1
|
Elftooh Awaga HA, Anwar YT, Ait-Allah AS, Abdelkareem AO. Oral versus intravenous antibiotic prophylaxis before obstetric and gynecological surgical interventions: A randomized clinical trial. Eur J Obstet Gynecol Reprod Biol 2025; 311:114020. [PMID: 40334373 DOI: 10.1016/j.ejogrb.2025.114020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVE The current study was conducted to compare the efficacy of prophylactic intravenous versus oral (Cephradin) for the prevention of surgical site infections (SSI) in Obstetric and Gynecological interventions. METHODS A randomized clinical trial was carried out on 230 patients fulfilling the eligibility criteria between 1st October 2023 and 31st July 2024, including pregnant women who were approached prior to their delivery and non-pregnant women who were approached prior to their intended interventions. (group A) 116 patients received oral antimicrobial prophylaxis, while (group B) 114 patients received intravenous antimicrobial prophylaxis. Furthermore, all included patients received oral antibiotics postoperatively (oral cephradine 500 mg/ 12 h for 7 days). Patients were evaluated twice; the 1st assessment was done 1 week, while the 2nd assessment was done a month after the intervention. RESULTS SSI rate was 14 % CI (0.07-0.20) in the oral group compared to 13 % CI (0.07-0.20) in the intravenous group after one week with no significant difference in between (p = 0.84). However, the SSI rate was 5.5 % CI (0.02-0.11) in the oral group compared to 6.3 % CI (0.02-0.12) in the intravenous group after one month of the intervention, with no significant difference in between (p = 0.78). CONCLUSION The current study showed that oral antibiotic prophylaxis before obstetric and gynecological interventions seems to be safe, easy, practical, cheap, and as effective as parenteral antibiotic prophylaxis in the prevention of SSI. However, more randomized trials with larger sample sizes are needed to pave the way for replacing parenteral with oral antibiotic chemoprophylaxis. TRIAL REGISTRATION The study was approved by the Sohag Ethical Committee, Faculty of Medicine, with IRB number (Soh-Med-23-09-05MS) and was registered on clinical trial.gov with T.R.N (NCT06255652).
Collapse
Affiliation(s)
- Hatem Abo Elftooh Awaga
- Department of Obstetrics and Gynecology, Faculty of Medicine, Sohag University, Sohag, Egypt.
| | - Yasmeen Tharwat Anwar
- Department of Obstetrics and Gynecology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Abdou Saeed Ait-Allah
- Department of Obstetrics and Gynecology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Amr Othman Abdelkareem
- Department of Obstetrics and Gynecology, Faculty of Medicine, Sohag University, Sohag, Egypt
| |
Collapse
|
2
|
Smeets RMA, van Beynum IM, van Kesteren C, Cornette JMJ, van der Zande JA, Roos-Hesselink JW, Beex-Oosterhuis MM, Flint RB. The maternal exposure of digoxin and flecainide in relation to the safety and effectiveness in the treatment of non-hydropic fetal tachycardia. Heart Rhythm 2025:S1547-5271(25)02499-3. [PMID: 40412599 DOI: 10.1016/j.hrthm.2025.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Sustained fetal tachycardia requires transplacental anti-arrhythmic therapy. Little is known about the dose-concentration-effect correlation and safety in the mother, fetus, and newborn. OBJECTIVE This study evaluates the relationship between maternal dose of digoxin and flecainide therapy for fetal tachycardia, maternal and umbilical cord concentrations, and side effects. METHODS This was a retrospective case series. We included 28 pregnant women initially treated with digoxin monotherapy for fetal tachycardia between June 2007 and January 2023. The main end point was the correlation between maternal drug exposure, effect, and side effects. RESULTS Oral digoxin monotherapy converted 9 fetuses (32%) to sinus rhythm after a median of 4.5 days (interquartile range [IQR] 3-6.5). Overall, 18 fetuses required additional oral flecainide (300 mg daily), resulting in a total conversion rate of 93% (26/28). Equal starting doses of digoxin caused similar maternal digoxin concentrations regardless of gestational age, with no significant difference between responders and non-responders (P = .504). Side effects, primarily nausea, led to dose reductions, but treatment remained effective. Maternal digoxin concentrations remained stable throughout pregnancy and little inter-patient variability was observed. Flecainide exposure varied both within and between patients. The median fetus/mother digoxin ratio was similar in both monotherapy (n = 3) and combination (n = 9) therapy groups (0.51 (IQR 0.28-0.76) vs 0.45 (IQR 0.39-0.64), P = .864). The median fetus/mother flecainide ratio was 0.82 (IQR 0.69-1.29). CONCLUSION Digoxin monotherapy successfully treated fetal tachycardia in only 32% of cases. Adding flecainide improved response to 93%, although it increased side effects, which could be managed with dose reductions. Maternal digoxin levels were stable throughout pregnancy.
Collapse
Affiliation(s)
- Rosa M A Smeets
- Department of Clinical Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Clinical Pharmacy, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Ingrid M van Beynum
- Division Pediatric Cardiology, Department of Paediatrics, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Jérôme M J Cornette
- Department of Obstetrics and Fetal Medicine, Erasmus University Medical Center - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johanna A van der Zande
- Department of Obstetrics and Fetal Medicine, Erasmus University Medical Center - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Cardiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | - Robert B Flint
- Department of Clinical Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands; Division of Neonatology, Department of Neonatal and Pediatric Intensive Care, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Motiwala ZY, Raza S, Fernet LY, Kamal FA, Misra S, Briceño Silva GD, Roy T, Anwer F, Hamza Habib M. Cancer-Related Pain Management in Pregnancy-A Review. J Palliat Med 2025. [PMID: 40401437 DOI: 10.1089/jpm.2024.0553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
Background: Cancer-related pain management in pregnant patients is a complex clinical challenge requiring a careful balance between effective maternal analgesia and fetal safety. The pathophysiology of cancer-related pain varies, necessitating a nuanced, individualized approach to treatment with careful consideration of maternal-fetal safety, and pharmacokinetics. Objectives: This review aims to explore safe and effective pharmacological and interventional strategies for managing cancer-related pain in pregnant patients while highlighting disparities in access to care. Design: A comprehensive literature review was conducted. A total of 4766 articles were identified, with 98 studies included for final analysis after exclusion. Setting/Subjects: Studies included pregnant individuals with various hematological and solid tumor malignancies across various community and academic settings. Measurements: Pain typology was categorized (nociceptive or neuropathic), with treatment options including commonly available over-the-counter medications, opioids, various neuropathic medications, and interventional pain management options. In addition to pharmacological efficacy and maternofetal safety profiles, interventional efficacy, and socio-cultural influences on care were also measured. Disparities in access to care were also analyzed. Results: Analgesic options in pregnancy include acetaminophen, opioids, certain neuropathic medications, with very cautious use of NSAIDs. Interventional methods-neuraxial anesthesia, nerve blocks, spinal cord stimulation-offer adjunctive benefit if done under ultrasound guidance, but fluoroscopy and radiation guided modalities are best avoided in pregnancy. Diverse populations often encountered significant barriers to adequate pain control. Conclusions: Optimal cancer pain management in pregnancy necessitates a multidisciplinary, and individually tailored approach. Enhancing clinical evidence, and ensuring culturally competent care are critical to improving outcomes in this patient population.
Collapse
Affiliation(s)
| | - Shahzad Raza
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Lucas Y Fernet
- University of Nottingham, Nottingham, Nottingham, England
| | - Faiza A Kamal
- University of Nottingham, Nottingham, Nottingham, England
| | | | | | - Tara Roy
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Faiz Anwer
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Muhammad Hamza Habib
- Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
4
|
Sawe S, Tsirizani L, Court R, Gausi K, Poswa A, Badat T, Wiesner L, Loveday M, Maartens G, Conradie F, Denti P. The effect of pregnancy on the population pharmacokinetics of levofloxacin in South Africans with rifampicin-resistant tuberculosis. Antimicrob Agents Chemother 2025; 69:e0162624. [PMID: 40167446 PMCID: PMC12057369 DOI: 10.1128/aac.01626-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/22/2025] [Indexed: 04/02/2025] Open
Abstract
Levofloxacin is a key drug in the prevention and treatment of rifampicin-resistant tuberculosis (RR-TB). There are limited data describing the effect of pregnancy on the pharmacokinetics of levofloxacin. We aimed to characterize the pharmacokinetics of levofloxacin in adults with RR-TB, including the effect of pregnancy. We pooled data from two studies conducted in adult participants treated for RR-TB in South Africa. Treatment regimens in both studies included levofloxacin dosed at 750/1000 mg daily, depending on body weight. We analyzed data from 47 participants, 31 (66%) living with HIV, using nonlinear mixed-effects modeling in NONMEM v7.5.1. Out of 33 female participants, 21 were pregnant, of whom 12 contributed matched antepartum and postpartum pharmacokinetic profiles. Levofloxacin followed one-compartment pharmacokinetics with first-order elimination and absorption with transit absorption compartments. The clearance and volume of distribution for a typical non-pregnant participant (weight: 58 kg; age: 32 years; serum creatinine: 56.2 µmol/L) were 6.06 (95% confidence interval [CI], 5.47 to 6.53) L/h and 85.9 (95% CI, 80.6 to 91.7) L, respectively. Higher serum creatinine levels were associated with lower levofloxacin clearance using a power function with an exponent of -0.367 (95% CI, -0.493 to -0.104). Pregnancy increased levofloxacin clearance by 38.1% (95% CI, 23.4% to 57.1%), with substantially lower exposures in pregnant compared with non-pregnant participants receiving equivalent weight-based doses. To achieve non-pregnant equivalent exposures of levofloxacin, an additional 250 mg tablet may be required, although further study is needed to assess the safety implications of a higher recommended dose in pregnant women.
Collapse
Affiliation(s)
- Sharon Sawe
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Lufina Tsirizani
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Richard Court
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Kamunkhwala Gausi
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Asanda Poswa
- Department of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Tasnim Badat
- Department of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Marian Loveday
- HIV and Other Infectious Diseases Research Unit (HIDRU), South African Medical Research Council, Durban, South Africa
| | - Gary Maartens
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Francesca Conradie
- Department of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Paolo Denti
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
5
|
Scherf-Clavel M, Leutritz AL, Gehrmann A, Unterecker S, Walther S, Kittel-Schneider S. Physiologically based pharmacokinetic modelling predicts altered maternal pharmacokinetics of amitriptyline during pregnancy. Br J Clin Pharmacol 2025. [PMID: 40312020 DOI: 10.1002/bcp.70084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/24/2025] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
AIMS Pharmacotherapy of maternal peripartum depression is an increasing challenge. Amitriptyline (AMI) is the most often used tricyclic antidepressant during pregnancy, but knowledge on pharmacokinetics in this special phase is lacking. Physiologically based pharmacokinetic (PBPK) modelling is a powerful tool to better understand pregnancy-induced pharmacokinetic changes of medication. We aimed to improve the knowledge about AMI pharmacokinetics during pregnancy using PBPK modelling. Consequently, we aimed to add new information for an effective and safe pharmacotherapy in pregnant women. METHODS A PBPK model, including AMI, but also its active metabolite nortriptyline (NOR), was developed to investigate pregnancy-induced pharmacokinetic changes after AMI administration. The predicted drug exposure was compared to observed concentrations in pregnant patients in clinical routine. The PBPK model was set up using PK-Sim Version 11. RESULTS Serum concentration profiles were described successfully. During pregnancy, active moiety serum concentration of AMI (AMI + NOR) did not change; however, AMI concentration increased, whereas NOR concentration decreased. CONCLUSIONS With this model, we added valuable information on AMI pharmacokinetics during pregnancy (increased AMI concentration, decreased NOR concentration). For clinical practice the treating physician should be aware that despite active moiety serum concentration comparable to before pregnancy, tolerability may be affected due to increased AMI serum concentrations and as consequence increased anticholinergic effects. To keep the risk of therapy discontinuation during pregnancy low, we suggest performing therapeutic drug monitoring, especially to check the AMI serum concentration.
Collapse
Affiliation(s)
- Maike Scherf-Clavel
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Anna Linda Leutritz
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Andrea Gehrmann
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Stefan Unterecker
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Social Foundation Bamberg, Teaching Hospital of the University of Erlangen, Germany
| | - Sebastian Walther
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
- APC Microbiome, University College Cork, Cork, Ireland
| |
Collapse
|
6
|
Fashe MM, Tiley JB, Lee CR. Mechanisms of altered hepatic drug disposition during pregnancy: small molecules. Expert Opin Drug Metab Toxicol 2025; 21:445-462. [PMID: 39992297 PMCID: PMC11961323 DOI: 10.1080/17425255.2025.2470792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Pregnancy alters the systemic exposure and clearance of many hepatically cleared drugs that are commonly used by obstetric patients. Understanding the molecular mechanisms underlying the changes in factors that affect hepatic drug clearance (blood flow, protein binding, and intrinsic clearance) is essential to more precisely predict systemic drug exposure and dose requirements in obstetric patients. AREAS COVERED This review (1) summarizes the anatomic, physiologic, and biochemical changes in maternal hepatic, cardiovascular, endocrine, and renal systems relevant to hepatic drug clearance and (2) reviews the molecular mechanisms underlying the altered hepatic metabolism and intrinsic clearance of drugs during pregnancy via a comprehensive PubMed search. It also identifies knowledge gaps in the molecular mechanisms and factors that modulate hepatic drug clearance during pregnancy. EXPERT OPINION Pharmacokinetic studies have shown that pregnancy alters systemic exposure, protein binding, and clearance of many drugs during gestation in part due to pregnancy-associated decreases in plasma albumin, increases in organ blood flow, and changes in the activity of drug-metabolizing enzymes (DMEs) and transporters. The changes in the activity of certain DMEs and transporters during pregnancy are likely driven by hormonal-changes that inhibit their activity or alter the expression of these proteins through activation of transcription factors.
Collapse
Affiliation(s)
- Muluneh M. Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Jacqueline B. Tiley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| |
Collapse
|
7
|
Quattrini E, Ireland DJ, Keelan JA. The risk tolerance and decision-making processes of Australian women regarding medication trials in pregnancy. Aust N Z J Obstet Gynaecol 2025; 65:218-226. [PMID: 39333018 PMCID: PMC12099191 DOI: 10.1111/ajo.13884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/27/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Pregnant women have historically been excluded from participation in medication trials, in part due to the perceived risks of drug exposure to mothers and fetuses. However, little is known about pregnant women's attitudes toward risk and participation in such trials. AIMS To address this knowledge gap and to identify factors that influence trial participation. MATERIALS AND METHODS Australian women over the age of 18, currently pregnant or within six months of delivery, were recruited to participate in an online survey (n = 623) and follow-up interviews (n = 11). The survey investigated willingness to participate in five hypothetical drug trial scenarios of varying risk. Demographic and obstetric information, including COVID-19 vaccination status, was also collected. The impact of these factors on trial participation was analysed using ordinal regression. Interviews were subjected to thematic framework analysis using a priori and emergent themes. RESULTS Nearly half of the respondents (48%) indicated a willingness to participate in at least one of the hypothetical trials. As trial risk increased participation likelihood decreased, especially if the risk was to the fetus, regardless of benefits to the mother. COVID-19 vaccination status and medication hesitancy were predictors of an unwillingness to participate. Three broad themes emerged from the qualitative data: risk-benefit analysis, quality of evidence, and trust. CONCLUSIONS Overall, participants expressed a positive attitude toward research and medication trials during pregnancy, but were concerned about fetal risk. The findings of this study may help enhance trial design and the participation of pregnant women in medication trials.
