1
|
Application of physiologically based pharmacokinetic modeling to predict the pharmacokinetics of telavancin in obesity with renal impairment. Eur J Clin Pharmacol 2021; 77:989-998. [PMID: 33447912 PMCID: PMC7808764 DOI: 10.1007/s00228-020-03072-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 12/15/2020] [Indexed: 11/08/2022]
Abstract
Purpose U.S. Food and Drug Administration (FDA) recommended telavancin dosing is based on total body weight (TBW) but lacks adjusted regimens for obese subjects with varying renal function. Our aim was to develop a physiologically based pharmacokinetic (PBPK) model of telavancin to design optimized dosing regimens for obese patients with hospital-acquired pneumonia (HAP) and varying renal function. Methods The PBPK model was verified using clinical pharmacokinetic (PK) data of telavancin in healthy populations with varying renal function and obese populations with normal renal function. Then, the PBPK model was applied to predict the PK in obese HAP patients with renal impairment (RI). Results The fold error values of PK parameters (AUC, Cmax, Tmax) were all within 1.5. The telavancin AUC0-inf was predicted to increase 1.07-fold in mild RI, 1.23-fold in moderate RI, 1.41-fold in severe RI, and 1.57-fold in end-stage renal disease (ESRD), compared with that in obese HAP with normal renal function. The PBPK model combined with Monte Carlo simulations (MCS) suggested that dose adjustment based on a 750-mg-fixed dose could achieve effectiveness with reduced risk of toxicity, compared with current TBW-based dosing recommendations. Conclusion The PBPK simulation proposed that using TBW-based regimen in obesity with RI should be avoided. Dose recommendations in obesity from the PBPK model are 750 mg daily for normal renal function and mild RI, 610 mg daily for moderate RI, 530 mg daily for severe RI, and 480 mg daily for ESRD. Supplementary Information The online version contains supplementary material available at 10.1007/s00228-020-03072-y.
Collapse
|
2
|
Meini S, Zini C, Passaleva MT, Frullini A, Fusco F, Carpi R, Piani F. Pneumatosis intestinalis in COVID-19. BMJ Open Gastroenterol 2020; 7:bmjgast-2020-000434. [PMID: 32522754 PMCID: PMC7287500 DOI: 10.1136/bmjgast-2020-000434] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/19/2020] [Accepted: 05/29/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction COVID-19 is a respiratory illness due to novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), described in December 2019 in Wuhan (China) and rapidly evolved into a pandemic. Gastrointestinal (GI) tract can also be involved. Case presentation A 44-year-old man was hospitalised for COVID-19-associated pneumonia. A rapid recovery of respiratory and general symptoms was observed after 1 week of treatment with lopinavir/ritonavir plus hydroxychloroquine and broad-spectrum antibiotics (piperacillin–tazobactam plus teicoplanin). No GI symptoms were reported during hospitalisation, but a lung contrast-enhancement CT (CE-CT) excluding thromboembolism showed, as collateral finding, intraperitoneal free bubbles not present on a previous CT examination; the subsequent abdominal CE-CT described pneumatosis intestinalis (PI) involving the caecum and the right colon. Ciprofloxacin plus metronidazole was started, and the 2-week follow-up CT showed the complete resolution of PI. Discussion The pathogenesis of PI is poorly understood. PI involving the caecum and right colon has been described for HIV and Cytomegalovirus infections, but, to our best knowledge, never before in COVID-19. We hypothesise a multifactorial aetiopathogenesis for PI, with a possible role of the bowel wall damage and microbiota impairment due to SARS-CoV-2 infection, and we suggest a conservative management in the absence of symptoms.
