1
|
Crump CJ, Abuelazm H, Ibrahim K, Shah S, El-Mallakh RS. An overview of the efficacy and safety of brexpiprazole for the treatment of schizophrenia in adolescents. Expert Rev Neurother 2024; 24:727-733. [PMID: 38864423 DOI: 10.1080/14737175.2024.2367695] [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: 09/26/2023] [Accepted: 06/10/2024] [Indexed: 06/13/2024]
Abstract
INTRODUCTION The onset of psychotic symptoms occurs prior to age 19 in 39% of the patients with schizophrenia. There are limited approved treatment options for adolescents with schizophrenia. Brexpiprazole was approved by the United States Food and Drug Administration (FDA) for treatment of schizophrenia in adolescents in 2022. AREAS COVERED Extrapolation of adult data to youth and use of pharmacologic modeling coupled with open long-term safety data were used by the FDA to approve brexpiprazole for adolescent schizophrenia. They were all reviewed herein. EXPERT OPINION D2 receptor partial agonist antipsychotic agents are preferred in the early phase of treatment of psychotic disorders. Approval of brexpiprazole in adolescent schizophrenia provides an additional option. Brexpiprazole was approved by the FDA on the basis of extrapolation of adult data without controlled trials in adolescents. This reduces placebo exposure in young people. Two previous agents (asenapine and ziprasidone) approved for adult schizophrenia failed to separate from placebo in adolescent schizophrenia studies; this partially undermines the process of extrapolation. For brexpiprazole, the paucity of data in adolescents relegates it to a second-line agent. More research on brexpiprazole is needed to delineate its relative role in the management of adolescent schizophrenia.
Collapse
Affiliation(s)
- Chesika J Crump
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Hagar Abuelazm
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kirolos Ibrahim
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Shaishav Shah
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Rif S El-Mallakh
- Mood Disorders Research Program, Depression Center, Department of Psychiatry and Behavioral Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
2
|
Anliker-Ort M, Rodieux F, Ziesenitz VC, Atkinson A, Bielicki JA, Erb TO, Gürtler N, Holland-Cunz S, Duthaler U, Rudin D, Haschke M, van den Anker J, Pfister M, Gotta V. Pharmacokinetics-Based Pediatric Dose Evaluation and Optimization Using Saliva - A Case Study. J Clin Pharmacol 2024; 64:810-819. [PMID: 38497339 DOI: 10.1002/jcph.2428] [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: 11/26/2023] [Accepted: 02/17/2024] [Indexed: 03/19/2024]
Abstract
Understanding pharmacokinetics (PK) in children is a prerequisite to determine optimal pediatric dosing. As plasma sampling in children is challenging, alternative PK sampling strategies are needed. In this case study we evaluated the suitability of saliva as alternative PK matrix to simplify studies in infants, investigating metamizole, an analgesic used off-label in infants. Six plasma and 6 saliva PK sample collections were scheduled after a single intravenous dose of 10 mg/kg metamizole. Plasma/saliva pharmacometric (PMX) modeling of the active metabolites 4-methylaminoantipyrine (4-MAA) and 4-aminoantipyrine (4-AA) was performed. Various reduced plasma sampling scenarios were evaluated by PMX simulations. Saliva and plasma samples from 25 children were included (age range, 5-70 months; weight range, 8.7-24.8 kg). Distribution of metamizole metabolites between plasma and saliva was without delay. Estimated mean (individual range) saliva/plasma fractions of 4-MAA and 4-AA were 0.32 (0.05-0.57) and 0.57 (0.25-0.70), respectively. Residual variability of 4-MAA (4-AA) in saliva was 47% (28%) versus 17% (11%) in plasma. A simplified sampling scenario with up to 6 saliva samples combined with 1 plasma sample was associated with similar PK parameter estimates as the full plasma sampling scenario. This case study with metamizole shows increased PK variability in saliva compared to plasma, compromising its suitability as single matrix for PK studies in infants. Nonetheless, rich saliva sampling can reduce the number of plasma samples required for PK characterization, thereby facilitating the conduct of PK studies to optimize dosing in pediatric patients.
Collapse
Affiliation(s)
- Marion Anliker-Ort
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Frédérique Rodieux
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Victoria C Ziesenitz
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
- Pediatric and Congenital Cardiology, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Andrew Atkinson
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
- Infectious Diseases Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Julia A Bielicki
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
- Pediatric Infectious Diseases, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Thomas O Erb
- Pediatric Anesthesiology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Nicolas Gürtler
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefan Holland-Cunz
- Pediatric Surgery, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Urs Duthaler
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Deborah Rudin
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Manuel Haschke
- Clinical Pharmacology and Toxicology, Department of General Internal Medicine, University Hospital Bern, University of Bern, Bern, Switzerland
| | - John van den Anker
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Verena Gotta
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| |
Collapse
|
3
|
Maher TM, Brown KK, Cunningham S, DeBoer EM, Deterding R, Fiorino EK, Griese M, Schwerk N, Warburton D, Young LR, Gahlemann M, Voss F, Stock C. Estimating the effect of nintedanib on forced vital capacity in children and adolescents with fibrosing interstitial lung disease using a Bayesian dynamic borrowing approach. Pediatr Pulmonol 2024. [PMID: 38289091 DOI: 10.1002/ppul.26882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/15/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND The rarity of childhood interstitial lung disease (chILD) makes it challenging to conduct powered trials. In the InPedILD trial, among 39 children and adolescents with fibrosing ILD, there was a numerical benefit of nintedanib versus placebo on change in forced vital capacity (FVC) over 24 weeks (difference in mean change in FVC % predicted of 1.21 [95% confidence interval: -3.40, 5.81]). Nintedanib has shown a consistent effect on FVC across populations of adults with different diagnoses of fibrosing ILD. METHODS In a Bayesian dynamic borrowing analysis, prespecified before data unblinding, we incorporated data on the effect of nintedanib in adults and the data from the InPedILD trial to estimate the effect of nintedanib on FVC in children and adolescents with fibrosing ILD. The data from adults were represented as a meta-analytic predictive (MAP) prior distribution with mean 1.69 (95% credible interval: 0.49, 3.08). The adult data were weighted according to expert judgment on their relevance to the efficacy of nintedanib in chILD, obtained in a formal elicitation exercise. RESULTS Combined data from the MAP prior and InPedILD trial analyzed within the Bayesian framework resulted in a median difference between nintedanib and placebo in change in FVC % predicted at Week 24 of 1.63 (95% credible interval: -0.69, 3.40). The posterior probability for superiority of nintedanib versus placebo was 95.5%, reaching the predefined success criterion of at least 90%. CONCLUSION These findings, together with the safety data from the InPedILD trial, support the use of nintedanib in children and adolescents with fibrosing ILDs.