Collapse
Affiliation(s)
- Eva Quattrini
- School of Biomedical SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Demelza J. Ireland
- School of Biomedical SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Jeffrey A. Keelan
- School of Biomedical SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
| |
Collapse
|
8
|
Thunbo M, Vendelbo J, Witte D, Larsen A, Pedersen L. Maternal Demographic Patterns in Medication use During Pregnancy: A Nationwide Register Study. Basic Clin Pharmacol Toxicol 2025; 136:e70020. [PMID: 40103276 PMCID: PMC11920599 DOI: 10.1111/bcpt.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
In recent years, medication use during pregnancy has increased, yet its association with maternal characteristics remains unclear. To address this gap, we aimed to investigate how maternal age, pre-gestational body mass index (BMI), smoking, parity, ethnic origin and employment status relate to medication use during pregnancy. We conducted a nationwide Danish registry study, including 698 447 clinically recognised pregnancies with a gestational age of at least 10 weeks, spanning from 2008 to 2018. Medication use was estimated based on prescription redemptions during pregnancy and stratified by the demographic factors of interest. Overall, 60.3% of pregnant women redeemed at least one prescription, while 28.9% redeemed multiple medications. Notably, higher usage was observed among women aged 35 or older, those with a BMI of 30 kg/m2 or more, smokers, multipara, Black women, and early retirees. Medication combination patterns differed with the demographic subgroups. These findings highlight notable differences in medication use among demographic groups during pregnancy, underscoring the need for tailored healthcare strategies during pregnancy.
Collapse
Affiliation(s)
- Mette Østergaard Thunbo
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of Clinical PharmacologyAarhus University HospitalAarhusDenmark
| | | | - Daniel R. Witte
- Department of Public HealthAarhus UniversityAarhusDenmark
- Steno Diabetes Centre AarhusAarhus University HospitalAarhusDenmark
| | - Agnete Larsen
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Lars Henning Pedersen
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Obstetrics and GynaecologyAarhus University HospitalAarhusDenmark
| |
Collapse
|
9
|
Decroocq J, Wu S, Barrois M, Pinson P, Boussaid I, Birsen R. [Haematological malignancies in pregnancy]. Med Sci (Paris) 2025; 41:374-379. [PMID: 40293151 DOI: 10.1051/medsci/2025044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
The management of haematological malignancies in pregnancy represents a complex medical challenge. These rare conditions require multidisciplinary collaboration to address both the maternal risks associated with physiological changes and the foetal risks, such as malformations or prematurity associated with treatment. However, data to guide the care of these patients are still limited. Comprehensive research is needed to better understand these diseases, assess the impact of treatments and optimise care, including the involvement of pregnant patients in clinical trials to improve their access to innovative therapies. This article provides an overview of the issues involved in the management of haematological malignancies during pregnancy, highlighting the diagnostic, therapeutic and prognostic challenges.
Collapse
Affiliation(s)
- Justine Decroocq
- Service d'hématologie clinique, Hôpital Cochin-Port Royal (AP-HP), Institut Cochin, Université Paris Cité, Paris, France
| | - Sangsong Wu
- Service d'hématologie clinique, Hôpital Cochin-Port Royal (AP-HP), Institut Cochin, Université Paris Cité, Paris, France
| | - Mathilde Barrois
- Service d'obstétrique-HAD, Hôpital Cochin-Port Royal (AP-HP), Paris, France
| | - Pierre Pinson
- Unité de recherche clinique, Hôpital Cochin-Port Royal (AP-HP), Institut Cochin, Paris, France
| | - Ismaël Boussaid
- Service d'hématologie biologique, Hôpital Cochin-Port Royal (AP-HP), Institut Cochin, Université Paris Cité, Paris, France
| | - Rudy Birsen
- Service d'hématologie clinique, Hôpital Cochin-Port Royal (AP-HP), Institut Cochin, Université Paris Cité, Paris, France
| |
Collapse
|
10
|
Dibbets AC, Koldeweij C, Osinga EP, Scheepers HCJ, de Wildt SN. Barriers and Facilitators for Bringing Model-Informed Precision Dosing to the Patient's Bedside: A Systematic Review. Clin Pharmacol Ther 2025; 117:633-645. [PMID: 39659053 PMCID: PMC11835426 DOI: 10.1002/cpt.3510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Model-informed precision dosing (MIPD) utilizes mathematical models to predict optimal medication doses for a specific patient or patient population. However, the factors influencing the implementation of MIPD have not been fully elucidated, hindering its widespread use in clinical practice. A systematic review was conducted in PubMed from inception to December 2022, aiming to identify barriers and facilitators for the implementation of MIPD into patient care. Articles with a focus on implementation of MIPD were eligible for this review. After screening titles and abstracts, full articles investigating the clinical implementation of MIPD were included for data extraction. Of 790 records identified, 15 publications were included. A total of 72 barriers and facilitators across seven categories were extracted through a hybrid thematic analysis. Barriers comprised limited data for model validation, unclear regulatory pathways for model endorsement and additional drug level measurements required for certain types of MIPD. Facilitators encompassed the development of user-friendly MIPD tools continuously updated based on user feedback and data. Collaborative efforts among diverse stakeholders for model validation and implementation, along with education of end-users, may promote the utilization of MIPD in patient care. Despite ongoing challenges, this systematic review revealed various strategies to facilitate the clinical implementation of MIPD.
Collapse
Affiliation(s)
- Anna Caroline Dibbets
- Division of Pharmacology and Toxicology, Department of PharmacyRadboud University Medical CenterNijmegenThe Netherlands
- Department of Obstetrics and GynaecologyMaastricht University Medical CenterMaastrichtThe Netherlands
- GROW, Institute for Oncology and ReproductionMaastrichtThe Netherlands
| | - Charlotte Koldeweij
- Division of Pharmacology and Toxicology, Department of PharmacyRadboud University Medical CenterNijmegenThe Netherlands
| | - Esra P. Osinga
- Division of Pharmacology and Toxicology, Department of PharmacyRadboud University Medical CenterNijmegenThe Netherlands
| | - Hubertina C. J. Scheepers
- Department of Obstetrics and GynaecologyMaastricht University Medical CenterMaastrichtThe Netherlands
- GROW, Institute for Oncology and ReproductionMaastrichtThe Netherlands
| | - Saskia N. de Wildt
- Division of Pharmacology and Toxicology, Department of PharmacyRadboud University Medical CenterNijmegenThe Netherlands
- Department of Pediatric and Neonatal Intensive CareErasmus MC‐Sophia Children's HospitalRotterdamThe Netherlands
- Department of Intensive CareRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
11
|
Ruiz P, Cheng PY, Desai S, Shin M, Jarrett JM, Ward CD, Shim YK. Prevalence of Exposure to Environmental Metal Mixtures Among Pregnant Women in the United States National Health and Nutrition Examination Survey (NHANES) 1999-2018. J Xenobiot 2025; 15:38. [PMID: 40126256 PMCID: PMC11932210 DOI: 10.3390/jox15020038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 03/25/2025] Open
Abstract
Although exposure to metals remains a public health concern, few studies have examined exposure to combinations of metals. This study characterized prevalent combinations of cadmium (Cd), mercury (Hg), and lead (Pb) in women (n = 10,152; aged 20-44 years) who participated in the U.S. National Health and Nutrition Examination Survey (NHANES) 1999-2018. To explore relative metal exposures within this population, Cd, Hg, and Pb blood levels were dichotomized as "high" and "low" categories using median values to represent the center of the metal concentrations in the study population, not thresholds for adverse health effects. The prevalence of the three metal combinations at "high" levels (singular, binary, tertiary combinations) was calculated. Multinomial logistic regression was used to calculate odds ratios for each combination relative to none of these combinations after adjusting for potential confounders. Among the pregnant women (n = 1297), singular Hg was most prevalent (19.2% [95% CI 15.0-23.3]), followed by singular Cd (14.7% [95% CI 11.2-18.2]), tertiary combination Cd/Hg/Pb (11.0% [95% CI 8.7-13.2]), binary combinations Cd/Pb (9.8% [95% CI 7.4-12.2]), Hg/Pb (9.2% [95% CI 6.5-11.8]), Cd/Hg (7.8% [95% CI 6.0-9.6]), and singular Pb (5.5% [95% CI 4.1-6.9]). We found significantly lower odds of having Cd/Hg/Pb (adjusted odds ratio (adjOR) = 0.49: p < 0.001) and Cd/Pb (adjOR = 0.68: p < 0.0364) combinations among pregnant women compared to non-pregnant women. The odds of having higher levels of singular Pb were significantly lower (adjOR = 0.31: p < 0.0001) in women pregnant in their first and second trimesters (n = 563) than in non-pregnant women (n = 6412), whereas, though nonsignificant, the odds were higher for women pregnant in their third trimester (n = 366) (adjOR = 1.25: p = 0.4715). These results indicate the possibility that the fetus might be exposed to higher levels of the metal mixtures due to placental transfer, particularly to Pb, during the early stages of pregnancy. Further research is warranted to understand the relationship between metal combination exposures during pregnancy and maternal and infant health.
Collapse
Affiliation(s)
- Patricia Ruiz
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| | - Po-Yung Cheng
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| | - Siddhi Desai
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Mikyong Shin
- Division of Environmental Health Science and Practice, National Center for Environmental Health, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| | - Jeffery M. Jarrett
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| | - Cynthia D. Ward
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| | - Youn K. Shim
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30329, USA
| |
Collapse
|
12
|
Fashe MM, Miner TA, Collazo VL, Grieco JT, Fallon JK, Jackson KD, Lee CR. Impact of sex and pregnancy on hepatic CYP3A4 expression and activity in a humanized mouse model. Drug Metab Dispos 2025; 53:100025. [PMID: 40023573 DOI: 10.1016/j.dmd.2024.100025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 03/04/2025] Open
Abstract
Cytochrome P450 (CYP) 3A4 is an essential drug-metabolizing enzyme in humans, which shows substantial interindividual variation in response to various intrinsic and extrinsic factors such as sex and pregnancy. In humans, higher CYP3A4 metabolism has been observed in females compared with that in males and in pregnant compared with that in nonpregnant individuals, which has been linked to increased CYP3A4 expression in liver. However, sex differences and pregnancy-mediated changes in hepatic CYP3A4 expression and activity in vivo are not fully understood. In this study, we investigated the utility of a genetically engineered humanized mouse model that carries human CYP3A4/7, pregnane X receptor (PXR) and constitutive androstane receptor (CAR) (huPXR/CAR/CYP3A4/7) to recapitulate sex-associated and pregnancy-associated differences in the hepatic CYP3A4 expression and metabolism observed in humans. We found that female huPXR/CAR/CYP3A4/7 mice exhibited higher basal CYP3A4 mRNA levels and CYP3A4 absolute protein concentrations in liver, and higher 1-hydroxymidazolam formation in liver microsomes, compared with male humanized mice. In contrast, pregnant huPXR/CAR/CYP3A4/7 mice exhibited lower CYP3A4 mRNA levels, CYP3A4 absolute protein concentrations, and 1-hydroxymidazolam formation compared with nonpregnant and postpartum humanized mice. Expression of CAR and Cyp2b10 (a CAR responsive gene) were also higher in females and decreased during pregnancy and were positively correlated with hepatic CYP3A4 mRNA levels. Overall, the huPXR/CAR/CYP3A4/7 mouse model demonstrated utility to study higher basal hepatic CYP3A4 metabolism in females compared with that in males in vivo; however, this humanized mouse model did not demonstrate utility to study pregnancy-mediated increases in CYP3A4 drug substrate metabolism and clearance observed in humans. SIGNIFICANCE STATEMENT: This study assessed the impact of sex and pregnancy on hepatic CYP3A4 protein concentrations and metabolism in humanized PXR/CAR/CYP3A4 mice. Consistent with humans, female mice demonstrated higher hepatic CYP3A4 expression and activity than male mice. In contrast, pregnant mice showed decreased CYP3A4 expression and metabolism compared with nonpregnant mice. The humanized mouse model appeared useful to evaluate sex differences in basal hepatic CYP3A4 metabolism in vivo, but not to study the pregnancy-mediated increase in CYP3A4 metabolism observed during human pregnancy.
Collapse
Affiliation(s)
- Muluneh M Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Taryn A Miner
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Valeria Laboy Collazo
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph T Grieco
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John K Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
13
|
Al Arashi W, Cloesmeijer ME, Leebeek FWG, Duvekot JJ, Kruip MJHA, Mathôt RAA, Cnossen MH, the OPTI‐CLOT/To WiN study group and SYMPHONY consortium. Replacement therapy in pregnant women with von Willebrand disease during delivery: Factor levels and pharmacokinetics. Hemasphere 2025; 9:e70061. [PMID: 39760000 PMCID: PMC11696247 DOI: 10.1002/hem3.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 01/07/2025] Open
Abstract
Limited data are available on VWF activity (VWF:Act) and factor VIII (FVIII:C) levels during delivery after VWF/FVIII concentrate administration in women with von Willebrand disease (VWD). We aimed to evaluate treatment with a specific VWF/FVIII concentrate on factor levels in women with VWD during delivery and the postpartum period. A retrospective single-center study was conducted between January 1, 2008, and August 1, 2022. Pregnant women treated with Haemate®P during delivery were included if they had ≥2 consecutive VWF:Act and FVIII:C measurements post-infusion. VWF:Act/FVIII:C levels were compared to predefined target levels. A population pharmacokinetic (PopPK) model was developed, estimating VWF and FVIII pharmacokinetics after Haemate®P administration. Nineteen women were included. Targeted VWF:Act/FVIII:C peak levels were achieved after the first infusion (≥1.00 IU/mL, n = 12; ≥1.50 IU/mL, n = 5), and all VWF:Act/FVIII:C trough levels remained ≥0.50 IU/mL during first 72 h of treatment. All women had pretreatment FVIII:C levels ≥1.00 IU/mL, except one woman with type 2N, which was significantly higher than FVIII:C levels during the third trimester (median increase: 0.42 IU/mL, interquartile range: [0.12-0.92]). FVIII:C trough levels increased during treatment, median 2.05 IU/mL [1.65-2.71]. Nine women (47%) experienced postpartum hemorrhage and no thrombosis occurred. A one-compartment PopPK model adequately described VWF:Act/FVIII:C levels. Targeted VWF:Act/FVIII:C peak levels were achieved with the prescribed dosing regimens. VWF clearance was similar to that in nonpregnant individuals. Both pretreatment and FVIIIC trough levels during treatment were high with reduced FVIII clearance. Monitoring VWF:Act/FVIII:C levels is recommended for optimizing target levels and enriching the current PopPK model, improving VWF:Act/FVIII:C level predictions, and achieving more effective dosing.