Collapse
Affiliation(s)
- Simone Meini
- Medicina Interna, Azienda USL Toscana centro, Ospedale Santa Maria Annunziata, Firenze, Italy
| | - Chiara Zini
- Radiologia, Azienda USL Toscana centro, Ospedale Santa Maria Annunziata, Firenze, Italy
| | - Maria Teresa Passaleva
- Medicina Interna, Azienda USL Toscana centro, Ospedale Santa Maria Annunziata, Firenze, Italy
| | - Anna Frullini
- Medicina Interna, Azienda USL Toscana centro, Ospedale Santa Maria Annunziata, Firenze, Italy
| | - Francesca Fusco
- Radiologia, Azienda USL Toscana centro, Ospedale Santa Maria Annunziata, Firenze, Italy
| | - Roberto Carpi
- Radiologia, Azienda USL Toscana centro, Ospedale Santa Maria Annunziata, Firenze, Italy
| | - Fiorella Piani
- Medicina Interna, Azienda USL Toscana centro, Ospedale Santa Maria Annunziata, Firenze, Italy
| |
Collapse
|
3
|
Zimmermann P, Curtis N. The effect of antibiotics on the composition of the intestinal microbiota - a systematic review. J Infect 2019; 79:471-489. [PMID: 31629863 DOI: 10.1016/j.jinf.2019.10.008] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/13/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Antibiotics change the composition of the intestinal microbiota. The magnitude of the effect of antibiotics on the microbiota and whether the effects are short-term or persist long-term remain uncertain. In this review, we summarise studies that have investigated the effect of antibiotics on the composition of the human intestinal microbiota. METHODS A systematic search was done to identify original studies that have investigated the effect of systemic antibiotics on the intestinal microbiota in humans. RESULTS We identified 129 studies investigating 2076 participants and 301 controls. Many studies reported a decrease in bacterial diversity with antibiotic treatment. Penicillin only had minor effects on the intestinal microbiota. Amoxicillin, amoxcillin/clavulanate, cephalosporins, lipopolyglycopeptides, macrolides, ketolides, clindamycin, tigecycline, quinolones and fosfomycin all increased abundance of Enterobacteriaea other than E. coli (mainly Citrobacter spp., Enterobacter spp. and Klebsiella spp.). Amoxcillin, cephalosporins, macrolides, clindamycin, quinolones and sulphonamides decreased abundance of E. coli, while amoxcillin/clavulante, in contrast to other penicillins, increased abundance of E. coli. Amoxicllin, piperacillin and ticarcillin, cephalosporins (except fifth generation cephalosporins), carbapenems and lipoglycopeptides were associated with increased abundance of Enterococcus spp., while macrolides and doxycycline decreased its abundance. Piperacillin and ticarcillin, carbapenems, macrolides, clindamycin and quinolones strongly decreased the abundance of anaerobic bacteria. In the studies that investigated persistence, the longest duration of changes was reported after treatment with ciprofloxacin (one year), clindamycin (two years) and clarithromycin plus metronidazole (four years). Many antibiotics were associated with a decrease in butyrate or butryrate-producing bacteria. CONCLUSION Antibiotics have profound and sometimes persisting effects on the intestinal microbiota, characterised by diminished abundance of beneficial commensals and increased abundance of potentially detrimental microorganisms. Understanding these effects will help tailor antibiotic treatment and the use of probiotics to minimise this 'collateral damage'.
Collapse
Affiliation(s)
- Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital HFR and Faculty of Science and Medicine, University of Fribourg, Switzerland; Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia.
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia
| |
Collapse
|
4
|
Das B, Sarkar C, Das D, Gupta A, Kalra A, Sahni S. Telavancin: a novel semisynthetic lipoglycopeptide agent to counter the challenge of resistant Gram-positive pathogens. Ther Adv Infect Dis 2017; 4:49-73. [PMID: 28634536 PMCID: PMC5467880 DOI: 10.1177/2049936117690501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Telavancin (TD-6424), a semisynthetic lipoglycopeptide vancomycin-derivative, is a novel antimicrobial agent developed by Theravance for overcoming resistant Gram-positive bacterial infections, specifically methicillin-resistant Staphylococcus aureus (MRSA). The US Food and Drug Administration (USFDA) had approved telavancin in 2009 for the treatment of complicated skin and skin structure infections (cSSSIs) caused by Gram-positive bacteria, including MRSA (S. aureus, Streptococcus agalactiae, Streptococcus pyogenes, Streptococcus anginosus group, or Enterococcus faecalis). Telavancin has two proposed mechanisms of action. In vitro, telavancin has a rapid, concentration-dependent bactericidal effect, due to disruption of cell membrane integrity. Telavancin has demonstrable in vitro activity against aerobic and anaerobic Gram-positive bacteria. Telavancin and vancomycin have similar spectra of activity. Gram-negative bacteria are usually non-susceptible to telavancin. Telavancin has been successfully tested in various animal models of bacteremia, endocarditis, meningitis, and pneumonia. Phase II Telavancin versus Standard Therapy for Treatment of Complicated Skin and Soft-Tissue Infections due to Gram-Positive Bacteria (FAST 1 and FAST 2) and phase III [Assessment of Telavancin in Complicated Skin and Skin Structure Infections 1 (ATLAS 1 and ATLAS 2)] clinical trials have been conducted for evaluating telavancin's efficacy and safety in cSSSIs. Phase III clinical trials have been carried out for evaluating telavancin's safety and efficacy in nosocomial pneumonia [Assessment of Telavancin for Treatment of Hospital acquired Pneumonia 1 and 2 (ATTAIN 1 and ATTAIN 2)]. A phase II randomized, double-blind, clinical trial has been carried out for evaluating telavancin's safety and efficacy in uncomplicated S. aureus bacteremia [Telavancin for Treatment of Uncomplicated S. aureus Bacteremia (ASSURE)]. Pacemaker lead-related infective endocarditis due to a vancomycin intermediate S. aureus (VISA) strain (non-daptomycin susceptible) was successfully treated with parenteral telavancin for 8 weeks. Telavancin extensively binds to serum albumin (~93%) and has a relatively small volume of distribution. Telavancin is not biotransformed by any cytochrome P450 microsomal enzymes and excreted mainly in the urine. Though well-tolerated, worrisome adverse effects, including renal dysfunction and QTc prolongation are of potential concern. Given its extensive binding to plasma proteins, long half-life, and a long post-antibiotic effect, it represents a promising addition to the therapeutic armamentarium in combating infections caused by resistant Gram-positive pathogens, namely, MRSA.