Collapse
Affiliation(s)
- Toby M Maher
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kevin K Brown
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Steven Cunningham
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Emily M DeBoer
- Section of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Colorado Denver, Denver, Colorado, USA
- The Children's Hospital Colorado, Aurora, Colorado, USA
| | - Robin Deterding
- Section of Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, University of Colorado Denver, Denver, Colorado, USA
- The Children's Hospital Colorado, Aurora, Colorado, USA
| | - Elizabeth K Fiorino
- Departments of Science Education and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Matthias Griese
- Hauner Children's Hospital, German Center for Lung Research (DZL), Ludwig Maximilians University, Munich, Germany
| | - Nicolaus Schwerk
- Clinic for Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - David Warburton
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Lisa R Young
- Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Florian Voss
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim am Rhein, Germany
| | - Christian Stock
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim am Rhein, Germany
| |
Collapse
|
4
|
Freerks L, Arien T, Mackie C, Inghelbrecht S, Klein S. A toolbox for mimicking gastrointestinal conditions in children: Design and evaluation of biorelevant dissolution media for mimicking paediatric gastric- and small intestinal conditions. Eur J Pharm Biopharm 2023; 193:144-157. [PMID: 37852543 DOI: 10.1016/j.ejpb.2023.10.011] [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: 08/10/2023] [Revised: 10/04/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
The goal of the present work was to develop an in vitro toolbox to evaluate the oral administration of dosage forms to children of different age groups and under different administration conditions (fasted/fed). Based on current data on the gastrointestinal physiology of children, a set of new biorelevant media was designed to mimic the composition and physicochemical properties of resting gastric and resting small intestinal fluid in children of different age groups. In addition, guidelines were developed on how to generate fasted and fed state gastric and small intestinal fluids by combining these media with age-specific drinking volumes or portions of already established simulated paediatric breakfast meals, respectively. These fluids can simulate the conditions in the paediatric stomach and small intestine after administration of a dosage form in the fasting state or after a breakfast. The in vitro toolbox was evaluated using the example of pre-school children with a total of five paediatric medicines. Results from the corresponding set of in vitro studies highlight the importance of addressing patient-specific characteristics rather than downscaling existing adult in vitro models.
Collapse
Affiliation(s)
- Lisa Freerks
- Department of Pharmacy, University of Greifswald, 17489 Greifswald, Germany
| | - Tina Arien
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Claire Mackie
- Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | | | - Sandra Klein
- Department of Pharmacy, University of Greifswald, 17489 Greifswald, Germany.
| |
Collapse
|
5
|
Goulooze SC, Vis PW, Krekels EHJ, Knibbe CAJ. Advances in pharmacokinetic-pharmacodynamic modelling for pediatric drug development: extrapolations and exposure-response analyses. Expert Rev Clin Pharmacol 2023; 16:1201-1209. [PMID: 38069812 DOI: 10.1080/17512433.2023.2288171] [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: 08/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023]
Abstract
INTRODUCTION Pharmacokinetic (PK)-Pharmacodynamic (PD) and exposure-response (E-R) modeling are critical parts of pediatric drug development. By integrating available knowledge and supportive data to support the design of future studies and pediatric dose selection, these techniques increase the efficiency of pediatric drug development and lowers the risk of exposing pediatric study participants to suboptimal or unsafe dose regimens. AREAS COVERED The role of PK, PK-PD and E-R modeling within pediatric drug development and pediatric dose selection is discussed. These models allow investigation of the impact of age and bodyweight on PK and PD in children, despite the often sparse data on the pediatric population. Also discussed is how E-R analyses strengthen the evidence basis to support (full or partial) extrapolation of drug efficacy from adults to children, and between different pediatric age groups. EXPERT OPINION Accelerated pediatric drug development and optimized pediatric dosing guidelines are expected from three future developments: (1) Increased focus on E-R modeling of currently approved drugs in children resulting in (novel) E-R modeling techniques and best practices, (2) increased use of real-world data for E-R (3) increased implementation of available population PK and E-R information in pediatric drug dosing guidelines.
Collapse
Affiliation(s)
| | - Peter W Vis
- LAP&P Consultants BV, Leiden, The Netherlands
| | - Elke H J Krekels
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Catherijne A J Knibbe
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
6
|
Walker R, Phillips B, Dias S. Comparison of Bayesian methods for incorporating adult clinical trial data to improve certainty of treatment effect estimates in children. PLoS One 2023; 18:e0281791. [PMID: 37319173 PMCID: PMC10270354 DOI: 10.1371/journal.pone.0281791] [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: 01/30/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023] Open
Abstract
There are challenges associated with recruiting children to take part in randomised clinical trials and as a result, compared to adults, in many disease areas we are less certain about which treatments are most safe and effective. This can lead to weaker recommendations about which treatments to prescribe in practice. However, it may be possible to 'borrow strength' from adult evidence to improve our understanding of which treatments work best in children, and many different statistical methods are available to conduct these analyses. In this paper we discuss four Bayesian methods for extrapolating adult clinical trial evidence to children. Using an exemplar dataset, we compare the effect of their modelling assumptions on the estimated treatment effect and associated heterogeneity. These modelling assumptions range from adult evidence being completely generalisable to being completely unrelated to the children's evidence. We finally discuss the appropriateness of these modelling assumptions in the context of estimating treatment effect in children.
Collapse
Affiliation(s)
- Ruth Walker
- Centre for Reviews and Dissemination, University of York, York, North Yorkshire, United Kingdom
| | - Bob Phillips
- Centre for Reviews and Dissemination, University of York, York, North Yorkshire, United Kingdom
- Department of Paediatric Haematology and Oncology, Leeds Teaching Hospitals NHS Trust, Leeds, West Yorkshire, United Kingdom
| | - Sofia Dias
- Centre for Reviews and Dissemination, University of York, York, North Yorkshire, United Kingdom
| |
Collapse
|
7
|
Franzese R, Riccobene T, Carrothers T, Vourvahis M, Winter E, Lovern M, McFadyen L. Population Pharmacokinetic Modeling for Ceftazidime-Avibactam Renal Dose Adjustments in Pediatric Patients 3 months and Older. Clin Pharmacol Ther 2023; 113:182-195. [PMID: 36239478 DOI: 10.1002/cpt.2764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/02/2022] [Indexed: 12/24/2022]
Abstract