Collapse
Affiliation(s)
- Wala Al Arashi
- Department of Pediatric Hematology and OncologyErasmus MC Sophia Children's Hospital, University Medical Center RotterdamRotterdamThe Netherlands
| | - Michael E. Cloesmeijer
- Department of Hospital Pharmacy & Clinical PharmacologyAmsterdam UMC–location AMCThe Netherlands
| | - Frank W. G. Leebeek
- Department of Hematology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Johannes J. Duvekot
- Department of Obstetrics and Gynecology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Marieke J. H. A. Kruip
- Department of Hematology, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Ron A. A. Mathôt
- Department of Hospital Pharmacy & Clinical PharmacologyAmsterdam UMC–location AMCThe Netherlands
| | - Marjon H. Cnossen
- Department of Pediatric Hematology and OncologyErasmus MC Sophia Children's Hospital, University Medical Center RotterdamRotterdamThe Netherlands
| | | |
Collapse
|
14
|
Amaeze O, Isoherranen N, Shum S. The absorption, distribution, metabolism and elimination characteristics of small interfering RNA therapeutics and the opportunity to predict disposition in pregnant women. Drug Metab Dispos 2025; 53:100018. [PMID: 39884813 DOI: 10.1124/dmd.123.001383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/19/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024] Open
Abstract
Small interfering RNA (siRNA) therapeutics represent an emerging class of pharmacotherapy with the potential to address previously hard-to-treat diseases. Currently approved siRNA therapeutics include lipid nanoparticle-encapsulated siRNA and tri-N-acetylated galactosamine-conjugated siRNA. These siRNA therapeutics exhibit distinct pharmacokinetic characteristics and unique absorption, distribution, metabolism, and elimination (ADME) properties. As a new drug modality, limited clinical data are available for siRNA therapeutics in specific populations, including pediatrics, geriatrics, individuals with renal or hepatic impairment, and pregnant women, making dosing challenging. In this Minireview, a mechanistic overview of the ADME properties of the 5 currently approved siRNA therapeutics is presented. A concise overview of the clinical data available for therapeutic siRNAs in special populations, focusing on the potential impact of physiologic changes during pregnancy on siRNA disposition, is provided. The utility of physiologically based pharmacokinetic (PBPK) modeling as a tool to elucidate the characteristics and disposition of siRNA therapeutics in pregnant women is explored. Additionally, opportunities to integrate known physiologic alterations induced by pregnancy into PBPK models that incorporate siRNA ADME mechanisms to predict the effects of pregnancy on siRNA disposition are discussed. Clinical data regarding the use of therapeutic siRNA in special populations remain limited. Data for precise parameterization of maternal-fetal siRNA PBPK models are lacking presently and underscore the need for further research in this area. Addressing this gap in knowledge will not only enhance our understanding of siRNA pharmacokinetics during pregnancy but also advance the possible development of siRNA therapeutics to treat pregnancy-related conditions. SIGNIFICANCE STATEMENT: This Minireview proposes a framework on how small interfering RNA (siRNA) disposition can be predicted in pregnancy based on mechanistic absorption, distribution, metabolism, and elimination (ADME) information using physiologically-based pharmacokinetic (PBPK) modeling. The mechanistic ADME information and available clinical data in special populations of currently Food and Drug Administration-approved siRNA therapeutics are summarized. Additionally, how physiological changes during pregnancy may affect siRNA disposition is reviewed, and the opportunities to use PBPK modeling to predict siRNA disposition in pregnant women is explored.
Collapse
Affiliation(s)
- Ogochukwu Amaeze
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Sara Shum
- ReNAgade Therapeutics Management Inc, Cambridge, Massachusetts.
| |
Collapse
|
15
|
Koldeweij C, Dibbets C, Franklin BD, Scheepers HCJ, de Wildt SN. A User-Driven Framework for Dose Selection in Pregnancy: Proof of Concept for Sertraline. Clin Pharmacol Ther 2025; 117:214-224. [PMID: 39248386 PMCID: PMC11652817 DOI: 10.1002/cpt.3429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024]
Abstract
Despite growing knowledge of pregnancy-induced changes in physiology that may alter maternal and fetal pharmacokinetics, evidence-based antenatal doses are lacking for most drugs. Pharmacokinetic modeling and expanding clinical data in pregnancy may support antenatal doses. We aimed to develop and pilot a comprehensive and user-driven Framework for Dose Selection in Pregnancy to support the clinical implementation of a best-evidence antenatal dose for sertraline. After initial development and evaluation by experts, the framework prototype was piloted to formulate an antenatal dosing strategy for sertraline in depression and anxiety disorders. Next, the framework was reviewed and assessed for usability by a multidisciplinary working committee of end-users comprising healthcare practitioners, experts from other disciplines including pharmacometrics, reproductive toxicology and medical ethics, alongside pregnant women and a partner. The resulting framework encompasses the following: rationale for drug selection, a comprehensive analysis of pharmacokinetic and dose-related efficacy and safety data, and implementation aspects including feasibility and desirability of the recommended antenatal dose based on a structured maternal and fetal benefit-risk assessment. An antenatal dose recommendation for sertraline, as a case study, was formulated using this approach and endorsed for clinical use by the working committee. Future applications of the framework for other drugs can further demonstrate its suitability for developing best evidence, acceptable and clinically feasible antenatal doses.
Collapse
Affiliation(s)
- Charlotte Koldeweij
- Division of Pharmacology and Toxicology, Department of PharmacyRadboud University Medical CenterNijmegenThe Netherlands
| | - Caroline Dibbets
- Division of Pharmacology and Toxicology, Department of PharmacyRadboud University Medical CenterNijmegenThe Netherlands
- Department of Obstetrics and GynaecologyMaastricht University Medical CentreMaastrichtThe Netherlands
| | - Bryony D. Franklin
- Centre for Medication Safety and Service QualityImperial College Healthcare NHS TrustLondonUK
- Department of Practice and PolicyUCL School of PharmacyLondonUK
| | - Hubertina C. J. Scheepers
- Department of Obstetrics and GynaecologyMaastricht University Medical CentreMaastrichtThe Netherlands
- Grow, School for Oncology and ReproductionMaastrichtThe Netherlands
| | - Saskia N. de Wildt
- Division of Pharmacology and Toxicology, Department of PharmacyRadboud University Medical CenterNijmegenThe Netherlands
- Department of Pediatric and Neonatal Intensive CareErasmus MC‐Sophia Children's HospitalRotterdamThe Netherlands
| |
Collapse
|
16
|
Bevers LAH, Jensen RL, Owen A, Colbers A, Carr DF, Burger DM. Genetic variation on dolutegravir pharmacokinetics and relation to safety and efficacy outcomes: a systematic review. Pharmacogenomics 2024; 25:623-635. [PMID: 39697075 DOI: 10.1080/14622416.2024.2441104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Dolutegravir (DTG) is an antiviral agent used for the treatment of HIV, however, there is uncertainty over the influence of genetic variation on DTG exposure, and whether it has clinical implications for the efficacy or toxicity in different populations. This systematic review aims to create an overview of the impact of pharmacogenomics (PGx) on DTG exposure, efficacy, and toxicity. METHODS Publications up to 14 November 2023 were searched and articles were selected on the following criteria: original research articles providing data on people with HIV, data on PGx and either PK or PD or both PD and PGx. RESULTS 711 records were identified, and after screening 10 articles were included. Commonly analyzed genes across the articles were UGT1A1, ABCB1, ABCG2, and NR1I2. The most reported variant associated with PD variability was in SLC22A2, with carriers at higher risk of neuropsychiatric adverse events. CONCLUSIONS This review concludes that while PGx testing may help explain some variability in DTG pharmacokinetics when combined with therapeutic drug monitoring (TDM), current evidence is insufficient to support its routine clinical use. The role of PGx research for DTG remains relevant, especially in specific patient populations where interindividual PK variations are still unexplained.
Collapse
Affiliation(s)
- Lisanne A H Bevers
- Department of Pharmacy, Radboudumc Research Institute for medical Innovation (RIMI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rebecca L Jensen
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - Andrew Owen
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - Angela Colbers
- Department of Pharmacy, Radboudumc Research Institute for medical Innovation (RIMI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daniel F Carr
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, UK
| | - David M Burger
- Department of Pharmacy, Radboudumc Research Institute for medical Innovation (RIMI), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Bobo WV, Moore KM, Betcher HK, Larish AM, Stoppel CM, VandeVoort JL, Chauhan M, Athreya AP, Talati A. The Association of Antidepressants in Late Pregnancy with Postpartum Hemorrhage: Systematic Review of Controlled Observational Studies. J Child Adolesc Psychopharmacol 2024; 34:428-446. [PMID: 39453674 PMCID: PMC11807859 DOI: 10.1089/cap.2024.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Introduction: Despite advances in obstetric care, postpartum hemorrhage (PPH) is a leading cause of maternal mortality worldwide. Prior reviews of studies published through 2016 suggest an association of antidepressant use during late pregnancy and increased risk of PPH. However, a causal link between prenatal antidepressants and PPH remains controversial. Objectives: This systematic literature review aimed to synthesize the empirical evidence on the association of antidepressant exposure in late pregnancy with the risk of PPH, including studies published before and after 2016. Methods: A systematic literature search was conducted using PubMed, OVID Medline, EMBASE, SCOPUS, PsycINFO, and CINAHL from inception to September 9, 2023. Original, peer-reviewed studies (published in English) that reported on the frequency or risk of PPH in women with evidence of antidepressant use during pregnancy and included at least one control group were included. Results: Twenty studies (eight published after 2016) met inclusion criteria, most of which focused on the risks of PPH associated with selective serotonin reuptake inhibitors (SSRIs) or serotonin-norepinephrine reuptake inhibitors (SNRIs). The main findings from the individual studies were mixed, but the majority documented statistically significant associations of PPH with late prenatal exposure, especially for exposures occurring within 30 days of delivery, compared with unexposed deliveries. Fourteen studies addressed underlying antidepressant indications or their correlates. Few studies focused on prenatal antidepressants and the risk of well-defined severe PPH or on antidepressant dose changes and general PPH risk. None examined competing risks of antidepressant discontinuation on mental health outcomes. Conclusions: Late pregnancy exposure to antidepressants may be a minor risk factor for PPH, but it is unclear to what extent reported associations are causal in nature, as opposed to correlational (effects related to nonpharmacological factors including maternal indication). For patients needing antidepressants during pregnancy, current evidence does not favor routinely discontinuing antidepressants specifically to reduce the risk of PPH.
Collapse
Affiliation(s)
- William V. Bobo
- Department of Behavioral Science and Social Medicine, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Katherine M. Moore
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Hannah K. Betcher
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Alyssa M. Larish
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | - Cynthis M. Stoppel
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - Mohit Chauhan
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | - Arjun P. Athreya
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ardesheer Talati
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
18
|
Koldeweij C, Jans VA, Waitt C, Greupink R, Auweele KLV, Franklin BD, Scheepers HC, de Wildt SN. Striving for balance in decisions on antenatal pharmacotherapy. Lancet 2024; 404:1779-1782. [PMID: 39488410 DOI: 10.1016/s0140-6736(24)02069-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 11/04/2024]
Abstract
Most individuals use medication during pregnancy. However, decision making on antenatal pharmacotherapy presents considerable ethical and scientific challenges. Amid a sociocultural paradigm prioritising the elimination of fetal risks, available evidence and guidance are limited, and current decision making on antenatal drugs mostly proceeds in an ad-hoc and, often, biased manner. This approach might undermine the health of both mother and child. The need for a systematic approach towards antenatal drug decisions is becoming even more pressing with the growing knowledge of pregnancy-induced changes in drug disposition and effects. With this new complexity, pregnancy-specific doses might be necessary, potentially altering the balance between maternal and fetal benefits and risks. In this Viewpoint, we argue that ethical principles and a pregnant individual's values must be integrated alongside existing evidence when making decisions on antenatal drug use and dosing. We use the example of sertraline to outline practical strategies for achieving this goal. This approach is urgently needed to foster better-informed and balanced decisions on antenatal pharmacotherapy.
Collapse
Affiliation(s)
- Charlotte Koldeweij
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Verna Aam Jans
- IQ Health, Radboud University Medical Center, Nijmegen, Netherlands
| | - Catriona Waitt
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK; Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Rick Greupink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Bryony D Franklin
- Centre for Medication Safety and Service Quality, Imperial College Healthcare NHS Trust, London, UK; Department of Practice and Policy, UCL School of Pharmacy, London, UK
| | - Hubertina Cj Scheepers
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands; Grow, Institute for Oncology and Reproduction, Maastricht, Netherlands
| | - Saskia N de Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands; Department of Intensive Care, Radboud University Medical Center, Nijmegen, Netherlands; Department of Pediatric and Neonatal Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands.
| |
Collapse
|
19
|
Glättli SC, Elzinga FA, van der Bijl W, Leuvenink HGD, Prins JR, van Goor H, Gordijn SJ, Olinga P, Touw DJ, Mian P. Variability in perfusion conditions and set-up parameters used in ex vivo human placenta models: A literature review. Placenta 2024; 157:37-49. [PMID: 38570213 DOI: 10.1016/j.placenta.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
The ex vivo human placenta perfusion model has proven to be clinically relevant to study transfer- and fetal exposure of various drugs. Although the method has existed for a long period, the setup of the perfusion model has not been generalized yet. This review aims to summarize the setups of ex vivo placental perfusion models used to examine drug transfer across the placenta to identify generalized properties and differences across setups. A literature search was carried out in PubMed September 26, 2022. Studies were labeled as relevant when information was reported, between 2000 and 2022, on the setups of ex vivo placental perfusion models used to study drug transfer across the placenta. The placenta perfusion process, and the data extraction, was divided into phases of preparation, control, drug, and experimental reflecting the chronological timeline of the different phases during the entire placental perfusion process. 135 studies describing an ex vivo human placental perfusion experiment were included. Among included studies, the majority (78.5%) analyzed drug perfusion in maternal to fetal direction, 18% evaluated bi-directional drug perfusion, 3% under equilibrium conditions, and one study investigated drug perfusion in fetal to maternal direction. This literature review facilitates the comparison of studies that employ similar placenta perfusion protocols for drug transfer studies and reveals significant disparities in the setup of these ex vivo placental perfusion models. Due to interlaboratory variability, perfusion studies are not readily comparable or interchangeable. Therefore, a stepwise protocol with multiple checkpoints for validating placental perfusion is needed.
