a high interindividual variability. Three significant PD covariates were identified: serum α1-acid glycoprotein levels (AAG), level of chemotherapy pretreatment, and treatment centre. Extensive pretreatment was associated with an increase in Slope values meaning a higher haematotoxicity. An increase in AAG was associated with a decrease of both Slope and docetaxel plasma clearance. Patients treated in one centre had both higher Slope and docetaxel clearance. The centre effect (most likely due to a bias in the PK part of the study between the two centres) reveals the robustness of the PK/PD model. Individual dosing of docetaxel should be based on previous chemotherapy but not on the AAG level since it has a similar influence on PD and PK docetaxel parameters. This methodology should be applied to further investigate elderly patients and to identify more precisely the characteristics of previous chemotherapy that contribute to the cumulative myelotoxicity.
Collapse
Affiliation(s)
- F Puisset
- Université de Toulouse et Institut Claudius-Regaud, EA3035, F-31052 Toulouse, France
| | - J Alexandre
- Faculté de Médecine Université Paris Descartes, Paris, France
| | - J-M Treluyer
- Faculté de Médecine Université Paris Descartes, Paris, France
| | - V Raoul
- Université de Toulouse et Institut Claudius-Regaud, EA3035, F-31052 Toulouse, France
| | - H Roché
- Université de Toulouse et Institut Claudius-Regaud, EA3035, F-31052 Toulouse, France
| | - F Goldwasser
- Faculté de Médecine Université Paris Descartes, Paris, France
| | - E Chatelut
- Université de Toulouse et Institut Claudius-Regaud, EA3035, F-31052 Toulouse, France
- E-mail:
| |
Collapse
|