Ceftazidime-avibactam is a novel β-lactam/β-lactamase inhibitor combination developed to treat serious Gram-negative bacterial infections; approved indications include complicated urinary tract infection, complicated intra-abdominal infection, and hospital-acquired pneumonia including ventilator-associated pneumonia in patients ≥ 3 months old. Because of the predominantly renal clearance of ceftazidime and avibactam, dose adjustments (reductions) are required for patients with estimated creatinine clearance (CrCL) ≤ 50 mL/min. We describe the application of combined adult and pediatric population pharmacokinetic models in developing ceftazidime-avibactam dose recommendations for pediatric patients ≥ 2 to < 18 years old with body surface area-normalized CrCL ≤ 50 mL/min/1.73 m2 , including moderate, severe, or very severe renal impairment, or end-stage renal disease requiring hemodialysis, and for patients ≥ 3 months to < 2 years old with mild, moderate, or severe renal impairment. Models included allometric scaling for all subjects and simulations (1,000 subjects per age group, renal function group, and indication) were performed nonparametrically using post hoc random effects. Doses were selected based on simulated pediatric patients achieving steady-state exposures similar to adults and high probability of target attainment (using a simultaneous joint target for both ceftazidime and avibactam). Because there were few children with renal impairment in the ceftazidime-avibactam clinical trials, selected pediatric doses were guided by extrapolation and matching of adult exposures associated with efficacy and within established safety margins. The recommended doses for pediatric patients with estimated CrCL ≤ 50 mL/min/1.73 m2 use equivalent adjustments in dose quantity and/or administration interval (vs. the corresponding age group with normal renal function) as those for adults.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark Lovern
- Certara Strategic Consulting, Raleigh, North Carolina, USA
| | | |
Collapse
|
8
|
Busner J, Pandina G, Day S, Mahableshwarkar A, Kempf L, Sheean M, Dunn J. Patient Centricity: Design and Conduct of Clinical Trials in Orphan Diseases: Third of Three Sets of Expanded Proceedings from the 2020 ISCTM Autumn Conference on Pediatric Drug Development. INNOVATIONS IN CLINICAL NEUROSCIENCE 2023; 20:25-31. [PMID: 37122576 PMCID: PMC10132279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
This article expands on a session, titled "Patient Centricity: Design and Conduct of Clinical Trials in Orphan Diseases," that was presented as part of a two-day meeting on Pediatric Drug Development at the International Society for Central Nervous System (CNS) Clinical Trials and Methodology (ISCTM) Autumn Conference in Boston, Massachusetts, in October 2020. Speakers from various areas of pediatric drug development addressed a variety of implications of including children in drug development programs, including implications for rare/orphan diseases. The speakers have written summaries of their talks. The session's lead Chair was Dr. Joan Busner, who wrote introductory and closing comments. Dr. Simon Day, regulatory consultant, outlined some of the past mistakes that have plagued trials that did not consult with patient groups in the early design phase. Dr. Atul Mahableshwarkar provided an industry perspective of a recent trial that benefited from the inclusion of patient input. Drs. Lucas Kempf and Maria Sheean provided regulatory input from the perspectives of the United States (US) Food and Drug Administration (FDA) and European Medicines Agency (EMA), respectively. Dr. Judith Dunn outlined a novel approach for assessing and rank ordering patient and clinician clinical meaningfulness and the disconnect that may occur. Dr. Busner provided closing comments, tied together the presented issues, and provided a synopsis of the lively discussion that followed the session. In addition to the speakers above, the discussion included two representatives from patient advocacy groups, as well as an additional speaker who described the challenges of conducting a pediatric trial in the US and European Union (EU), given the often competing regulatory requirements. This article should serve as an expert-informed reference to those interested and involved in CNS drug development programs that are aimed at children and rare diseases and seek to ensure a patient-centric approach.
Collapse
Affiliation(s)
- Joan Busner
- Dr. Busner is with Signant Health in Blue Bell, Pennsylvania and Department of Psychiatry, Virginia Commonwealth University School of Medicine in Richmond, Virginia
| | - Gahan Pandina
- Dr. Pandina is with Janssen Pharmaceuticals in Titusville, New Jersey
| | - Simon Day
- Dr. Day is with Clinical Trials Consulting and Training, Ltd. in North Marston, England, United Kingdom. At the time of this writing
| | - Atul Mahableshwarkar
- Dr. Mahableshwarkar was with Emalex Biosciences; he is now with ARM Pharma Consulting in Deerfield, Illinois
| | | | - Maria Sheean
- Dr. Sheean is with the European Medicines Agency in Amsterdam, North Holland, Netherlands
| | - Judith Dunn
- Dr. Dunn is with Fulcrum Therapeutics in Cambridge, Massachusetts
| |
Collapse
|
9
|
Smits A, Annaert P, Cavallaro G, De Cock PAJG, de Wildt SN, Kindblom JM, Lagler FB, Moreno C, Pokorna P, Schreuder MF, Standing JF, Turner MA, Vitiello B, Zhao W, Weingberg AM, Willmann R, van den Anker J, Allegaert K. Current knowledge, challenges and innovations in developmental pharmacology: A combined conect4children Expert Group and European Society for Developmental, Perinatal and Paediatric Pharmacology White Paper. Br J Clin Pharmacol 2022; 88:4965-4984. [PMID: 34180088 PMCID: PMC9787161 DOI: 10.1111/bcp.14958] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/22/2021] [Accepted: 05/30/2021] [Indexed: 12/30/2022] Open
Abstract
Developmental pharmacology describes the impact of maturation on drug disposition (pharmacokinetics, PK) and drug effects (pharmacodynamics, PD) throughout the paediatric age range. This paper, written by a multidisciplinary group of experts, summarizes current knowledge, and provides suggestions to pharmaceutical companies, regulatory agencies and academicians on how to incorporate the latest knowledge regarding developmental pharmacology and innovative techniques into neonatal and paediatric drug development. Biological aspects of drug absorption, distribution, metabolism and excretion throughout development are summarized. Although this area made enormous progress during the last two decades, remaining knowledge gaps were identified. Minimal risk and burden designs allow for optimally informative but minimally invasive PK sampling, while concomitant profiling of drug metabolites may provide additional insight in the unique PK behaviour in children. Furthermore, developmental PD needs to be considered during drug development, which is illustrated by disease- and/or target organ-specific examples. Identifying and testing PD targets and effects in special populations, and application of age- and/or population-specific assessment tools are discussed. Drug development plans also need to incorporate innovative techniques such as preclinical models to study therapeutic strategies, and shift from sequential enrolment of subgroups, to more rational designs. To stimulate appropriate research plans, illustrations of specific PK/PD-related as well as drug safety-related challenges during drug development are provided. The suggestions made in this joint paper of the Innovative Medicines Initiative conect4children Expert group on Developmental Pharmacology and the European Society for Developmental, Perinatal and Paediatric Pharmacology, should facilitate all those involved in drug development.