Collapse
Affiliation(s)
- S C Glättli
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - F A Elzinga
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - W van der Bijl
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - H G D Leuvenink
- Department of Surgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - J R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - H van Goor
- Department of Pathology and Medical Biology, Pathology Section, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - S J Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - P Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deunsinglaan 1, 9713 AV, Groningen, the Netherlands
| | - D J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands; Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deunsinglaan 1, 9713 AV, Groningen, the Netherlands
| | - P Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.
| |
Collapse
|
20
|
Jiang H, Guo J, Li J, Li C, Du W, Canavese F, Xie F, Li H, Yang J, Ying H, Hua J. Do birth outcomes mediate the association between drug use in pregnancy and neonatal metabolic bone disease? A prospective cohort study of 10,801 Chinese women. Front Public Health 2024; 12:1377070. [PMID: 39403442 PMCID: PMC11472830 DOI: 10.3389/fpubh.2024.1377070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/06/2024] [Indexed: 10/30/2024] Open
Abstract
Background Prenatal drug use may cause toxicity to bone health in newborns. We aimed to examine whether birth outcomes mediate the association between medication use and neonatal metabolic bone disease (MBD). Methods A prospective cohort of 10,801 pregnant women (17-49 years) and their infants followed at a single center from 1 January 2012 to 31 December 2021 were included. Based on four single drugs, comprehensive medication use was determined and categorized into three groups using latent-class analysis: group 1 included antibiotics and furosemide or less than two drugs except for MgSO4; group 2 included MgSO4 without antibiotics or furosemide; and group 3 encompassed dexamethasone and antibiotics. Mediation analysis was conducted to assess the mediating effects of prematurity, low birth weight (LBW), and small for gestational age (SGA). Results There were 138 (1.3%) infants with MBD; 2,701 (25%) were born preterm, 1717 (15.9%) had LBW, and 303 (2.8%) were SGA. Pregnant women in groups 2 and 3 were 2.52 to 14.66 times more likely to deliver an infant with MBD than those in group 1. Only LBW showed a significant mediating effect on the association between comprehensive medication use and MBD, with a mediation proportion of 51.8% (45.0-64.1%, p < 0.001). Conclusion Comprehensive medication use during pregnancy was associated with an increased risk of neonatal MBD, largely mediated by LBW. Early antepartum monitoring and prevention targeting adverse birth outcomes are necessary to mitigate the risk of MBD.
Collapse
Affiliation(s)
- Honglin Jiang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jialin Guo
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Li
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunlin Li
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenchong Du
- NTU Psychology, Nottingham Trent University, Nottingham, United Kingdom
| | - Federico Canavese
- CHU Lille, Department of Pediatric Orthopedic Surgery, Lille University, Lille, France
| | - Feng Xie
- Department of Orthopedics and Sports Medicine, Shanghai United Family Hospital, Shanghai, China
| | - Huajing Li
- Aigora Technology PTE. LTD, TRIVEX, Singapore, Singapore
| | - Jian Yang
- Aigora Technology PTE. LTD, TRIVEX, Singapore, Singapore
| | - Hao Ying
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Hua
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Costa B, Gouveia MJ, Vale N. PBPK Modeling of Lamotrigine and Efavirenz during Pregnancy: Implications for Personalized Dosing and Drug-Drug Interaction Management. Pharmaceutics 2024; 16:1163. [PMID: 39339201 PMCID: PMC11435310 DOI: 10.3390/pharmaceutics16091163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to model the pharmacokinetics of lamotrigine (LTG) and efavirenz (EFV) in pregnant women using physiologically based pharmacokinetic (PBPK) and pregnancy-specific PBPK (p-PBPK) models. For lamotrigine, the adult PBPK model demonstrated accurate predictions for pharmacokinetic parameters. Predictions for the area under the curve (AUC) and peak plasma concentration (Cmax) generally agreed well with observed values. During pregnancy, the PBPK model accurately predicted AUC and Cmax with a prediction error (%PE) of less than 25%. The evaluation of the EFV PBPK model revealed mixed results. While the model accurately predicted certain parameters for non-pregnant adults, significant discrepancies were observed in predictions for higher doses (600 vs. 400 mg) and pregnant individuals. The model's performance during pregnancy was poor, indicating the need for further refinement to account for genetic polymorphism. Gender differences also influenced EFV pharmacokinetics, with lower exposure levels in females compared to males. These findings highlight the complexity of modeling EFV, in general, but specifically in pregnant populations, and the importance of validating such models for accurate clinical application. The study highlights the importance of tailoring dosing regimens for pregnant individuals to ensure both safety and efficacy, particularly when using combination therapies with UGT substrate drugs. Although drug-drug interactions between LTG and EFV appear minimal, further research is needed to improve predictive models and enhance their accuracy.
Collapse
Affiliation(s)
- Bárbara Costa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055 Porto, Portugal
| | - Maria João Gouveia
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055 Porto, Portugal
- Center for Study in Animal Science of University of Porto (CECA-ICETA UP), Praça Coronel Pacheco 15, 4050-453 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
22
|
Koldeweij CJM, Dibbets AC, Ceulemans M, de Vries LC, Franklin BD, Scheepers HCJ, de Wildt SN. Willingness-to-use and preferences for model-informed antenatal doses: a cross-sectional study among European healthcare practitioners and pregnant women. Front Pharmacol 2024; 15:1403747. [PMID: 39211781 PMCID: PMC11358599 DOI: 10.3389/fphar.2024.1403747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Background: Physiological changes in pregnancy may affect drug safety and efficacy, sometimes requiring dose adjustments. Pregnancy-adjusted doses, however, are missing for most medications. Increasingly, pharmacokinetic models can be used for antenatal dose finding. Given the novelty of this technique and questions regarding dose credibility, the acceptability of model-informed antenatal doses should be explored. Objective: We aimed to assess the willingness-to-use and preferred features for model-informed antenatal doses among healthcare practitioners (HCPs) and pregnant women in European countries. Methods: A cross-sectional, web-based study drawing on two open surveys was performed between 8 September and 30 November 2022. Each survey comprised statements drawn from prior focus groups, associated with Likert-scales. Themes included respondents' information needs, search behaviours along with their willingness-to-use and preferred features for model-informed antenatal doses. The surveys were disseminated through professional societies, pregnancy websites and social media. A descriptive analysis was performed. Results: In total, 608 HCPs from different specialties and 794 pregnant women across 15 countries participated, with 81% of respondents across both groups in the Netherlands or Belgium. Among pregnant women, 31% were medical professionals and 85% used medication during pregnancy. Eighty-three percent of HCPs found current antenatal pharmacotherapy suboptimal and 97% believed that model-informed antenatal doses would enhance the quality of antenatal care. Most HCPs (93%) and pregnant women (75%) would be willing to follow model-informed antenatal doses. Most HCPs desired access to the evidence (88%), including from pharmacokinetic modelling (62%). Most pregnant women (96%) wanted to understand antenatal dosing rationales and to be involved in dosing decisions (97%). Conclusion: The willingness-to-use model-informed antenatal doses is high among HCPs and pregnant women provided that certain information needs are met.
Collapse
Affiliation(s)
- C. J. M. Koldeweij
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - A. C. Dibbets
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - M. Ceulemans
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KULeuven, Belgium
- IQ Health, Radboud University Medical Center, Nijmegen, Netherlands
- L-C&Y, KU Leuven Child and Youth Institute, Leuven, Belgium
| | - L. C. de Vries
- Teratology Information Service, Netherlands Pharmacovigilance Centre Lareb, S’Hertogenbosch, Netherlands
| | - B. D. Franklin
- Centre for Medication Safety and Service Quality, Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Practice and Policy, UCL School of Pharmacy, London, United Kingdom
| | - H. C. J. Scheepers
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
- Grow, School for Oncology and Reproduction, Maastricht, Netherlands
| | - S. N. de Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Paediatric and Neonatal Intensive Care, Erasmus MC-Sophia Children’s Hospital, Rotterdam, Netherlands
| |
Collapse
|
23
|
Barry CJS, Walker VM, Burden C, Havdahl A, Davies NM. Genetic Insights Into Perinatal Outcomes of Maternal Antihypertensive Therapy During Pregnancy. JAMA Netw Open 2024; 7:e2426234. [PMID: 39190310 PMCID: PMC11350473 DOI: 10.1001/jamanetworkopen.2024.26234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/10/2024] [Indexed: 08/28/2024] Open
Abstract
Importance Limited information exists regarding the impact of pharmacotherapy in pregnancy due to ethical concerns of unintended fetal harm. Yet, maternal prescriptive drug use for chronic conditions such as hypertension is common. Objective To investigate potential causal relationships between perturbing maternal genetic variants influencing antihypertensive drug targets and perinatal outcomes among offspring using mendelian randomization (MR). Design, Setting, and Participants This 2-sample MR study used individual-level single-nucleotide variation (SNV) outcome data from mother-father-offspring trios with complete genetic and phenotypic information from the Norwegian Mother, Father and Child Cohort Study (MoBa) and summary-level SNV exposure data from UK Biobank participants sourced from the Integrative Epidemiology Unit OpenGWAS project. Pregnant individuals were recruited across Norway during their routine ultrasonography examination at 18 weeks' gestation between June 1999 and December 2008, and mothers, fathers, and offspring were followed up after birth. Novel genetic instruments for maternal antihypertensive drug targets that act via systolic blood pressure (SBP) were derived from individual-level data analyzed in January 2018. Two-sample multivariable MR analysis of these maternal drug targets and offspring outcomes were performed between January 2023 and April 2024. Exposures Maternal genetic variants associated with drug targets for treatments of hypertension, as specified in the National Health Service dictionary of medicines and devices. Main Outcomes and Measures Offspring outcomes were Apgar score at 1 minute and 5 minutes, offspring developmental score at 6 months, birth length, birth weight z score, gestational age, head circumference, and congenital malformation. Maternal hypertensive disorders of pregnancy were a positive control. Results The MoBa sample contained 29 849 family trios, with a mean (SD) maternal age of 30.2 (18.6) years and a mean (SD) paternal age of 32.8 (13.1) years; 51.1% of offspring were male. Seven independent SNVs were identified as influencing maternal SBP via the antihypertensive drug target instruments. For higher levels of maternal SBP acting through the CACNB2 calcium channel blocker target, the estimated change in gestational age was 3.99 days (95% CI, 0.02-7.96 days) per 10-mm Hg decrease in SBP. There was no evidence of differential risk for measured perinatal outcomes from maternal SBP acting through drug targets for multiple hypertensive subclasses, such as between the ADRB1 β-adrenoceptor-blocking target and risk of congenital malformation (estimated odds ratio, 0.28 [95% CI, 0.02-4.71] per 10-mm Hg decrease in SBP). Maternal and paternal SBP acting through the EDNRA vasodilator antihypertensive target did not have a potential causal effect on birth weight z score, with respective β estimates of 0.71 (95% CI, -0.09 to 1.51) and 0.72 (95% CI, -0.08 to 1.53) per 10-mm Hg decrease in SBP. Conclusions and Relevance The findings provided little evidence to indicate that perturbation of maternal genetic variants for SBP that influence antihypertensive drug targets had potential causal relationships with measures of perinatal development and health within this study. These findings may be triangulated with existing literature to guide physicians and mothers in decisions about antihypertensive use during pregnancy.
Collapse
Affiliation(s)
- Ciarrah-Jane S. Barry
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Venexia M. Walker
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Christy Burden
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Alexandra Havdahl
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Nic Waals Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- Center for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- PROMENTA, Department of Psychology, University of Oslo, Oslo, Norway
| | - Neil M. Davies
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Division of Psychiatry, University College London, London, United Kingdom
- Department of Statistical Science, University College London, London, United Kingdom
| |
Collapse
|
24
|
Li D, Franco S, Pennell PB. The impact of pregnancy-related hormonal and physiological changes on antiseizure medications: expert perspective. Expert Rev Clin Pharmacol 2024; 17:655-663. [PMID: 38748860 DOI: 10.1080/17512433.2024.2356617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Epilepsy is a disorder of recurrent, unprovoked seizures affecting approximately 15 million individuals of childbearing potential worldwide. Patients with epilepsy rely on regular daily therapy with antiseizure medications (ASMs). Furthermore, ASMs are also prescribed for other neuropsychiatric indications (e.g. bipolar disorder, pain, migraines) with over 2% of the pregnancies in the United States involving prenatal exposure to ASMs. AREAS COVERED ASM concentrations are affected by hormonal and physiological changes in pregnancy, including increases in renal and hepatic blood flow, decreased protein binding, and changes in enzyme activity. Clearance changes typically reverse within a few weeks after delivery. During pregnancy, many ASMs, such as lamotrigine, levetiracetam, and oxcarbazepine, should have serum concentrations monitored and doses increased to maintain the individualized target range for seizure control. ASMs metabolized via glucuronidation, primarily lamotrigine, undergo marked increases in clearance throughout pregnancy, requiring about 3-fold the pre-pregnancy daily dose by delivery. Postpartum, ASM doses are usually decreased over several weeks to prevent drug toxicity. EXPERT OPINION In the future, the development of a physiologically-based pharmacokinetic model for various ASMs may enable empiric dose adjustments in pregnancy without the difficulties of frequent therapeutic drug monitoring.
Collapse
Affiliation(s)
- Denise Li
- Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Susannah Franco
- Department of Pharmacy, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Page B Pennell
- Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Fixler JS. Surgical-site infection in multifetal cesarean delivery. Arch Gynecol Obstet 2024; 310:1049-1053. [PMID: 38448709 DOI: 10.1007/s00404-024-07384-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/07/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE The relationship between multifetal cesarean delivery and surgical-site infection (SSI) is unclear. If SSI is more common in multifetal cesareans, adjustment of practices such as antibiotic dosing could be warranted. The purpose of this study was to determine whether patients undergoing multifetal cesarean delivery are more likely to experience SSI than those undergoing singleton cesarean delivery. METHODS This was a retrospective cohort study including all cesarean deliveries at a tertiary hospital from 10/1/2009 to 12/28/2018. The primary outcome was rate of SSI in women after multifetal cesarean delivery as compared to those who underwent singleton cesarean delivery. Univariable analysis and multivariable logistic regression were used to assess independent clinical factors associated with SSI in multifetal cesarean deliveries. RESULTS 34,340 women underwent cesarean delivery during this period. 33,211 were singletons (96.7%), and 1,129 were multifetal (3.3%). There was no difference in the rate of SSI in multifetal gestations (15/1,129, 1.3%) as compared to singletons (493/33,211, 1.5%) (p = 0.670, OR 0.89 [95% CI 0.53, 1.50], aOR 1.06 [95% CI 0.61, 1.84]). Limiting analysis to multifetal deliveries, prolonged rupture of membranes (p < 0.004, OR 5.43 [95% CI 1.49, 19.88]), labor augmentation (p < 0.001, OR 15.84 [1.74, 144.53]), and chorioamnionitis (p < 0.001, OR 15.43 [95% CI 3.11, 76.62]) were more common in women with SSI. DISCUSSION SSI is not increased in multifetal cesarean delivery as compared to singleton cesarean delivery. In multifetal cesareans, chorioamnionitis, prolonged rupture of membranes, and labor augmentation were associated with increased odds of SSI.