Collapse
Affiliation(s)
- Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| | - Chayna Sarkar
- Department of Pharmacology & Clinical Pharmacology, North Eastern Indira Gandhi Regional Institute of Health & Medical Sciences (NEIGRIHMS) Shillong, Shillong, India
| | - Debasmita Das
- Department of Computer Science & Engineering, Faculty of Engineering, Manipal University Jaipur, Dehmi Kalan, Jaipur Ajmer Expressway, Rajasthan, India
| | - Amit Gupta
- Department of Surgery, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| | - Arnav Kalra
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| | - Shubham Sahni
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| |
Collapse
|
5
|
Toutain PL, Ferran AA, Bousquet-Melou A, Pelligand L, Lees P. Veterinary Medicine Needs New Green Antimicrobial Drugs. Front Microbiol 2016; 7:1196. [PMID: 27536285 PMCID: PMC4971058 DOI: 10.3389/fmicb.2016.01196] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/19/2016] [Indexed: 01/08/2023] Open
Abstract
Given that: (1) the worldwide consumption of antimicrobial drugs (AMDs) used in food-producing animals will increase over the coming decades; (2) the prudent use of AMDs will not suffice to stem the rise in human antimicrobial resistance (AMR) of animal origin; (3) alternatives to AMD use are not available or not implementable, there is an urgent need to develop novel AMDs for food-producing animals. This is not for animal health reasons, but to break the link between human and animal resistomes. In this review we establish the feasibility of developing for veterinary medicine new AMDs, termed "green antibiotics," having minimal ecological impact on the animal commensal and environmental microbiomes. We first explain why animal and human commensal microbiota comprise a "turnstile" exchange, between the human and animal resistomes. We then outline the ideal physico-chemical, pharmacokinetic, and pharmacodynamic properties of a veterinary green antibiotic and conclude that they can be developed through a rational screening of currently used AMD classes. The ideal drug will be hydrophilic, of relatively low potency, slow clearance and small volume of distribution. It should be eliminated principally by the kidney as inactive metabolite(s). For oral administration, bioavailability can be enhanced by developing lipophilic pro-drugs. For parenteral administration, slow-release formulations of existing eco-friendly AMDs with a short elimination half-life can be developed. These new eco-friendly veterinary AMDs can be developed from currently used drug classes to provide alternative agents to those currently used in veterinary medicine and mitigate animal contributions to the human AMR problem.
Collapse
Affiliation(s)
- Pierre-Louis Toutain
- Ecole Nationale Vétérinaire de Toulouse, Institut National de la Recherche Agronomique, TOXALIM, Université de ToulouseToulouse, France
| | - Aude A. Ferran
- Ecole Nationale Vétérinaire de Toulouse, Institut National de la Recherche Agronomique, TOXALIM, Université de ToulouseToulouse, France
| | - Alain Bousquet-Melou
- Ecole Nationale Vétérinaire de Toulouse, Institut National de la Recherche Agronomique, TOXALIM, Université de ToulouseToulouse, France
| | - Ludovic Pelligand
- Comparative Biomedical Sciences, The Royal Veterinary CollegeHatfield, UK
| | - Peter Lees
- Comparative Biomedical Sciences, The Royal Veterinary CollegeHatfield, UK
| |
Collapse
|
6
|
Toutain PL, Bousquet-Melou A. Rebuttal to the reaction of the EGGVP to the review article 'the consequences of generic marketing on antibiotic consumption and the spread of microbial resistance: the need for new antibiotics'. J Vet Pharmacol Ther 2015; 37:618-23. [PMID: 25399727 DOI: 10.1111/jvp.12166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- P L Toutain
- Ecole Nationale Veterinaire de Toulouse, Toulouse, France
| | | |
Collapse
|
7
|
Wang X, Paul JA, Nanovskaya TN, Hankins GDV, Ahmed MS. Quantitative determination of telavancin in pregnant baboon plasma by solid-phase extraction and LC-ESI-MS. J Pharm Biomed Anal 2014; 98:107-12. [PMID: 24905291 PMCID: PMC4127372 DOI: 10.1016/j.jpba.2014.04.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 04/22/2014] [Accepted: 04/28/2014] [Indexed: 11/15/2022]
Abstract
The increasing incidence and severity of methicillin- and vancomycin-resistant infections during pregnancy prompted further development of telavancin. The understanding of the pharmacokinetics of telavancin during pregnancy is critical to optimize dosing. Due to ethical and safety concerns the study is conducted on the pregnant baboons. A method using solid-phase extraction coupled with liquid chromatography-single quadrupole mass spectrometry for the quantitative determination of telavancin in baboon plasma samples was developed and validated. Teicoplanin was used as an internal standard. Telavancin was extracted from baboon plasma samples by using Waters Oasis(®) MAX 96-Well SPE plate and achieved extraction recovery was >66% with variation <12%. Telavancin was separated on Waters Symmetry C18 column with gradient elution. Two SIM channels were monitored at m/z 823 and m/z 586 to achieve quantification with simultaneous confirmation of telavancin identification in baboon plasma samples. The linearity was assessed in the range of 0.188μg/mL to75.0μg/mL, with a correlation coefficient of 0.998. The relative standard deviation of this method was <11% for within- and between-run assays, and the accuracy ranged between 96% and 114%.