Collapse
Affiliation(s)
- Anne Smits
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Neonatal intensive Care unit, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Giacomo Cavallaro
- Neonatal intensive care unit, Fondazione IRCCS Ca' Grande Ospedale Maggiore Policlinico, Milan, Italy
| | - Pieter A J G De Cock
- Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium.,Heymans Institute of Pharmacology, Ghent University, Ghent, Belgium.,Department of Pharmacy, Ghent University Hospital, Ghent, Belgium
| | - Saskia N de Wildt
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Pharmacology and Toxicology, Radboud Institute Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jenny M Kindblom
- Pediatric Clinical Research Center, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Florian B Lagler
- Institute for Inherited Metabolic Diseases and Department of Pediatrics, Paracelsus Medical University, Clinical Research Center Salzburg, Salzburg, Austria
| | - Carmen Moreno
- Institute of Psychiatry and Mental Health, Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Paula Pokorna
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Department of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.,Department of Physiology and Pharmacology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Amalia Children's Hospital, Nijmegen, the Netherlands
| | - Joseph F Standing
- UCL Great Ormond Street Institute of Child Health, London, UK.,Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Mark A Turner
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool Health Partners, Liverpool, UK
| | - Benedetto Vitiello
- Division of Child and Adolescent Neuropsychiatry, Department of Public Health and Pediatrics, University of Torino, Torino, Italy
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, China.,Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Clinical Research Centre, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | | | | | - John van den Anker
- Intensive Care and Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.,Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Department of Hospital Pharmacy, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
10
|
Bouazza N, Dokoumetzidis A, Knibbe CAJ, de Wildt SN, Ambery C, De Cock PA, Gasthuys E, Foissac F, Urien S, Hamberg AK, Poggesi I, Zhao W, Vermeulen A, Standing JF, Tréluyer JM. General clinical and methodological considerations on the extrapolation of pharmacokinetics and optimization of study protocols for small molecules and monoclonal antibodies in children. Br J Clin Pharmacol 2022; 88:4985-4996. [PMID: 36256514 DOI: 10.1111/bcp.15571] [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: 10/21/2021] [Revised: 09/05/2022] [Accepted: 09/20/2022] [Indexed: 12/01/2022] Open
Abstract
Pharmacometric modelling plays a key role in both the design and analysis of regulatory trials in paediatric drug development. Studies in adults provide a rich source of data to inform the paediatric investigation plans, including knowledge on drug pharmacokinetics (PK), safety and efficacy. In children, drug disposition differs widely from birth to adolescence but extrapolating adult to paediatric PK, safety and efficacy either with pharmacometric or physiologically based approaches can help design or in some cases reduce the need for clinical studies. Aspects to consider when extrapolating PK include the maturation of drug metabolizing enzyme expression, glomerular filtration, drug excretory systems, and the expression and activity of specific transporters in conjunction with other drug properties such as fraction unbound. Knowledge of these can be used to develop extrapolation tools such as allometric scaling plus maturation functions or physiologically based PK. PK/pharmacodynamic approaches and well-designed clinical trials in children are of key importance in paediatric drug development. In this white paper, state-of-the-art of current methods used for paediatric extrapolation will be discussed. This paper is part of a conect4children implementation of innovative methodologies including pharmacometric and physiologically based PK modelling in clinical trial design/paediatric drug development through dissemination of expertise and expert advice. The suggestions arising from this white paper should define a minimum set of standards in paediatric modelling and contribute to the regulatory science.
Collapse
Affiliation(s)
- Naïm Bouazza
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | | | - Catherijne A J Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Intensive Care and Paediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Claire Ambery
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline plc, London, UK
| | - Pieter A De Cock
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium.,Department of Pharmacy, Ghent University Hospital, Ghent, Belgium.,Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium
| | - Elke Gasthuys
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, University of Ghent, Ghent, Belgium
| | - Frantz Foissac
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | - Saïk Urien
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | - Anna-Karin Hamberg
- Department of Clinical Pharmacology, Uppsala University Hospital, Uppsala, Sweden
| | - Italo Poggesi
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Beerse, Belgium
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China.,Clinical Research Centre, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - An Vermeulen
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, University of Ghent, Ghent, Belgium
| | - Joseph F Standing
- Infection, Inflammation and Immunology, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Pharmacy, Great Ormond Street Hospital for Children, London, UK
| | - Jean-Marc Tréluyer
- Pediatric and Perinatal Drug Evaluation and Pharmacology, Université Paris Cité, Paris, France.,Unité de Recherche Clinique Université Paris Cité Necker-Cochin, AP-HP, Paris, France.,CIC-1419 Inserm, Cochin-Necker, Paris, France
| | | |
Collapse
|
11
|
Gross AS, Harry AC, Clifton CS, Pasqua OD. Clinical Trial Diversity: An Opportunity for Improved Insight into the Determinants of Variability in Drug Response. Br J Clin Pharmacol 2022; 88:2700-2717. [PMID: 35088432 PMCID: PMC9306578 DOI: 10.1111/bcp.15242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 11/27/2022] Open
Abstract
Although the number of countries participating in pivotal trials submitted to enable drug registration has nearly doubled over the past 25 years, there has not been a substantial increase in the diversity of clinical trial populations. In parallel, our understanding of factors that influence medicine response and variability has continued to evolve. The notion of intrinsic and extrinsic sources of variability has been embedded into different regulatory guidelines, including the recent guideline on the importance of enhancing the diversity of clinical trial populations. In addition to presenting the clinical and scientific reasons for ensuring that clinical trial populations represent the demographics of patient populations, this overview outlines the efforts of regulatory agencies, patient advocacy groups and clinical researchers to attain this goal through strategies to meet representation in recruitment targets and broaden eligibility criteria. Despite these efforts, challenges to participation in clinical trials remain, and certain groups continue to be underrepresented in development programmes. These challenges are amplified when the representativeness of specific groups may vary across countries and regions in a global clinical programme. Whilst enhanced trial diversity is a critical step towards ensuring that results will be representative of patient populations, a concerted effort is required to characterise further the factors influencing interindividual and regional differences in response for global populations. Quantitative clinical pharmacology principles should be applied to allow extrapolation of data across groups or regions as well as provide insight into the effect of patient‐specific characteristics on a medicine's dose rationale and efficacy and safety profiles.
Collapse
Affiliation(s)
- Annette S Gross
- Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline R&D, Sydney, Australia.,Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Anya C Harry
- Global Demographics & Diversity, Global Clinical Sciences and Delivery, GlaxoSmithKline R&D, Upper Providence, USA.,Current Address: West Pharmaceutical Services, King of Prussia, USA
| | - Christine S Clifton
- Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline R&D, Sydney, Australia
| | - Oscar Della Pasqua
- Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline R&D, Brentford, United Kingdom.,Clinical Pharmacology & Therapeutics Group, School of Pharmacy - University College London, London, UK
| |
Collapse
|
12
|
van Rongen A, Krekels EH, Calvier EA, de Wildt SN, Vermeulen A, Knibbe CA. An update on the use of allometric and other scaling methods to scale drug clearance in children: towards decision tables. Expert Opin Drug Metab Toxicol 2022; 18:99-113. [PMID: 35018879 DOI: 10.1080/17425255.2021.2027907] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION When pediatric data are not available for a drug, allometric and other methods are applied to scale drug clearance across the pediatric age-range from adult values. This is applied when designing first-in-child studies, but also for off-label drug prescription. AREAS COVERED This review provides an overview of the systematic accuracy of allometric and other pediatric clearance scaling methods compared to gold-standard PBPK predictions. The findings are summarized in decision tables to provide a priori guidance on the selection of appropriate pediatric clearance scaling methods for both novel drugs for which no pediatric data are available and existing drugs in clinical practice. EXPERT OPINION While allometric scaling principles are commonly used to scale pediatric clearance, there is no universal allometric exponent (i.e., 1, 0.75 or 0.67) that can accurately scale clearance for all drugs from adults to children of all ages. Therefore, pediatric scaling decision tables based on age, drug elimination route, binding plasma protein, fraction unbound, extraction ratio, and/or isoenzyme maturation are proposed to a priori select the appropriate (allometric) clearance scaling method, thereby reducing the need for full PBPK-based clearance predictions. Guidance on allometric scaling when estimating pediatric clearance values is provided as well.