Collapse
Affiliation(s)
- Joseph S Fixler
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St, Little Rock, AR, 72205, USA.
| |
Collapse
|
26
|
Wu YE, Zheng YY, Li QY, Yao BF, Cao J, Liu HX, Hao GX, van den Anker J, Zheng Y, Zhao W. Model-informed drug development in pediatric, pregnancy and geriatric drug development: States of the art and future. Adv Drug Deliv Rev 2024; 211:115364. [PMID: 38936664 DOI: 10.1016/j.addr.2024.115364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 06/09/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
The challenges of drug development in pediatric, pregnant and geriatric populations are a worldwide concern shared by regulatory authorities, pharmaceutical companies, and healthcare professionals. Model-informed drug development (MIDD) can integrate and quantify real-world data of physiology, pharmacology, and disease processes by using modeling and simulation techniques to facilitate decision-making in drug development. In this article, we reviewed current MIDD policy updates, reflected on the integrity of physiological data used for MIDD and the effects of physiological changes on the drug PK, as well as summarized current MIDD strategies and applications, so as to present the state of the art of MIDD in pediatric, pregnant and geriatric populations. Some considerations are put forth for the future improvements of MIDD including refining regulatory considerations, improving the integrity of physiological data, applying the emerging technologies, and exploring the application of MIDD in new therapies like gene therapies for special populations.
Collapse
Affiliation(s)
- Yue-E Wu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuan-Yuan Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiu-Yue Li
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Cao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui-Xin Liu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Medical Center, Washington, DC, USA; Departments of Pediatrics, Pharmacology & Physiology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA; Department of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Basel, Switzerland
| | - Yi Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
27
|
Costa B, Vale N. Advances in Psychotropic Treatment for Pregnant Women: Efficacy, Adverse Outcomes, and Therapeutic Monitoring. J Clin Med 2024; 13:4398. [PMID: 39124665 PMCID: PMC11312735 DOI: 10.3390/jcm13154398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Advancements in psychotropic therapy for pregnant women are pivotal for addressing maternal mental health during the perinatal period. Screening for mood and anxiety symptoms during pregnancy is recommended to enable early intervention. Psychotropic medications, including antidepressants, benzodiazepines, antipsychotics, and mood stabilizers, are commonly used, but challenges remain regarding their safety and efficacy during pregnancy. Pregnancy induces significant changes in pharmacokinetics, necessitating personalized dosing strategies and careful monitoring. Real-time monitoring technologies, such as smartphone-integrated platforms and home-based monitoring, enhance accessibility and accuracy. Prospective studies and collaboration among healthcare providers are essential for evidence-based guidelines and optimal treatment strategies. Reducing stigma around mental health during pregnancy is crucial to ensure women seek help and discuss treatment options, promoting understanding and acceptance within the community.
Collapse
Affiliation(s)
- Bárbara Costa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
28
|
Costa B, Gouveia MJ, Vale N. Safety and Efficacy of Antiviral Drugs and Vaccines in Pregnant Women: Insights from Physiologically Based Pharmacokinetic Modeling and Integration of Viral Infection Dynamics. Vaccines (Basel) 2024; 12:782. [PMID: 39066420 PMCID: PMC11281481 DOI: 10.3390/vaccines12070782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Addressing the complexities of managing viral infections during pregnancy is essential for informed medical decision-making. This comprehensive review delves into the management of key viral infections impacting pregnant women, namely Human Immunodeficiency Virus (HIV), Hepatitis B Virus/Hepatitis C Virus (HBV/HCV), Influenza, Cytomegalovirus (CMV), and SARS-CoV-2 (COVID-19). We evaluate the safety and efficacy profiles of antiviral treatments for each infection, while also exploring innovative avenues such as gene vaccines and their potential in mitigating viral threats during pregnancy. Additionally, the review examines strategies to overcome challenges, encompassing prophylactic and therapeutic vaccine research, regulatory considerations, and safety protocols. Utilizing advanced methodologies, including PBPK modeling, machine learning, artificial intelligence, and causal inference, we can amplify our comprehension and decision-making capabilities in this intricate domain. This narrative review aims to shed light on diverse approaches and ongoing advancements, this review aims to foster progress in antiviral therapy for pregnant women, improving maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Bárbara Costa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, 4000-055 Porto, Portugal;
| | - Maria João Gouveia
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, 4000-055 Porto, Portugal;
- Center for the Study in Animal Science (CECA/ICETA), University of Porto, 4051-401 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
29
|
Schoretsanitis G, Deligiannidis KM, Kasperk N, Schmidt CT, Kittel-Schneider S, Ter Horst P, Berlin M, Kohn E, Poels EMP, Zutshi D, Tomson T, Spigset O, Paulzen M. The impact of pregnancy on the pharmacokinetics of antiseizure medications: A systematic review and meta-analysis of data from 674 pregnancies. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111030. [PMID: 38762161 DOI: 10.1016/j.pnpbp.2024.111030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/04/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
OBJECTIVE Increasing evidence suggests that the physiological changes of pregnancy may impact pharmacokinetics of antiseizure medications (ASM), and this may affect treatment outcomes. The aim of this study was to quantify the pregnancy impact on the ASM pharmacokinetics. METHODS A systematic literature search was conducted in PubMed/EMBASE in November 2022 and updated in August 2023 for studies comparing levels of ASM in the same individuals during pregnancy and in the preconception/postpartum period. Alteration ratios between the 3rd trimester and baseline were estimated. We also performed a random-effects meta-analysis calculating between-timepoint differences in mean differences (MDs) and 95% confidence intervals (95%CIs) for dose-adjusted plasma concentrations (C/D ratios). Study quality was assessed using the ClinPK guidelines. RESULTS A total of 65 studies investigating 15 ASMs in 674 pregnancies were included. The largest differences were reported for lamotrigine, oxcarbazepine and levetiracetam (alteration ratio 0.42, range 0.07-2.45, 0.42, range 0.08-0.82 and 0.52, range 0.04-2.77 respectively): accordingly, C/D levels were lower in the 3rd trimester for lamotrigine, levetiracetam and the main oxcarbazepine metabolite monohydroxycarbazepine (MD = -12.33 × 10-3, 95%CI = -16.08 to -8.58 × 10-3 (μg/mL)/(mg/day), p < 0.001, MD = -7.16 (μg/mL)/(mg/day), 95%CI = -9.96 to -4.36, p < 0.001, and MD = -4.87 (μg/mL)/(mg/day), 95%CI = -9.39 to -0.35, p = 0.035, respectively), but not for oxcarbazepine (MD = 1.16 × 10-3 (μg/mL)/(mg/day), 95%CI = -2.55 to 0.24 × 10-3, p = 0.10). The quality of studies was acceptable with an average rating score of 11.5. CONCLUSIONS Data for lamotrigine, oxcarbazepine (and monohydroxycarbazepine) and levetiracetam demonstrate major changes in pharmacokinetics during pregnancy, suggesting the importance of therapeutic drug monitoring to assist clinicians in optimizing treatment outcomes.
Collapse
Affiliation(s)
- Georgios Schoretsanitis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Hospital of Psychiatry, University of Zurich, Zurich, Switzerland; The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, NY, USA; Department of Psychiatry at the Donald and Barbara Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY, USA.
| | - Kristina M Deligiannidis
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, NY, USA; The Departments of Obstetrics & Gynecology and Molecular Medicine at the Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Department of Psychiatry at the Donald and Barbara Zucker School of Medicine at Northwell/Hofstra, Hempstead, NY, USA.
| | - Nicholas Kasperk
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, and JARA - Translational Brain Medicine, Aachen, Germany.
| | - Chiara Theresa Schmidt
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, and JARA - Translational Brain Medicine, Aachen, Germany.
| | - Sarah Kittel-Schneider
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Acute Mental Health Unit, Cork University Hospital, Wilton, Cork, Ireland.
| | - Peter Ter Horst
- Department of Clinical Pharmacy, Isala Medical Centre, Dokter van Heesweg 2, 8025 AB Zwolle, the Netherlands.
| | - Maya Berlin
- Clinical Pharmacology and Toxicology Unit, Shamir (Assaf Harofeh) Medical Center, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| | - Elkana Kohn
- Clinical Pharmacology and Toxicology Unit, Shamir (Assaf Harofeh) Medical Center, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| | - Eline M P Poels
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Deepti Zutshi
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Torbjörn Tomson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Olav Spigset
- Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Michael Paulzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, and JARA - Translational Brain Medicine, Aachen, Germany; Alexianer Hospital Aachen, Aachen, Germany.
| |
Collapse
|
30
|
Collins HE, Alexander BT, Care AS, Davenport MH, Davidge ST, Eghbali M, Giussani DA, Hoes MF, Julian CG, LaVoie HA, Olfert IM, Ozanne SE, Bytautiene Prewit E, Warrington JP, Zhang L, Goulopoulou S. Guidelines for assessing maternal cardiovascular physiology during pregnancy and postpartum. Am J Physiol Heart Circ Physiol 2024; 327:H191-H220. [PMID: 38758127 PMCID: PMC11380979 DOI: 10.1152/ajpheart.00055.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Maternal mortality rates are at an all-time high across the world and are set to increase in subsequent years. Cardiovascular disease is the leading cause of death during pregnancy and postpartum, especially in the United States. Therefore, understanding the physiological changes in the cardiovascular system during normal pregnancy is necessary to understand disease-related pathology. Significant systemic and cardiovascular physiological changes occur during pregnancy that are essential for supporting the maternal-fetal dyad. The physiological impact of pregnancy on the cardiovascular system has been examined in both experimental animal models and in humans. However, there is a continued need in this field of study to provide increased rigor and reproducibility. Therefore, these guidelines aim to provide information regarding best practices and recommendations to accurately and rigorously measure cardiovascular physiology during normal and cardiovascular disease-complicated pregnancies in human and animal models.
Collapse
Grants
- HL169157 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HD088590 NICHD NIH HHS
- HD083132 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- The Biotechnology and Biological Sciences Research Council
- P20GM103499 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- British Heart Foundation (BHF)
- R21 HD111908 NICHD NIH HHS
- Distinguished University Professor
- The Lister Insititute
- ES032920 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL149608 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Royal Society (The Royal Society)
- U.S. Department of Defense (DOD)
- HL138181 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- MC_00014/4 UKRI | Medical Research Council (MRC)
- RG/17/8/32924 British Heart Foundation
- Jewish Heritage Fund for Excellence
- HD111908 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL163003 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- APP2002129 NHMRC Ideas Grant
- HL159865 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL131182 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL163818 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NS103017 HHS | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- HL143459 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL146562 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL138181 NHLBI NIH HHS
- 20CSA35320107 American Heart Association (AHA)
- RG/17/12/33167 British Heart Foundation (BHF)
- National Heart Foundation Future Leader Fellowship
- P20GM121334 HHS | NIH | National Institute of General Medical Sciences (NIGMS)
- HL146562-04S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL155295 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HD088590-06 HHS | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- HL147844 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- WVU SOM Synergy Grant
- R01 HL146562 NHLBI NIH HHS
- R01 HL159865 NHLBI NIH HHS
- Canadian Insitute's of Health Research Foundation Grant
- R01 HL169157 NHLBI NIH HHS
- HL159447 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- ES034646-01 HHS | NIH | National Institute of Environmental Health Sciences (NIEHS)
- HL150472 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 2021T017 Dutch Heart Foundation Dekker Grant
- MC_UU_00014/4 Medical Research Council
- R01 HL163003 NHLBI NIH HHS
- Christenson professor In Active Healthy Living
- National Heart Foundation
- Dutch Heart Foundation Dekker
- WVU SOM Synergy
- Jewish Heritage
- Department of Health | National Health and Medical Research Council (NHMRC)
- Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de recherche en santé du Canada)
Collapse
Affiliation(s)
- Helen E Collins
- University of Louisville, Louisville, Kentucky, United States
| | - Barbara T Alexander
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Alison S Care
- University of Adelaide, Adelaide, South Australia, Australia
| | | | | | - Mansoureh Eghbali
- University of California Los Angeles, Los Angeles, California, United States
| | | | | | - Colleen G Julian
- University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Holly A LaVoie
- University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - I Mark Olfert
- West Virginia University School of Medicine, Morgantown, West Virginia, United States
| | | | | | - Junie P Warrington
- University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Lubo Zhang
- Loma Linda University School of Medicine, Loma Linda, California, United States
| | | |
Collapse
|
31
|
Wanninayake S, Ochoa‐Ferraro A, Patel K, Ramachandran R, Wierzbicki AS, Dawson C. Two successful pregnancies -in patients taking Volanesorsen for familial chylomicronemia syndrome. JIMD Rep 2024; 65:249-254. [PMID: 38974616 PMCID: PMC11224504 DOI: 10.1002/jmd2.12435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 07/09/2024] Open
Abstract
Familial chylomicronemia syndrome (FCS) is a rare inherited disorder characterized by severe hypertriglyceridemia, posing a heightened risk of acute pancreatitis. Recently, Volanesorsen, an APOC3 antisense oligonucleotide, gained approval for FCS treatment in the UK. Caution is advised during pregnancy due to limited safety data, although animal studies show no toxicity/teratogenicity. Two case scenarios are presented: In the first case, a patient with FCS continued Volanesorsen injections without having thrombocytopenia during an unplanned pregnancy until third trimester, maintaining triglyceride control. Upon discovering the pregnancy at 38 weeks, Volanesorsen was ceased, and a low-fat diet reinstated. Despite a heightened risk of pancreatitis, no episodes of pancreatitis occurred during the pregnancy. In the second case, stopping Volanesorsen before conception led to elevated triglycerides, and an episode of acute pancreatitis at 22 weeks, despite strict very low-fat diet and fibrate therapy from 14 weeks. At 23 weeks, Volanesorsen was reintroduced concurrently with regular therapeutic plasma exchange. No further episodes of pancreatitis occurred. In both case, fetal health was maintained throughout pregnancy, fetal scans revealed no anomalies, and planned C-sections delivered healthy babies without congenital issues. Both babies are well and developing normally at 24 and 19 months.