Collapse
Affiliation(s)
- Xiaoming Wang
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Jonathan A Paul
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Tatiana N Nanovskaya
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Gary D V Hankins
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA
| | - Mahmoud S Ahmed
- Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-0587, USA.
| |
Collapse
|
8
|
Rashid MU, Dalhoff A, Bäckström T, Björkhem-Bergman L, Panagiotidis G, Weintraub A, Nord CE. Ecological impact of MCB3837 on the normal human microbiota. Int J Antimicrob Agents 2014; 44:125-30. [PMID: 24931053 DOI: 10.1016/j.ijantimicag.2014.03.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/26/2014] [Accepted: 03/30/2014] [Indexed: 11/30/2022]
Abstract
MCB3837 is a novel, water-soluble, injectable prodrug that is rapidly converted to the active substance MCB3681 in vivo following intravenous (i.v.) administration. Both MCB3837 and MCB3681 are oxazolidinone-quinolone hybrid molecules. The purpose of the present study was to investigate the effect of MCB3681 on the human skin, nose, oropharyngeal and intestinal microbiota following administration of MCB3837. Twelve healthy male subjects received i.v. MCB3837 (6 mg/kg body weight) once daily for 5 days. Skin, nose, saliva and faecal samples were collected on Day -1 (pre dose), during administration on Days 2 and 5, and post dose on Days 8, 12 and 19. Micro-organisms were identified to genus level. No measurable concentrations of MCB3681 were found in any saliva samples or in the faecal samples on Day -1. On Day 2, 10 volunteers had faecal MCB3681 concentrations between 16.5 mg/kg faeces and 275.1mg/kg faeces; no MCB3681 in faeces could be detected in two of the volunteers. On Day 5, all volunteers had faecal concentrations of MCB3681 ranging from 98.9 to 226.3 mg/kg. MCB3681 caused no ecological changes in the skin, nasal and oropharyngeal microbiota. The numbers of enterococci, bifidobacteria, lactobacilli and clostridia decreased in the intestinal microbiota during administration of the drug. Numbers of Escherichia coli, other enterobacteria and Candida were not affected during the study. There was no impact on the number of Bacteroides. The faecal microbiota was normalised on Day 19. No new colonising aerobic or anaerobic Gram-positive bacteria with MCB3681 minimum inhibitory concentrations of ≥4 mg/L were found.
Collapse
Affiliation(s)
- Mamun-Ur Rashid
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Axel Dalhoff
- University Medical Center Schleswig-Holstein, Institute for Infection Medicine, Brunswiker Str. 4, D-24105 Kiel, Germany
| | - Tobias Bäckström
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Linda Björkhem-Bergman
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Georgios Panagiotidis
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Andrej Weintraub
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Carl Erik Nord
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
9
|
Rashid MU, Weintraub A, Nord CE. Effect of new antimicrobial agents on the ecological balance of human microflora. Anaerobe 2011; 18:249-53. [PMID: 22155131 DOI: 10.1016/j.anaerobe.2011.11.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 11/21/2011] [Accepted: 11/23/2011] [Indexed: 11/26/2022]
Abstract
The human normal microflora is relatively stable at each ecological habitat under normal circumstances and acts as a barrier against colonization by potentially pathogenic microorganisms and against overgrowth of already present opportunistic microorganisms. Administration of antimicrobial agents causes disturbances in the ecological balance between the host and the normal microflora. The risk of emergence and spread of resistant strains between patients and dissemination of resistant determinants between microorganisms is reduced if colonization resistance is not disturbed by antimicrobial agents. In this article, the potential ecological effects of administration of new antimicrobial agents on the intestinal and oropharyngeal microflora are summarized. The review is based on clinical studies published during the past 10 years.
Collapse
Affiliation(s)
- Mamun-Ur Rashid
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|