Collapse
Affiliation(s)
- Anne van Rongen
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elke Hj Krekels
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Elisa Am Calvier
- Sanofi Pharmacokinetics-Dynamics and Metabolism (PKDM), Translational Medicine and Early Development, Sanofi R&D, Montpellier, France
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands.,Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - An Vermeulen
- Laboratory of Medical Biochemistry and Clinical Analysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium.,Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Catherijne Aj Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
13
|
Extrapolation as a Default Strategy in Pediatric Drug Development. Ther Innov Regul Sci 2022; 56:883-894. [PMID: 35006587 DOI: 10.1007/s43441-021-00367-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023]
Abstract
Pediatric drug development lags adult development by about 8 years (Mulugeta et al. in Pediatr Clin 64(6):1185-1196, 2017). In such context, many incentives, regulations, and innovative techniques have been proposed to address the disparity for pediatric patients. One such strategy is extrapolation of efficacy from a reference population. Extrapolation is currently justified by providing evidence in support of the effective use of drugs in children when the course of the disease and the expected treatment response would be sufficiently similar in the pediatric and reference population. This paper's position is that, despite uncertainties, pediatric drug development programs should initially assume some degree of extrapolation. The degree to which extrapolation can be used lies along a continuum representing the uncertainties to be addressed through generation of new pediatric evidence. In addressing these uncertainties, the extrapolation strategy should reflect the level of tolerable uncertainty concerning the decision to expose a child to the risks of a new drug. This judgment about the level of tolerable uncertainty should vary with the context (e.g., disease severity, existing therapeutic options) and can be embedded into pediatric drug development archetypes to ascertain the extent of studies needed and whether simultaneous development for adults and adolescents be considered.
Collapse
|
14
|
Higi L, Käser K, Wälti M, Grotzer M, Vonbach P. Description of a clinical decision support tool with integrated dose calculator for paediatrics. Eur J Pediatr 2022; 181:679-689. [PMID: 34524516 PMCID: PMC8821055 DOI: 10.1007/s00431-021-04261-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/02/2021] [Accepted: 09/05/2021] [Indexed: 12/02/2022]
Abstract
Medication errors, especially dosing errors are a leading cause of preventable harm in paediatric patients. The paediatric patient population is particularly vulnerable to dosing errors due to immaturity of metabolising organs and developmental changes. Moreover, the lack of clinical trial data or suitable drug forms, and the need for weight-based dosing, does not simplify drug dosing in paediatric or neonatal patients. Consequently, paediatric pharmacotherapy often requires unlicensed and off-label use including manipulation of adult dosage forms. In practice, this results in the need to calculate individual dosages which in turn increases the likelihood of dosing errors. In the age of digitalisation, clinical decision support (CDS) tools can support healthcare professionals in their daily work. CDS tools are currently amongst the gold standards in reducing preventable errors. In this publication, we describe the development and core functionalities of the CDS tool PEDeDose, a Class IIa medical device software certified according to the European Medical Device Regulation. The CDS tool provides a drug dosing formulary with an integrated calculator to determine individual dosages for paediatric, neonatal, and preterm patients. Even a technical interface is part of the CDS tool to facilitate integration into primary systems. This enables the support of the paediatrician directly during the prescribing process without changing the user interface.Conclusion: PEDeDose is a state-of-the-art CDS tool for individualised paediatric drug dosing that includes a certified calculator. What is Known: • Dosing errors are the most common type of medication errors in paediatric patients. • Clinical decision support tools can reduce medication errors effectively. Integration into the practitioner's workflow improves usability and user acceptance. What is New: • A clinical decision support tool with a certified integrated dosing calculator for paediatric drug dosing. • The tool was designed to facilitate integration into clinical information systems to directly support the prescribing process. Any clinical information system available can interoperate with the PEDeDose web service.
Collapse
Affiliation(s)
- Lukas Higi
- PEDeus Ltd, Zurich, Switzerland. .,Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| | | | | | - Michael Grotzer
- PEDeus Ltd, Zurich, Switzerland ,University Children’s Hospital of Zurich, Zurich, Switzerland
| | | |
Collapse
|
15
|
Esposito S, Carrothers TJ, Riccobene T, Stone GG, Kantecki M. Ceftaroline Fosamil for Treatment of Pediatric Complicated Skin and Soft Tissue Infections and Community-Acquired Pneumonia. Paediatr Drugs 2021; 23:549-563. [PMID: 34462863 PMCID: PMC8563558 DOI: 10.1007/s40272-021-00468-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
Community-acquired pneumonia (CAP)/community-acquired bacterial pneumonia (CABP) and complicated skin and soft tissue infection (cSSTI)/acute bacterial skin and skin structure infection (ABSSSI) represent major causes of morbidity and mortality in children. β-Lactams are the cornerstone of antibiotic treatment for many serious bacterial infections in children; however, most of these agents have no activity against methicillin-resistant Staphylococcus aureus (MRSA). Ceftaroline fosamil, a β-lactam with broad-spectrum in vitro activity against Gram-positive pathogens (including MRSA and multidrug-resistant Streptococcus pneumoniae) and common Gram-negative organisms, is approved in the European Union and the United States for children with CAP/CABP or cSSTI/ABSSSI. Ceftaroline fosamil has completed a pediatric investigation plan including safety, efficacy, and pharmacokinetic evaluations in patients with ages ranging from birth to 17 years. It has demonstrated similar clinical and microbiological efficacy to best available existing treatments in phase III-IV trials in patients aged ≥ 2 months to < 18 years with CABP or ABSSSI, with a safety profile consistent with the cephalosporin class. It is also approved in the European Union for neonates with CAP or cSSTI, and in the US for neonates with ABSSSI. Ceftaroline fosamil dosing for children (including renal function adjustments) is supported by pharmacokinetic/pharmacodynamic modeling and simulations in appropriate age groups, and includes the option of 5- to 60-min intravenous infusions for standard doses, and a high dose for cSSTI patients with MRSA isolates, with a ceftaroline minimum inhibitory concentration of 2-4 mg/L. Considered together, these data suggest ceftaroline fosamil may be beneficial in the management of CAP/CABP and cSSTI/ABSSSI in children.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Building 15, Via Gramsci 14, 43126, Parma, Italy.
| | | | | | | | | |
Collapse
|
16
|
Tabriz AG, Fullbrook DHG, Vilain L, Derrar Y, Nandi U, Grau C, Morales A, Hooper G, Hiezl Z, Douroumis D. Personalised Tasted Masked Chewable 3D Printed Fruit-Chews for Paediatric Patients. Pharmaceutics 2021; 13:pharmaceutics13081301. [PMID: 34452262 PMCID: PMC8400795 DOI: 10.3390/pharmaceutics13081301] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/18/2022] Open
Abstract
The development of personalised paediatric dosage forms using 3D printing technologies has gained significant interest over the last few years. In the current study extruded filaments of the highly bitter Diphenhydramine Hydrochloride (DPH) were fabricated by using suitable hydrophilic carries such as hydroxypropyl cellulose (Klucel ELFTM) and a non-ionic surfactant (Gelucire 48/16TM) combined with sweetener (Sucralose) and strawberry flavour grades. The thermoplastic filaments were used to print 3D fruit-chew designs by Fused Deposition Modelling (FDM) technology. Physicochemical characterisation confirmed the formation of glass solution where DPH was molecularly dispersed within the hydrophilic carriers. DPH was released rapidly from the 3D printed fruit-chew designs with >85% within the first 30 min. Trained panellists performed a full taste and sensory evaluation of the sweetener intensity and the strawberry aroma. The evaluation showed complete taste masking of the bitter DPH and revealed a synergistic effect of the sweetener and the strawberry flavour with enhanced sweet strawberry, fruity and aftertaste perception. The findings of the study can be used for the development of paediatric dosage forms with enhanced organoleptic properties, palatability and medication adherence.