Collapse
Affiliation(s)
- Subadra Wanninayake
- Department of Diabetes, Endocrinology and MetabolismUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Antonio Ochoa‐Ferraro
- Department of Diabetes, Endocrinology and MetabolismUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | | | - Radha Ramachandran
- Department of Adult Inherited Metabolic Diseases, Metabolic Medicine and Chemical PathologyGuys and St Thomas' Hospitals NHS Foundation TrustLondonUK
| | - Anthony S. Wierzbicki
- Department of Adult Inherited Metabolic Diseases, Metabolic Medicine and Chemical PathologyGuys and St Thomas' Hospitals NHS Foundation TrustLondonUK
| | - Charlotte Dawson
- Department of Diabetes, Endocrinology and MetabolismUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| |
Collapse
|
32
|
Dai W, Piquette-Miller M. Altered Expression of BCRP Impacts Fetal Accumulation of Rosuvastatin in a Rat Model of Preeclampsia. Pharmaceutics 2024; 16:884. [PMID: 39065581 PMCID: PMC11279450 DOI: 10.3390/pharmaceutics16070884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/14/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Expression of the breast cancer resistance protein (BCRP/ABCG2) transporter is downregulated in placentas from women with preeclampsia (PE) and in an immunological rat model of PE. While many drugs are substrates of this important efflux transporter, the impact of PE associated BCRP downregulation on maternal and fetal drug exposure has not been investigated. Using the PE rat model, we performed a pharmacokinetic study with rosuvastatin (RSV), a BCRP substrate, to investigate this impact. PE was induced in rats during gestational days (GD) 13 to 16 with daily low-dose endotoxin. On GD18, RSV (3 mg/kg) was administrated intravenously, and rats were sacrificed at time intervals between 0.5 and 6 h. As compared to controls, placental expression of Bcrp and Oatp2b1 significantly decreased in PE rats. A corresponding increase in RSV levels was seen in fetal tissues and amniotic fluid of the PE group (p < 0.05), while maternal plasma concentrations remained unchanged from the controls. An increase in Bcrp expression and decreased RSV concentration were seen in the livers of PE dams. This suggests that PE-mediated transporter dysregulation leads to significant changes in the maternal and fetal RSV disposition. Overall, our findings demonstrate that altered placental expression of transporters in PE can increase fetal accumulation of their substrates.
Collapse
|
33
|
Koldeweij C, Kleuskens M, Litjens C, Franklin BD, Scheepers HCJ, de Wildt SN. Perceived barriers and facilitators for model-informed dosing in pregnancy: a qualitative study across healthcare practitioners and pregnant women. BMC Med 2024; 22:248. [PMID: 38886762 PMCID: PMC11184760 DOI: 10.1186/s12916-024-03450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/28/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Most women use medication during pregnancy. Pregnancy-induced changes in physiology may require antenatal dose alterations. Yet, evidence-based doses in pregnancy are missing. Given historically limited data, pharmacokinetic models may inform pregnancy-adjusted doses. However, implementing model-informed doses in clinical practice requires support from relevant stakeholders. PURPOSE To explore the perceived barriers and facilitators for model-informed antenatal doses among healthcare practitioners (HCPs) and pregnant women. METHODS Online focus groups and interviews were held among healthcare practitioners (HCPs) and pregnant women from eight countries across Europe, Africa and Asia. Purposive sampling was used to identify pregnant women plus HCPs across various specialties prescribing or providing advice on medication to pregnant women. Perceived barriers and facilitators for implementing model-informed doses in pregnancy were identified and categorised using a hybrid thematic analysis. RESULTS Fifty HCPs and 11 pregnant women participated in 12 focus groups and 16 interviews between January 2022 and March 2023. HCPs worked in the Netherlands (n = 32), the UK (n = 7), South Africa (n = 5), Uganda (n = 4), Kenya, Cameroon, India and Vietnam (n = 1 each). All pregnant women resided in the Netherlands. Barriers and facilitators identified by HCPs spanned 14 categories across four domains whereas pregnant women described barriers and facilitators spanning nine categories within the same domains. Most participants found current antenatal dosing information inadequate and regarded model-informed doses in pregnancy as a valuable and for some, much-needed addition to antenatal care. Although willingness-to-follow model-informed antenatal doses was high across both groups, several barriers for implementation were identified. HCPs underlined the need for transparent model validation and endorsement of the methodology by recognised institutions. Foetal safety was deemed a critical knowledge gap by both groups. HCPs' information needs and preferred features for model-informed doses in pregnancy varied. Several pregnant women expressed a desire to access information and partake in decisions on antenatal dosing. CONCLUSIONS Given the perceived limitations of current pharmacotherapy for pregnant women and foetuses, model-informed dosing in pregnancy was seen as a promising means to enhance antenatal care by pregnant women and healthcare practitioners.
Collapse
Affiliation(s)
- Charlotte Koldeweij
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Mirèse Kleuskens
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carlijn Litjens
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| | - Bryony Dean Franklin
- Centre for Medication Safety and Service Quality, Imperial College Healthcare NHS Trust, London, UK
- Department of Practice and Policy, UCL School of Pharmacy, London, UK
| | - Hubertina C J Scheepers
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, The Netherlands
- Grow, School for Oncology and Reproduction, Maastricht, The Netherlands
| | - Saskia N de Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pediatric and Neonatal Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
34
|
Kotta-Loizou I, Pritsa A, Antasouras G, Vasilopoulos SN, Voulgaridou G, Papadopoulou SK, Coutts RHA, Lechouritis E, Giaginis C. Fetus Exposure to Drugs and Chemicals: A Holistic Overview on the Assessment of Their Transport and Metabolism across the Human Placental Barrier. Diseases 2024; 12:114. [PMID: 38920546 PMCID: PMC11202568 DOI: 10.3390/diseases12060114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The placenta exerts a crucial role in fetus growth and development during gestation, protecting the fetus from maternal drugs and chemical exposure. However, diverse drugs and chemicals (xenobiotics) can penetrate the maternal placental barrier, leading to deleterious, adverse effects concerning fetus health. Moreover, placental enzymes can metabolize drugs and chemicals into more toxic compounds for the fetus. Thus, evaluating the molecular mechanisms through which drugs and chemicals transfer and undergo metabolism across the placental barrier is of vital importance. In this aspect, this comprehensive literature review aims to provide a holistic approach by critically summarizing and scrutinizing the potential molecular processes and mechanisms governing drugs and chemical transfer and metabolism across the placental barrier, which may lead to fetotoxicity effects, as well as analyzing the currently available experimental methodologies used to assess xenobiotics placental transfer and metabolism. METHODS A comprehensive and in-depth literature review was conducted in the most accurate scientific databases such as PubMed, Scopus, and Web of Science by using relevant and effective keywords related to xenobiotic placental transfer and metabolism, retrieving 8830 published articles until 5 February 2024. After applying several strict exclusion and inclusion criteria, a final number of 148 relevant published articles were included. RESULTS During pregnancy, several drugs and chemicals can be transferred from the mother to the fetus across the placental barrier by either passive diffusion or through placental transporters, resulting in fetus exposure and potential fetotoxicity effects. Some drugs and chemicals also appear to be metabolized across the placental barrier, leading to more toxic products for both the mother and the fetus. At present, there is increasing research development of diverse experimental methodologies to determine the potential molecular processes and mechanisms of drug and chemical placental transfer and metabolism. All the currently available methodologies have specific strengths and limitations, highlighting the strong demand to utilize an efficient combination of them to obtain reliable evidence concerning drug and chemical transfer and metabolism across the placental barrier. To derive the most consistent and safe evidence, in vitro studies, ex vivo perfusion methods, and in vivo animal and human studies can be applied together with the final aim to minimize potential fetotoxicity effects. CONCLUSIONS Research is being increasingly carried out to obtain an accurate and safe evaluation of drug and chemical transport and metabolism across the placental barrier, applying a combination of advanced techniques to avoid potential fetotoxic effects. The improvement of the currently available techniques and the development of novel experimental protocols and methodologies are of major importance to protect both the mother and the fetus from xenobiotic exposure, as well as to minimize potential fetotoxicity effects.
Collapse
Affiliation(s)
- Ioly Kotta-Loizou
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK;
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Georgios Antasouras
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| | - Spyridon N. Vasilopoulos
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece;
| | - Gavriela Voulgaridou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Sousana K. Papadopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Robert H. A. Coutts
- Department of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK;
| | - Eleftherios Lechouritis
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| |
Collapse
|
35
|
Berezowska M, Sharma P, Pilla Reddy V, Coppola P. Physiologically Based Pharmacokinetic modelling of drugs in pregnancy: A mini-review on availability and limitations. Fundam Clin Pharmacol 2024; 38:402-409. [PMID: 37968879 DOI: 10.1111/fcp.12967] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/14/2023] [Accepted: 10/17/2023] [Indexed: 11/17/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) modelling in pregnancy is a relatively new approach that is increasingly being used to assess drug systemic exposure in pregnant women to potentially inform dosing adjustments. Physiological changes throughout pregnancy are incorporated into mathematical models to simulate drug disposition in the maternal and fetal compartments as well as the transfer of drugs across the placenta. This mini-review gathers currently available pregnancy PBPK models for drugs commonly used during pregnancy. In addition, information about the main PBPK modelling platforms used, metabolism pathways, drug transporters, data availability and drug labels were collected. The aim of this mini-review is to provide a concise overview, demonstrate trends in the field, highlight understudied areas and identify current gaps of PBPK modelling in pregnancy. Possible future applications of this PBPK approach are discussed from a clinical, regulatory and industry perspective.
Collapse
Affiliation(s)
- Monika Berezowska
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Pradeep Sharma
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Venkatesh Pilla Reddy
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Paola Coppola
- Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
36
|
Samimi MN, Hale A, Schults J, Fischer A, Roberts JA, Dhanani J. Clinical guidance for unfractionated heparin dosing and monitoring in critically ill patients. Expert Opin Pharmacother 2024; 25:985-997. [PMID: 38825778 DOI: 10.1080/14656566.2024.2364057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/31/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION Unfractionated heparin is a widely used anticoagulant in critically ill patients. It has a well-established safety profile and remains an attractive option for clinicians due to its short half-life and reversibility. Heparin has a unique pharmacokinetic profile, which contributes to significant inter-patient and intra-patient variability in effect. The variability in anticoagulant effect combined with heparin's short half-life mean close monitoring is required for clinical efficacy and preventing adverse effects. To optimize heparin use in critically ill patients, effective monitoring assays and dose adjustment strategies are needed. AREAS COVERED This paper explores the use of heparin as an anticoagulant and optimal approaches to monitoring in critically ill patients. EXPERT OPINION Conventional monitoring assays for heparin dosing have significant limitations. Emerging data appear to favor using anti-Xa assay monitoring for heparin anticoagulation, which many centers have successfully adopted as the standard. The anti-Xa assay appears have important benefits relative to the aPTT for heparin monitoring in critically ill patients, and should be considered for broader use.
Collapse
Affiliation(s)
- May N Samimi
- Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- Department of Pharmacy, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Andrew Hale
- Discipline of Pharmacy, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Jessica Schults
- Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- School of Nursing, Midwifery and Social Work, University of Queensland, Brisbane, Australia
- Herston Infectious Diseases Institute (HeIDI), Metro North Health, Brisbane, Australia
| | - Andreas Fischer
- Pharmacy Department, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Jason A Roberts
- Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- Department of Pharmacy, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Herston Infectious Diseases Institute (HeIDI), Metro North Health, Brisbane, Australia
- Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Jayesh Dhanani
- Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia
- Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
37
|
Sabbag A, Aabel EW, Castrini AI, Siontis KC, Laredo M, Nizard J, Duthoit G, Asirvatham S, Sehrawat O, Kirkels FP, van Rosendael PJ, Beinart R, Acha MR, Peichl P, Lim HS, Sohns C, Martins R, Font J, Truong NNK, Estensen M, Haugaa KH. Mitral valve prolapse: arrhythmic risk during pregnancy and postpartum. Eur Heart J 2024; 45:1831-1839. [PMID: 38740526 PMCID: PMC11129793 DOI: 10.1093/eurheartj/ehae224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/02/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND AIMS Arrhythmic mitral valve prolapse (AMVP) is linked to life-threatening ventricular arrhythmias (VAs), and young women are considered at high risk. Cases of AMVP in women with malignant VA during pregnancy have emerged, but the arrhythmic risk during pregnancy is unknown. The authors aimed to describe features of women with high-risk AMVP who developed malignant VA during the perinatal period and to assess if pregnancy and the postpartum period were associated with a higher risk of malignant VA. METHODS This retrospective international multi-centre case series included high-risk women with AMVP who experienced malignant VA and at least one pregnancy. Malignant VA included ventricular fibrillation, sustained ventricular tachycardia, or appropriate shock from an implantable cardioverter defibrillator. The authors compared the incidence of malignant VA in non-pregnant periods and perinatal period; the latter defined as occurring during pregnancy and within 6 months after delivery. RESULTS The authors included 18 women with AMVP from 11 centres. During 7.5 (interquartile range 5.8-16.6) years of follow-up, 37 malignant VAs occurred, of which 18 were pregnancy related occurring in 13 (72%) unique patients. Pregnancy and 6 months after delivery showed increased incidence rate of malignant VA compared to the non-pregnancy period (univariate incidence rate ratio 2.66, 95% confidence interval 1.23-5.76). CONCLUSIONS The perinatal period could impose increased risk of malignant VA in women with high-risk AMVP. The data may provide general guidance for pre-conception counselling and for nuanced shared decision-making between patients and clinicians.
Collapse
Affiliation(s)
- Avi Sabbag
- Sheba Medical Centre, Ramat-Gan, affiliated with the School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eivind W Aabel
- ProCardio Center for Research Based Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, and University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Anna Isotta Castrini
- ProCardio Center for Research Based Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, and University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | | | - Mikael Laredo
- Sorbonne Université, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jacky Nizard
- Sorbonne Université, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Guillaume Duthoit
- Sorbonne Université, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Samuel Asirvatham
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ojasay Sehrawat
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Feddo P Kirkels
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Roy Beinart
- Sheba Medical Centre, Ramat-Gan, affiliated with the School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Rav Acha
- Jesselson Integrated Heart Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Petr Peichl
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Han S Lim
- Austin and Northern Health, University of Melbourne, Melbourne, Australia
| | - Christian Sohns
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | | | - Jonaz Font
- LTSI, Rennes University Hospital, Rennes, France
| | - Nguyen N K Truong
- Department of Interventional Cardiology, Medical University Center of Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Mette Estensen
- ProCardio Center for Research Based Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, and University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Kristina H Haugaa
- ProCardio Center for Research Based Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, and University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
38
|
Bozdag D, van Voorthuizen J, Korpel N, Lentz S, Gurer-Orhan H, Kamstra JH. Dysregulation of adipogenesis and disrupted lipid metabolism by the antidepressants citalopram and sertraline. Toxicol Appl Pharmacol 2024; 486:116937. [PMID: 38643950 DOI: 10.1016/j.taap.2024.116937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024]
Abstract
Selective Serotonin Reuptake Inhibitors (SSRIs) are widely used medications for the treatment of major depressive disorder. However, long-term SSRI use has been associated with weight gain and altered lipid profiles. These findings suggest that SSRIs may have negative effects on metabolism. Exposure to certain chemicals called 'obesogens' is known to promote lipid accumulation and obesity by modulating adipogenesis. Here, we investigated whether citalopram (CIT) and sertraline (SER) interfere with the process of adipogenesis, using human mesenchymal stem cells (MSCs) in a 2D and a 3D model. Assessment of intracellular lipid accumulation by fluorescence staining was used as a measure for enhanced adipogenesis. To explore possible mechanisms behind SSRIs' effects, receptor mediated activity was studied using responsive cell lines for various nuclear receptors. Furthermore, RNA sequencing was performed in the 3D model, followed by differential gene expression and pathway analysis. A dose dependent increase in lipid accumulation was observed in both models with CIT and SER. For the 3D model, the effect was seen in a range close to reported steady-state plasma concentrations (0.065-0.65 μM for SER and 0.12-0.92 μM for CIT). Pathway analysis revealed unexpected results of downregulation in adipogenesis-related pathways and upregulation in phospholipids and lysosomal pathways. This was confirmed by an observed increase in lysosomes in the 2D model. Our findings suggest lysosomal dysfunction and disrupted lipid metabolism in mature adipocytes, leading to excessive phospholipid synthesis. Moreover, important adipogenic processes are inhibited, potentially leading to dysfunctional adipocytes, which might have implications in the maintenance of a healthy metabolic balance.