Collapse
Affiliation(s)
- Atabak Ghanizadeh Tabriz
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent ME4 4TB, UK; (A.G.T.); (D.H.G.F.); (G.H.); (Z.H.)
| | - Daniel Henri George Fullbrook
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent ME4 4TB, UK; (A.G.T.); (D.H.G.F.); (G.H.); (Z.H.)
| | - Lilian Vilain
- Polytech Marseille, School of Engineering, Aix Marseille Université, 163 Avenue of Luminy, 13009 Marseille, France; (L.V.); (Y.D.)
| | - Youri Derrar
- Polytech Marseille, School of Engineering, Aix Marseille Université, 163 Avenue of Luminy, 13009 Marseille, France; (L.V.); (Y.D.)
| | - Uttom Nandi
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Chatham, Kent ME4 4TB, UK;
| | - Clara Grau
- School of Chemistry of Mulhouse (ENSCMu), University of Haute-Alsace (UHA), 3 Street Alfred Werner, 68093 Mulhouse, France; (C.G.); (A.M.)
| | - Anaïs Morales
- School of Chemistry of Mulhouse (ENSCMu), University of Haute-Alsace (UHA), 3 Street Alfred Werner, 68093 Mulhouse, France; (C.G.); (A.M.)
| | - Gemma Hooper
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent ME4 4TB, UK; (A.G.T.); (D.H.G.F.); (G.H.); (Z.H.)
| | - Zoltan Hiezl
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent ME4 4TB, UK; (A.G.T.); (D.H.G.F.); (G.H.); (Z.H.)
| | - Dennis Douroumis
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent ME4 4TB, UK; (A.G.T.); (D.H.G.F.); (G.H.); (Z.H.)
- Correspondence:
| |
Collapse
|
17
|
Acceptability of small-sized oblong tablets in comparison to syrup and mini-tablets in infants and toddlers: A randomized controlled trial. Eur J Pharm Biopharm 2021; 166:126-134. [PMID: 34153451 DOI: 10.1016/j.ejpb.2021.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/04/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE There is limited evidence for the acceptability of various drug formulations holding the potential to improve medicines administration to children. Suitable formulations need to meet the requirements of pediatric patients. Previous studies have demonstrated the acceptance of mini-tablets. Oblong tablets may carry more active ingredient content per unit than mini-tablets and could be an important alternative when the drug substance requires administration of higher doses. The primary objective was to demonstrate non-inferiority of acceptability of oblong tablets in comparison to 3 ml glucose syrup in children aged 1 to 5 years. Secondary objectives were investigation of acceptability, swallowability and palatability of mini-tablets, oblong tablets and glucose syrup in children between 1 and 5 years. METHODS An open, randomized, single dose two-way cross-over design in two parallel study arms was applied. 280 children were stratified to one of five age groups and randomized to receiving one oblong tablet (2.5 × 6 mm) in comparison either to 3 ml glucose syrup or to three mini-tablets (2 × 2 mm). Acceptability and swallowability were assessed according to pre-defined evaluation criteria. The application of the formulations was video documented to evaluate the palatability. RESULTS As primary objective, non-inferiority was observed regarding acceptability of the oblong tablet compared to syrup in all age groups (84.4% vs 80.1%, difference 4,29% points with 95% CI of -3.00%,11.57%). For swallowability, superiority of the oblong tablet compared to syrup could be shown (74.5% vs. 53.2%, difference 21.26% points, 95% CI of 11.29%, 31.23%). Regarding palatability, <10% of children demonstrated unpleasant reaction after intake of the oblong tablet or mini-tablets as graded by both raters, however, in contrast up to 40% of children after intake of syrup. CONCLUSION Oblong tablets are a promising, safe alternative to liquid drug formulations and administration of multiple mini-tablets in children.
Collapse
|
18
|
Chan PL, McFadyen L, Quaye A, Leister‐Tebbe H, Hendrick VM, Hammond J, Raber S. The use of extrapolation based on modeling and simulation to support high-dose regimens of ceftaroline fosamil in pediatric patients with complicated skin and soft-tissue infections. CPT Pharmacometrics Syst Pharmacol 2021; 10:551-563. [PMID: 33687148 PMCID: PMC8213416 DOI: 10.1002/psp4.12608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/14/2022] Open
Abstract
A model-informed drug development approach was used to select ceftaroline fosamil high-dose regimens for pediatric patients with complicated skin and soft-tissue infections caused by Staphylococcus aureus with a ceftaroline minimum inhibitory concentration (MIC) of 2 or 4 mg/L. Steady-state ceftaroline concentrations were simulated using a population pharmacokinetics (PK) model for ceftaroline fosamil and ceftaroline including data from 304 pediatric subjects and 944 adults. Probability of target attainment (PTA) for various simulated pediatric high-dose regimens and renal function categories were calculated based on patients achieving 35% fT>MIC (S. aureus PK/pharmacodynamic target for 2-log10 bacterial killing). For extrapolation of efficacy, simulated exposures and PTA were compared to adults with normal renal function receiving high-dose ceftaroline fosamil (600 mg 2-h infusions every 8 h). For safety, predicted ceftaroline exposures were compared with observed pediatric and adult data. Predicted ceftaroline exposures for the approved pediatric high-dose regimens (12, 10, or 8 mg/kg by 2-h infusions every 8 h for patients aged >2 to <18 years with normal/mild, moderate, or severe renal impairment, respectively; 10 mg/kg by 2-h infusions every 8 h for patients aged ≥2 months to <2 years with normal renal function/mild impairment) were well matched to adults with normal renal function. Median predicted maximum concentration at steady state (Cmax,ss ) and area under the plasma concentration-time curve over 24 h at steady state pediatric to adult ratios were 0.907-1.33 and 0.940-1.41, respectively. PTAs (>99% and ≥81% for MICs of 2 and 4 mg/L, respectively) matched or exceeded the adult predictions. Simulated Cmax,ss values were below the maximum observed data in other indications, including a high-dose pediatric pneumonia trial, which reported no adverse events related to high exposure.