Collapse
Affiliation(s)
- Deniz Bozdag
- Faculty of Veterinary Medicine, Department of Population Health Sciences, Institute for Risk Assessment Sciences, Utrecht University, 3584 CM Utrecht, the Netherlands; Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ege University, 35040 Izmir, Turkey.
| | - Jeroen van Voorthuizen
- Faculty of Veterinary Medicine, Department of Population Health Sciences, Institute for Risk Assessment Sciences, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Nikita Korpel
- Faculty of Veterinary Medicine, Department of Population Health Sciences, Institute for Risk Assessment Sciences, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Sander Lentz
- Faculty of Veterinary Medicine, Department of Population Health Sciences, Institute for Risk Assessment Sciences, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Hande Gurer-Orhan
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ege University, 35040 Izmir, Turkey
| | - Jorke H Kamstra
- Faculty of Veterinary Medicine, Department of Population Health Sciences, Institute for Risk Assessment Sciences, Utrecht University, 3584 CM Utrecht, the Netherlands.
| |
Collapse
|
39
|
Ludna Duarte M, Mikaelle Brandão Silva A, Wellithom Viturino da Silva J, Pereira Santana D, Victor de Castro W, Cláudio Arraes de Alencar L, César Galindo Bedor D, Bastos Leal L. A validated method for simultaneous quantification of four antiretrovirals in dried blood spot and plasma using LC-MS/MS: Application to efavirenz therapeutic drug monitoring in pregnant patients. Clin Biochem 2024; 127-128:110765. [PMID: 38649089 DOI: 10.1016/j.clinbiochem.2024.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION The specific physiological background induced by pregnancy leads to significant changes in maternal pharmacokinetics, suggesting potential variability in plasma concentrations of antiretrovirals. Pregnant HIV patients exposed to subtherapeutic doses, particularly in the last trimester of the pregnancy, have higher chances to transmit the infection to their children. Therefore, the therapeutic drug monitoring of antiretrovirals in HIV pregnant patients would be of great value. OBJECTIVES This study aimed to develop and validate a sensitive liquid chromatograph tandem mass spectrometry (LC-MS/MS) method for simultaneous quantification of efavirenz, raltegravir, atazanavir, and ritonavir in dried blood spots (DBS) and plasma. DESIGN AND METHODS The analytes were extracted from the DBS punch and plasma with a mixture of methanol:zinc sulfate 200 mM (50:50, v/v) and 100 % methanol, respectively. For the chromatographic separation a Shim-pack® C18, 4.6 mm × 150 mm, 5 μm column was used. Detection was performed in a 3200-QTRAP® mass spectrometer, with a run time of 6 min. RESULTS The assay was linear in the range of 15-1,000 ng/mL for raltegravir, 50-10,000 ng/mL for both atazanavir and ritonavir, 50-5,000 ng/mL for efavirenz. Precision and accuracy at these concentrations were less than 15 % for all analytes. Raltegravir, atazanavir, and ritonavir were stable for seven days at 23 °C and 40 °C, whereas efavirenz was stable for twenty-four hours at the same conditions. CONCLUSIONS The method was successfully applied to quantify efavirenz in DBS samples obtained from HIV-1 infected pregnant volunteers under antiretroviral therapy. The concentrations of efavirenz in DBS and plasma were comparable according to Passing-Bablok regression and Bland-Altman analysis.
Collapse
Affiliation(s)
- Maira Ludna Duarte
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Campus Recife, S/n, Recife, Pernambuco, CEP 50740-521, Brazil.
| | - Aurylanne Mikaelle Brandão Silva
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Campus Recife, S/n, Recife, Pernambuco, CEP 50740-521, Brazil
| | - José Wellithom Viturino da Silva
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Campus Recife, S/n, Recife, Pernambuco, CEP 50740-521, Brazil
| | - Davi Pereira Santana
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Campus Recife, S/n, Recife, Pernambuco, CEP 50740-521, Brazil.
| | - Whocely Victor de Castro
- Graduate Program in Pharmaceutical Sciences, Federal University of São João del-Rei, Av. Sebastião Gonçalves Coelho 400, Chanadour, Divinópolis, Minas Gerais, CEP 35501-296, Brazil.
| | - Luiz Cláudio Arraes de Alencar
- Department of Tropical Medicine, Federal University of Pernambuco, Campus Recife, S/n, Recife, Pernambuco, CEP 50740-521, Brazil
| | - Danilo César Galindo Bedor
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Campus Recife, S/n, Recife, Pernambuco, CEP 50740-521, Brazil.
| | - Leila Bastos Leal
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Campus Recife, S/n, Recife, Pernambuco, CEP 50740-521, Brazil.
| |
Collapse
|
40
|
Zhang X, Liu X, Liu C, Hua C. Clinical considerations of emergent oral manifestations during pregnancy. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2024; 42:142-153. [PMID: 38597074 PMCID: PMC11034408 DOI: 10.7518/hxkq.2024.2023367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/06/2024] [Indexed: 04/11/2024]
Abstract
Pregnancy is a special period for developing and treating oral diseases. Oral emergencies during pregnancy need to be handled appropriately. Changes in the physiological environments and personal habits of pregnant women increase susceptibility to some oral diseases. However, clinical treatment strategies are limited due to the need to ensure the safety of pregnant women and fetuses. Pregnant women should obtain oral health knowledge and enhance their awareness. Dentists should adhere to the principle of "prevention before pregnancy, controlling symptoms during pregnancy, and treating diseases after pregnancy" for different pregnancy periods. They should also formulate appropriate treatment plans to control emergencies, prevent disease progression, and avoid harmful effects on pregnant women by using the safest, simplest, and most effective strategies that avoid adverse effects on fetuses. Pregnant women and dentists should combine prevention and treatment while collaborating in maintaining oral health during pregnancy. This article focuses on the principles of treatment during pregnancy, and the treatment timing, clinical management, and treatment strategies of different diseases causing oral emergencies during pregnancy are reviewed.
Collapse
Affiliation(s)
- Xuefeng Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xian Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chengge Hua
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
41
|
He Z, Liu C, Lin L, Feng G, Wu G. Real-world safety of Levetiracetam: Mining and analysis of its adverse drug reactions based on FAERS database. Seizure 2024; 117:253-260. [PMID: 38537425 DOI: 10.1016/j.seizure.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
INTRODUCTION Levetiracetam is a relatively new and widely utilized anti-seizure medication; however, limited information is available regarding its adverse effects. This study aims to thoroughly investigate, evaluate, and present evidence on the safety profile of Levetiracetam, relying on data from the FDA Adverse Event Reporting System (FAERS) database to facilitate informed clinical decision-making. METHODS We employed various statistical measures, including Reporting Odds Ratio (ROR), Proportionate Reporting Ratio (PRR), and analysis by the Medicines and Healthcare Products Regulatory Agency (MHRA), to identify signals of adverse reactions associated with Levetiracetam. Positive signals consistent with Designated Medical Event (DME) were singled out for focused comparison and discussion. RESULTS The analysis of 26,182 adverse events linked to Levetiracetam as the primary suspected drug revealed 692 positive signals spanning 22 System Organ Classes (SOCs). Nervous system disorders were the most frequently reported, followed by psychiatric disorders, and general disorders and administration site conditions. 11 positive signals consistent with Preferred Terms (PTs) in DME were identified, predominantly concentrated in 6 SOCs. Among these, rhabdomyolysis, Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), drug reaction with eosinophilia and systemic symptoms (DRESS) exhibited relatively large values of A, ROR, and Chi-squared. Additionally, PTs related to spontaneous abortion, drug interaction, urethral atresia, ventricular septal defect, and atrial septal defect showed significant strength. CONCLUSIONS The study indicates that Levetiracetam carries a potential risk of causing rhabdomyolysis, SJS, TEN, DRESS as well as spontaneous abortion. Signals related to drug interaction, urethral atresia, ventricular septal defect, and atrial septal defect warrant heightened attention in clinical use.
Collapse
Affiliation(s)
- Zhimin He
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Cuimin Liu
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, PR China
| | - Lin Lin
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, PR China; School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Guowen Feng
- Department of Pharmacy, Langzhong People's Hospital, Nanchong, Sichuan, 637400, PR China.
| | - Gang Wu
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, PR China.
| |
Collapse
|
42
|
Ward K, Citrome L. Tolerability and safety outcomes of first-line oral second-generation antipsychotics in patients with schizophrenia. Expert Opin Drug Saf 2024; 23:399-409. [PMID: 38467517 DOI: 10.1080/14740338.2024.2328812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Antipsychotics are the foundation of pharmacologic treatment for schizophrenia. There are many oral antipsychotics available and given that these medications are generally considered comparably efficacious when titrated to an adequate dose, their varied tolerability, and safety profiles become critically important for medication selection. AREAS COVERED This paper reviews tolerability and safety considerations for first-line second-generation oral antipsychotics currently approved for the treatment of schizophrenia in the USA. Excluded from consideration are clozapine and non-oral formulations. EXPERT OPINION Among antipsychotics, there are many differences in adverse reactions observed in clinical trials, such as variable likelihood to cause sedation vs insomnia, weight gain and abnormalities in glucose/lipid metabolism, hyperprolactinemia, potential for impact on the QT interval, and motoric adverse effects. Additional safety data that can help with medication selection include safety in pregnancy and lactation, and potential for drug-drug interactions. Ultimately, working with patients to personalize treatment by focusing on safety and individual tolerability considerations for various adverse effects can help in building a therapeutic alliance and improving patients' outcomes.
Collapse
Affiliation(s)
- Kristen Ward
- Clinical Pharmacy Department, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Leslie Citrome
- Department of Psychiatry and Behavioral Science, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
43
|
Versluis J, Bourgonje AR, Touw DJ, Meinderts JR, Prins JR, de Jong MFC, Mian P. Pharmacokinetics of Tacrolimus in Pregnant Solid-Organ Transplant Recipients: A Retrospective Study. J Clin Pharmacol 2024; 64:428-436. [PMID: 38084781 DOI: 10.1002/jcph.2393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024]
Abstract
Data on the pharmacokinetics of tacrolimus during pregnancy are limited. Therefore, the aim of this retrospective study was to characterize the whole-blood pharmacokinetics of tacrolimus throughout pregnancy. In this single-center retrospective cohort study, whole-blood tacrolimus trough concentrations corrected for the dose (concentration-to-dose [C/D] ratios) were compared before, monthly during, and after pregnancy in kidney, liver, and lung transplant recipients who became pregnant and gave birth between 2000 and 2022. Descriptive statistics and linear mixed models were used to characterize changes in tacrolimus C/D ratios before, during, and after pregnancy. The total study population included 46 pregnancies (31 pregnant women). Nineteen, 21, and 6 pregnancies were following kidney, liver, and lung transplantation, respectively. Immediate-release or extended-release formulations were used in 54.5% and 45.5% of the women, respectively. Tacrolimus C/D ratios significantly (P < .001) decreased (-48%) compared to the prepregnancy state at 7 months of pregnancy. These ratios recovered within 3 months postpartum (P = .002). C/D ratios tended to be lower during treatment with an extended-release formulation than with an immediate-release formulation (P = .071). Transplantation type did not significantly affect C/D ratios during pregnancy (P = .873). In conclusion, we found that tacrolimus whole-blood pharmacokinetics change throughout pregnancy, with the lowest C/D ratios (48% decrease) in the 7th month of pregnancy. In general, the decrease in C/D ratios seems to stabilize from month 4 onward compared to prepregnancy.
Collapse
Affiliation(s)
- Jorn Versluis
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Jildau R Meinderts
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Margriet F C de Jong
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Paola Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| |
Collapse
|
44
|
Fashe MM, Le TV, Gower MN, Mulrenin IR, Dorman KF, Smith S, Fallon JK, Dumond JB, Boggess KA, Lee CR. Impact of Pregnancy on the Pharmacokinetics and Metabolism of Nicotinamide in Humans. Clin Pharmacol Ther 2024; 115:556-564. [PMID: 38093631 PMCID: PMC11250906 DOI: 10.1002/cpt.3146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023]
Abstract
In pre-eclampsia models, nicotinamide (NAM) has protective effects in pre-eclampsia and is being evaluated as a therapeutic nutraceutical in clinical studies. NAM undergoes extensive hepatic metabolism by NAM N-methyltransferase to methylnicotinamide (MNA), which is subsequently metabolized to methyl-2-pyridone-5-carboxamide (M2PY) by aldehyde oxidase. However, the pharmacokinetics of NAM and its major metabolites has never been studied in pregnant individuals. Blood samples were collected before and 1, 2, 4, 8, and 24 hours after single 1 g oral NAM dose in healthy pregnant (gestational age 24-33 weeks) and nonpregnant female volunteers (n = 6/group). Pooled urine was collected from 0 to 8 hours. NAM, MNA, and M2PY area under the concentration-time curve (AUC) data were analyzed by noncompartmental analysis. No difference in the plasma AUC0→24 of NAM (median (25%-75%): 463 (436-576) vs. 510 (423, 725) μM*hour, P = 0.430) and its intermediate metabolite MNA (89.1 (60.4, 124.4) vs. 83.8 (62.7, 93.7) μM*hour, P = 0.515) was observed in pregnant and nonpregnant volunteers, respectively; however, the terminal metabolite M2PY AUC0 → 24 was significantly lower in pregnant individuals (218 (188, 254) vs. 597 (460, 653) μM*hour, P < 0.001). NAM renal clearance (CLR ; P = 0.184), MNA CLR (P = 0.180), and total metabolite formation clearance (P = 0.405) did not differ across groups; however, M2PY CLR was significantly higher in pregnant individuals (10.5 (9.3-11.3) vs. 7.5 (6.4-8.5) L/h, P = 0.002). These findings demonstrate that the PK of NAM and systemic exposure to its intermediate metabolite MNA are not significantly altered during pregnancy, and systemic exposure to NAM's major metabolite M2PY was reduced during pregnancy due to increased renal elimination.