Collapse
|
19
|
Bhatnagar M, Sheehan S, Sharma I, Baer G, Green D, McCune S, Joffe S, Snyder D. Prospect of Direct Benefit in Pediatric Trials: Practical Challenges and Potential Solutions. Pediatrics 2021; 147:peds.2020-049602. [PMID: 33906929 PMCID: PMC8262097 DOI: 10.1542/peds.2020-049602] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/16/2021] [Indexed: 11/24/2022] Open
Abstract
Clinical research in pediatric patients is necessary to develop safe and effective medicines for children. US Food and Drug Administration (FDA) human subject protection regulations (21 Code of Federal Regulations 50, subpart D) require that, with limited exceptions, research in children that exceeds a defined level of risk must offer a prospect of direct benefit to the individual child that is sufficient to justify those risks. Growing attention to the merits of initiating pediatric clinical trials earlier in the drug and biological product development process has led the FDA to look more closely at the meaning of the regulatory term prospect of direct benefit. In collaboration with the FDA, the Duke-Margolis Center for Health Policy convened a workshop with leading experts in the fields of biomedical ethics, pediatric clinical research, and pediatric product development, as well as patient representatives, to discuss the FDA's approach to characterizing prospect of direct benefit in the context of scientific advances in product development. Workshop topics included the extrapolation of adult efficacy data to children, use of nonclinical models of disease, use of modeling and simulation to support pediatric dosing, and reliance on biomarkers and surrogate end points in clinical research. Discussion from the workshop is provided herein to communicate the challenges that investigators, industry sponsors, regulators, and institutional review boards face when evaluating pediatric research and to outline several approaches to maximize prospect of direct benefit, minimize unnecessary risks and burden, and facilitate timely access to safe and effective medicines for children.
Collapse
Affiliation(s)
| | - Sarah Sheehan
- Duke-Margolis Center for Health Policy, Washington, District of Columbia; and
| | - Isha Sharma
- Duke-Margolis Center for Health Policy, Washington, District of Columbia
| | - Gerri Baer
- Food and Drug Administration, Silver Spring, Maryland
| | - Dionna Green
- Food and Drug Administration, Silver Spring, Maryland
| | - Susan McCune
- Food and Drug Administration, Silver Spring, Maryland
| | - Steven Joffe
- Division of Medical Ethics, Department of Medical Ethics and Health Policy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Donna Snyder
- Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
20
|
Hill-McManus D, Marshall S, Liu J, Willke RJ, Hughes DA. Linked Pharmacometric-Pharmacoeconomic Modeling and Simulation in Clinical Drug Development. Clin Pharmacol Ther 2020; 110:49-63. [PMID: 32936931 DOI: 10.1002/cpt.2051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022]
Abstract
Market access and pricing of pharmaceuticals are increasingly contingent on the ability to demonstrate comparative effectiveness and cost-effectiveness. As such, it is widely recognized that predictions of the economic potential of drug candidates in development could inform decisions across the product life cycle. This may be challenging when safety and efficacy profiles in terms of the relevant clinical outcomes are unknown or highly uncertain early in product development. Linking pharmacometrics and pharmacoeconomics, such that outputs from pharmacometric models serve as inputs to pharmacoeconomic models, may provide a framework for extrapolating from early-phase studies to predict economic outcomes and characterize decision uncertainty. This article reviews the published studies that have implemented this methodology and used simulation to inform drug development decisions and/or to optimize the use of drug treatments. Some of the key practical issues involved in linking pharmacometrics and pharmacoeconomics, including the choice of final outcome measures, methods of incorporating evidence on comparator treatments, approaches to handling multiple intermediate end points, approaches to quantifying uncertainty, and issues of model validation are also discussed. Finally, we have considered the potential barriers that may have limited the adoption of this methodology and suggest that closer alignment between the disciplines of clinical pharmacology, pharmacometrics, and pharmacoeconomics, may help to realize the potential benefits associated with linked pharmacometric-pharmacoeconomic modeling and simulation.
Collapse
Affiliation(s)
- Daniel Hill-McManus
- Centre for Health Economics and Medicines Evaluation, Bangor University, Bangor, UK
| | | | - Jing Liu
- Clinical Pharmacology, Pfizer Inc, Groton, Connecticut, USA
| | | | - Dyfrig A Hughes
- Centre for Health Economics and Medicines Evaluation, Bangor University, Bangor, UK
| |
Collapse
|
21
|
Conklin LS, Hoffman EP, van den Anker J. Developmental Pharmacodynamics and Modeling in Pediatric Drug Development. J Clin Pharmacol 2020; 59 Suppl 1:S87-S94. [PMID: 31502687 DOI: 10.1002/jcph.1482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
Challenges in pediatric drug development include small patient numbers, limited outcomes research, ethical barriers, and sparse biosamples. Increasingly, pediatric drug development is focusing on extrapolation: leveraging knowledge about adult disease and drug responses to inform projections of drug and clinical trial performance in pediatric subpopulations. Pharmacokinetic-pharmacodynamic (PK-PD) modeling and extrapolation aim to reduce the numbers of patients and data points needed to establish efficacy. Planning for PK-PD and biomarker studies should begin early in the adult drug development program. Extrapolation relies on the assumption that both the underlying disease and the mechanism of action of the drug used to treat that disease are similar in adults and pediatric subpopulations. Clearly, developmental changes in PK and PD need to be considered to enhance the quality of PK-PD modeling and, therefore, increase the success of extrapolation. This article focuses on the influence of differences in PD between adults and pediatric subpopulations that are highly relevant for the use of extrapolation.
Collapse
Affiliation(s)
- Laurie S Conklin
- Division of Gastroenterology, Hepatology, and Nutrition, Children's National Health System, Washington, DC, USA.,ReveraGen BioPharma, Rockville, MD, USA
| | - Eric P Hoffman
- ReveraGen BioPharma, Rockville, MD, USA.,Binghamton University-SUNY, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - John van den Anker
- ReveraGen BioPharma, Rockville, MD, USA.,Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA
| |
Collapse
|
22
|
Ollivier C, Sun H, Amchin W, Beghetti M, Berger RMF, Breitenstein S, Garnett C, Gullberg N, Hassel P, Ivy D, Kawut SM, Klein A, Lesage C, Migdal M, Nije B, Odermarsky M, Strait J, de Graeff PA, Stockbridge N. New Strategies for the Conduct of Clinical Trials in Pediatric Pulmonary Arterial Hypertension: Outcome of a Multistakeholder Meeting With Patients, Academia, Industry, and Regulators, Held at the European Medicines Agency on Monday, June 12, 2017. J Am Heart Assoc 2020; 8:e011306. [PMID: 31088189 PMCID: PMC6585335 DOI: 10.1161/jaha.118.011306] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
| | - Haihao Sun
- 2 Food and Drug Administration Silver Spring MD
| | | | - Maurice Beghetti
- 3 Pediatric Cardiology Unit Centre Universitaire de Cardiologie et Chirurgie Cardiaque Pédiatrique University Hospitals of Geneva City of Geneva Switzerland
| | - Rolf M F Berger
- 4 Center for Congenital Heart Diseases Department of Pediatric Cardiology Beatrix Children's Hospital University Medical Center Groningen University of Groningen the Netherlands
| | | | | | | | | | - Dunbar Ivy
- 7 Heart Institute Children's Hospital Colorado University of Colorado School of Medicine Aurora CO
| | - Steven M Kawut
- 8 Perelman School of Medicine University of Pennsylvania Philadelphia PA
| | | | | | - Marek Migdal
- 11 Childrens Memorial Health Institute Warsaw Poland
| | | | - Michal Odermarsky
- 12 Department of Paediatric Cardiology Paediatric Heart Center Lund University and Skåne University Hospital Lund Sweden
| | | | | | | |
Collapse
|
23
|
Eichler H, Koenig F, Arlett P, Enzmann H, Humphreys A, Pétavy F, Schwarzer‐Daum B, Sepodes B, Vamvakas S, Rasi G. Are Novel, Nonrandomized Analytic Methods Fit for Decision Making? The Need for Prospective, Controlled, and Transparent Validation. Clin Pharmacol Ther 2020; 107:773-779. [PMID: 31574163 PMCID: PMC7158212 DOI: 10.1002/cpt.1638] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022]
Abstract
Real-world data and patient-level data from completed randomized controlled trials are becoming available for secondary analysis on an unprecedented scale. A range of novel methodologies and study designs have been proposed for their analysis or combination. However, to make novel analytical methods acceptable for regulators and other decision makers will require their testing and validation in broadly the same way one would evaluate a new drug: prospectively, well-controlled, and according to a pre-agreed plan. From a European regulators' perspective, the established methods qualification advice procedure with active participation of patient groups and other decision makers is an efficient and transparent platform for the development and validation of novel study designs.