Collapse
Affiliation(s)
- Muluneh M. Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tien V. Le
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Megan N. Gower
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ian R. Mulrenin
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Karen F. Dorman
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Spenser Smith
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John K. Fallon
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Julie B. Dumond
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kim A. Boggess
- Department of Obstetrics & Gynecology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
45
|
Matjuda EN, Engwa GA, Mungamba MM, Sewani-Rusike CR, Goswami N, Nkeh-Chungag BN. Cardio-Metabolic Health of Offspring Exposed in Utero to Human Immuno-Deficiency Virus and Anti-Retroviral Treatment: A Systematic Review. BIOLOGY 2024; 13:32. [PMID: 38248463 PMCID: PMC10813696 DOI: 10.3390/biology13010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND Antiretroviral treatment (ART) use during pregnancy continues to rise as it is known to decrease the likelihood of HIV transmission from mother to child. However, it is still unknown whether foetal exposure to (ART) may affect the foetal environment, predisposing the offspring to cardiometabolic risk. Therefore, the aim of this study was to systematically review the cardio-metabolic effects of in utero exposure to HIV/ART on offspring. METHODS We carried out a systematic review and obtained literature from the Google scholar, PubMed, ProQuest, Web of Science, and Scopus databases. Two independent reviewers evaluated the titles, abstracts, and full-length English contents. Data from the eligible studies were included. RESULTS The search yielded 7596 records. After assessing all of these records, 35 of the full-length articles were included in this systematic review. Several studies showed that low birth weight, small head circumference, and altered mitochondrial content were more common among HIV-exposed uninfected (HEU) children compared to HIV-unexposed uninfected children (HUU). A few studies demonstrated elevated triglyceride levels, lower levels of insulin, and increased blood pressure, oxidative stress, vascular dysfunction, cardiac damage, and myocardial dysfunction among HEU children compared with HUU children. CONCLUSION Most findings showed that there were cardio-metabolic health risk factors among HEU children, indicating that maternal exposure to HIV and ART may negatively affect foetal health, which may lead to cardio-metabolic morbidity later in life.
Collapse
Affiliation(s)
- Edna Ngoakoana Matjuda
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (E.N.M.); (M.M.M.); (C.R.S.-R.)
| | - Godwill Azeh Engwa
- Department of Biological and Environmental Sciences, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (G.A.E.); (N.G.)
| | - Muhulo Muhau Mungamba
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (E.N.M.); (M.M.M.); (C.R.S.-R.)
| | - Constance Rufaro Sewani-Rusike
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (E.N.M.); (M.M.M.); (C.R.S.-R.)
| | - Nandu Goswami
- Department of Biological and Environmental Sciences, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (G.A.E.); (N.G.)
- Physiology Division, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstrasse 6, D-5 A, 8036 Graz, Austria
- Department of Health Sciences, Alma Mater Europaea, 2000 Maribor, Slovenia
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Benedicta Ngwenchi Nkeh-Chungag
- Department of Biological and Environmental Sciences, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (G.A.E.); (N.G.)
| |
Collapse
|
46
|
Morel C, Paoli J, Emond C, Debaugnies F, Hardy EM, Creta M, Montagne M, Borde P, Nieuwenhuyse AV, Duca RC, Schroeder H, Grova N. Pharmacokinetic characterisation of a valproate Autism Spectrum Disorder rat model in a context of co-exposure to α-Hexabromocyclododecane. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 105:104343. [PMID: 38122861 DOI: 10.1016/j.etap.2023.104343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Assessing the role of α-hexabromocyclododecane α-HBCDD as a factor of susceptibility for Autism Spectrum disorders by using valproic acid-exposed rat model (VPA) required characterizing VPA pharmacokinetic in the context of α-HBCDD-co-exposure in non-pregnant and pregnant rats. The animals were exposed to α-HBCDD by gavage (100 ng/kg/day) for 12 days. This was followed by a single intraperitoneal dose of VPA (500 mg/kg) or a daily oral dose of VPA (500 mg/kg) for 3 days. Exposure to α-HBCDD did not affect the pharmacokinetics of VPA in pregnant or non-pregnant rats. Surprisingly, VPA administration altered the pharmacokinetics of α-HBCDD. VPA also triggered higher foetal toxicity and lethality with the PO than IP route. α-HBCDD did not aggravate the embryotoxicity observed with VPA, regardless of the route of exposure. Based on this evidence, a single administration of 500 mg/kg IP is the most suitable VPA model to investigate α-HBCDD co-exposure.
Collapse
Affiliation(s)
- C Morel
- Calbinotox EA-7488, Faculty of Science and Technology, University of Lorraine, Campus Aiguillettes, B.P. 70239, 54506 Vandœuvre-lès-Nancy, France.
| | - J Paoli
- Calbinotox EA-7488, Faculty of Science and Technology, University of Lorraine, Campus Aiguillettes, B.P. 70239, 54506 Vandœuvre-lès-Nancy, France; UMR Inserm 1256 nGERE, Nutrition-Génétique et exposition aux risques environnementaux, Institute of Medical Research (Pôle BMS) - University of Lorraine, B.P. 184, 54511 Nancy, France.
| | - C Emond
- Calbinotox EA-7488, Faculty of Science and Technology, University of Lorraine, Campus Aiguillettes, B.P. 70239, 54506 Vandœuvre-lès-Nancy, France; PKSH Inc., Crabtree, Quebec, Canada; School of Public Health, DSEST, University of Montreal, Montreal, Quebec, Canada.
| | - F Debaugnies
- Department of Medical Biology, National Health Laboratory (LNS), Dudelange, Grand Duchy of Luxembourg.
| | - E M Hardy
- Department of Health Protection, National Health Laboratory (LNS), Dudelange, Grand Duchy of Luxembourg.
| | - M Creta
- Department of Health Protection, National Health Laboratory (LNS), Dudelange, Grand Duchy of Luxembourg.
| | - M Montagne
- Department of Health Protection, National Health Laboratory (LNS), Dudelange, Grand Duchy of Luxembourg.
| | - P Borde
- Department of Medical Biology, National Health Laboratory (LNS), Dudelange, Grand Duchy of Luxembourg.
| | - A Van Nieuwenhuyse
- Department of Health Protection, National Health Laboratory (LNS), Dudelange, Grand Duchy of Luxembourg; Environment and Health, Department of Public Health and Primary Care, University of Leuven (KU Leuven), Leuven, Belgium.
| | - R C Duca
- Department of Health Protection, National Health Laboratory (LNS), Dudelange, Grand Duchy of Luxembourg; Environment and Health, Department of Public Health and Primary Care, University of Leuven (KU Leuven), Leuven, Belgium.
| | - H Schroeder
- Calbinotox EA-7488, Faculty of Science and Technology, University of Lorraine, Campus Aiguillettes, B.P. 70239, 54506 Vandœuvre-lès-Nancy, France; UMR Inserm 1256 nGERE, Nutrition-Génétique et exposition aux risques environnementaux, Institute of Medical Research (Pôle BMS) - University of Lorraine, B.P. 184, 54511 Nancy, France.
| | - N Grova
- Calbinotox EA-7488, Faculty of Science and Technology, University of Lorraine, Campus Aiguillettes, B.P. 70239, 54506 Vandœuvre-lès-Nancy, France; UMR Inserm 1256 nGERE, Nutrition-Génétique et exposition aux risques environnementaux, Institute of Medical Research (Pôle BMS) - University of Lorraine, B.P. 184, 54511 Nancy, France; Immune Endocrine Epigenetics Research Group, Department of Infection and Immunity-Luxembourg Institute of Health, 29 rue Henri Koch, L-4354, Esch-Sur-Alzette, Grand Duchy of Luxembourg.
| |
Collapse
|
47
|
Yang X, Grimstein M, Pressly M, Fletcher EP, Shord S, Leong R. Utility of Physiologically Based Pharmacokinetic Modeling to Investigate the Impact of Physiological Changes of Pregnancy and Cancer on Oncology Drug Pharmacokinetics. Pharmaceutics 2023; 15:2727. [PMID: 38140068 PMCID: PMC10748010 DOI: 10.3390/pharmaceutics15122727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The treatment of cancer during pregnancy remains challenging with knowledge gaps in drug dosage, safety, and efficacy due to the under-representation of this population in clinical trials. Our aim was to investigate physiological changes reported in both pregnancy and cancer populations into a PBPK modeling framework that allows for a more accurate estimation of PK changes in pregnant patients with cancer. METHODS Paclitaxel and docetaxel were selected to validate a population model using clinical data from pregnant patients with cancer. The validated population model was subsequently used to predict the PK of acalabrutinib in pregnant patients with cancer. RESULTS The Simcyp pregnancy population model reasonably predicted the PK of docetaxel in pregnant patients with cancer, while a modified model that included a 2.5-fold increase in CYP2C8 abundance, consistent with the increased expression during pregnancy, was needed to reasonably predict the PK of paclitaxel in pregnant patients with cancer. Changes in protein binding levels of patients with cancer had a minimal impact on the predicted clearance of paclitaxel and docetaxel. PBPK modeling predicted approximately 60% lower AUC and Cmax for acalabrutinib in pregnant versus non-pregnant patients with cancer. CONCLUSIONS Our results suggest that PBPK modeling is a promising approach to investigate the effects of pregnancy and cancer on the PK of oncology drugs and potentially inform dosing for pregnant patients with cancer. Further evaluation and refinement of the population model are needed for pregnant patients with cancer with additional compounds and clinical PK data.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruby Leong
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA; (X.Y.); (M.G.); (S.S.)
| |
Collapse
|
48
|
Wanar A, Saia K, Field TA. Accelerated Fentanyl Metabolism During Pregnancy and Impact on Prenatal Drug Testing. Matern Child Health J 2023; 27:1944-1948. [PMID: 37269392 DOI: 10.1007/s10995-023-03664-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2023] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To evaluate the metabolic pattern of illicit fentanyl in a sample of pregnant patients with opioid use disorder. Fentanyl pharmacokinetics during pregnancy are currently understudied yet the interpretation of a fentanyl immunoassay during pregnancy has significant implications on maternal legal custody and child welfare. Through this medical-legal lens, we demonstrate the utility of an emerging metric, the metabolic ratio, for accurate analysis of fentanyl pharmacokinetics during pregnancy. METHODS We conducted a retrospective cohort analysis using the electronic medical records of 420 patients receiving integrated prenatal and opioid use disorder care at a large urban safety net hospital. Data related to maternal health and substance use were collected for each subject. The metabolic ratio was calculated for each subject to measure their rate of metabolism. The sample's (n = 112) metabolic ratios were compared with a large non-pregnant sample (n = 4366). RESULTS The metabolic ratios of our pregnant sample were significantly (p = .0001) higher than the metabolic ratios of our non-pregnant sample, indicating that the rate of conversion to the major metabolite was faster in pregnant people. The effect size for this difference between the pregnant and non-pregnant sample was large (d = 0.86). CONCLUSIONS FOR PRACTICE Our findings characterize the unique metabolic pattern of fentanyl in pregnant people who use opioids, providing guidance for institutional policies around fentanyl drug testing. Additionally, our study warns of misinterpretation of toxicology results and stresses the importance of physician advocacy on behalf of pregnant women who use illicit opioids.
Collapse
Affiliation(s)
- Amita Wanar
- OB/GYN, Boston University School of Medicine, Boston, MA, USA
- Departments of Obstetrics and Gynecology and Psychiatry, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Kelley Saia
- OB/GYN, Boston University School of Medicine, Boston, MA, USA.
- Departments of Obstetrics and Gynecology and Psychiatry, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA.
| | - Thomas A Field
- OB/GYN, Boston University School of Medicine, Boston, MA, USA
- Departments of Obstetrics and Gynecology and Psychiatry, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
49
|
Zitoun N, Campbell MK, Matsui D, Garcia-Bournissen F. Prospective evaluation of pregnancy outcomes after gestational exposure to prazosin. Br J Clin Pharmacol 2023; 89:3324-3329. [PMID: 37323115 DOI: 10.1111/bcp.15829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/12/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023] Open
Abstract
AIMS Prazosin is an antihypertensive medication which can be used to help with post-traumatic stress disorder (PTSD) symptoms. Little data is currently available on its safety in pregnancy. The aim of this study was to assess the fetal and pregnancy safety associated with prazosin exposures in early pregnancy. METHODS Subjects were 11 patients who took prazosin during pregnancy and were counselled at the FRAME clinic in London Health Sciences Centre (Ontario, Canada) between 1 January 2000 and 31 December 2021. Data on their other exposures and pregnancy outcomes were collected from medical records and through telephone questionnaires. RESULTS It was found that 6/11 (54.5%) subjects did not report any adverse outcomes and experienced uneventful pregnancies. There were two miscarriages. Birthweights were within the normal range for the remaining nine pregnancies. Adverse events reported were consistent with background population expectation, including: one postpartum haemorrhage, one case of preeclampsia, one preterm birth, two NICU admissions, and two caesarean sections. CONCLUSIONS For these 11 subjects, pregnancy outcomes after exposure to prazosin were consistent with typical outcomes from unexposed pregnancies. More data are needed to conclude that prazosin is safe for use in pregnant subjects. However, the lack of adverse effects above baseline is reassuring to future patients who may be unintentionally exposed to prazosin while pregnant. Therefore, this study contributes valuable data towards monitoring safety of prazosin in pregnancy.
Collapse
Affiliation(s)
- Natalie Zitoun
- Department of Epidemiology & Biostatistics, University of Western Ontario, London, ON, Canada
| | - M Karen Campbell
- Department of Epidemiology & Biostatistics, University of Western Ontario, London, ON, Canada
- Department of Pediatrics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
- Department of Obstetrics & Gynecology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Doreen Matsui
- Department of Pediatrics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Facundo Garcia-Bournissen
- Department of Epidemiology & Biostatistics, University of Western Ontario, London, ON, Canada
- Department of Pediatrics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
50
|
Coppola P, Butler A, Cole S, Kerwash E. Total and Free Blood and Plasma Concentration Changes in Pregnancy for Medicines Highly Bound to Plasma Proteins: Application of Physiologically Based Pharmacokinetic Modelling to Understand the Impact on Efficacy. Pharmaceutics 2023; 15:2455. [PMID: 37896215 PMCID: PMC10609738 DOI: 10.3390/pharmaceutics15102455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Free drug concentrations are generally considered the pharmacologically active moiety and are important for cellular diffusion and distribution. Pregnancy-related changes in plasma protein binding and blood partitioning are due to decreases in plasma albumin, alpha-1-acid glycoprotein, and haematocrit; this may lead to increased free concentrations, tissue distribution, and clearance during pregnancy. In this paper we highlight the importance and challenges of considering changes in total and free concentrations during pregnancy. For medicines highly bound to plasma proteins, such as tacrolimus, efavirenz, clindamycin, phenytoin, and carbamazepine, differential changes in concentrations of free drug during pregnancy may be clinically significant and have important implications for dose adjustment. Therapeutic drug monitoring usually relies on the measurement of total concentrations; this can result in dose adjustments that are not necessary when changes in free concentrations are considered. We explore the potential of physiologically based pharmacokinetic (PBPK) models to support the understanding of the changes in plasma proteins binding, using tacrolimus and efavirenz as example drug models. The exposure to either drug was predicted to be reduced during pregnancy; however, the decrease in the exposure to the total tacrolimus and efavirenz were significantly larger than the reduction in the exposure to the free drug. These data show that PBPK modelling can support the impact of the changes in plasma protein binding and may be used for the simulation of free concentrations in pregnancy to support dosing decisions.
Collapse
|