Collapse
Affiliation(s)
- Hans‐Georg Eichler
- European Medicines Agency (EMA)AmsterdamThe Netherlands
- Medical University of ViennaViennaAustria
| | | | - Peter Arlett
- European Medicines Agency (EMA)AmsterdamThe Netherlands
| | - Harald Enzmann
- Federal Institute for Drugs and Medical Devices (BfArM)BonnGermany
- EMA's Committee for Medicinal Products for Human Use (CHMP)AmsterdamThe Netherlands
| | | | - Frank Pétavy
- European Medicines Agency (EMA)AmsterdamThe Netherlands
| | - Brigitte Schwarzer‐Daum
- Medical University of ViennaViennaAustria
- EMA's Committee for Orphan Medicinal Products (COMP)AmsterdamThe Netherlands
| | - Bruno Sepodes
- EMA's Committee for Medicinal Products for Human Use (CHMP)AmsterdamThe Netherlands
- EMA's Committee for Orphan Medicinal Products (COMP)AmsterdamThe Netherlands
- Universidade de LisboaLisbonPortugal
| | | | - Guido Rasi
- European Medicines Agency (EMA)AmsterdamThe Netherlands
- University Tor VergataRomeItaly
| |
Collapse
|
24
|
Burckart GJ, Seo S, Pawlyk AC, McCune SK, Yao LP, Giacoia GP, Wang Y, Zineh I. Scientific and Regulatory Considerations for an Ontogeny Knowledge Base for Pediatric Clinical Pharmacology. Clin Pharmacol Ther 2020; 107:707-709. [PMID: 31983072 PMCID: PMC7875197 DOI: 10.1002/cpt.1763] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
Understanding all aspects of developmental biology, or pediatric ontogeny, that affect drug therapy from the fetus to the adolescent child is the holy grail of pediatric scientists and clinical pharmacologists. The scientific community is now close to being able to tie together the vast amount of information collected on pediatric ontogeny over the past 60 years. An organized knowledge base and new tools would allow us to utilize this information effectively in pediatric drug development.
Collapse
Affiliation(s)
- Gilbert J. Burckart
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Shirley Seo
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Aaron C. Pawlyk
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Susan K. McCune
- Office of Pediatric Therapeutics, Office of the Commissioner, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lynne P. Yao
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - George P. Giacoia
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Yaning Wang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Issam Zineh
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
25
|
Kopp‐Schneider A, Calderazzo S, Wiesenfarth M. Power gains by using external information in clinical trials are typically not possible when requiring strict type I error control. Biom J 2020; 62:361-374. [PMID: 31265159 PMCID: PMC7079072 DOI: 10.1002/bimj.201800395] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/30/2022]
Abstract
In the era of precision medicine, novel designs are developed to deal with flexible clinical trials that incorporate many treatment strategies for multiple diseases in one trial setting. This situation often leads to small sample sizes in disease-treatment combinations and has fostered the discussion about the benefits of borrowing of external or historical information for decision-making in these trials. Several methods have been proposed that dynamically discount the amount of information borrowed from historical data based on the conformity between historical and current data. Specifically, Bayesian methods have been recommended and numerous investigations have been performed to characterize the properties of the various borrowing mechanisms with respect to the gain to be expected in the trials. However, there is common understanding that the risk of type I error inflation exists when information is borrowed and many simulation studies are carried out to quantify this effect. To add transparency to the debate, we show that if prior information is conditioned upon and a uniformly most powerful test exists, strict control of type I error implies that no power gain is possible under any mechanism of incorporation of prior information, including dynamic borrowing. The basis of the argument is to consider the test decision function as a function of the current data even when external information is included. We exemplify this finding in the case of a pediatric arm appended to an adult trial and dichotomous outcome for various methods of dynamic borrowing from adult information to the pediatric arm. In conclusion, if use of relevant external data is desired, the requirement of strict type I error control has to be replaced by more appropriate metrics.
Collapse
Affiliation(s)
| | - Silvia Calderazzo
- Division of BiostatisticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Manuel Wiesenfarth
- Division of BiostatisticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
26
|
Menditto E, Orlando V, De Rosa G, Minghetti P, Musazzi UM, Cahir C, Kurczewska-Michalak M, Kardas P, Costa E, Sousa Lobo JM, Almeida IF. Patient Centric Pharmaceutical Drug Product Design-The Impact on Medication Adherence. Pharmaceutics 2020; 12:E44. [PMID: 31947888 PMCID: PMC7023035 DOI: 10.3390/pharmaceutics12010044] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
Medication adherence is a growing concern for public health and poor adherence to therapy has been associated with poor health outcomes and higher costs for patients. Interventions for improving adherence need to consider the characteristics of the individual therapeutic regimens according to the needs of the patients. In particular, geriatric and paediatric populations as well as dermatological patients have special needs/preferences that should be considered when designing drug products. Patient Centric Drug Product Pharmaceutical Design (PCDPD) offers the opportunity to meet the needs and preferences of patients. Packaging, orodispersible formulations, fixed dose combinations products, multiparticulate formulations, topical formulations and 3D printing are of particular relevance in a PCDPD process. These will be addressed in this review as well as their impact on medication adherence.
Collapse
Affiliation(s)
- Enrica Menditto
- CIRFF, Centre of Pharmacoeconomics, Department of Pharmacy, University of Naples Federico II, Corso Umberto I, 40, 80138 Napoli NA, Italy; (E.M.); (V.O.)
| | - Valentina Orlando
- CIRFF, Centre of Pharmacoeconomics, Department of Pharmacy, University of Naples Federico II, Corso Umberto I, 40, 80138 Napoli NA, Italy; (E.M.); (V.O.)
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II Corso Umberto I, 40, 80138 Napoli NA, Italy;
| | - Paola Minghetti
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via G. Colombo 71, 20133 Milan, Italy; (P.M.); (U.M.M.)
| | - Umberto Maria Musazzi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via G. Colombo 71, 20133 Milan, Italy; (P.M.); (U.M.M.)
| | - Caitriona Cahir
- Division of Population Health Sciences, Royal College of Surgeons, Beaux Lane House, Mercer Street, Dublin 2, Ireland;
| | - Marta Kurczewska-Michalak
- Department of Family Medicine, Medical University of Lodz, 60, Narutowicza St., 90-136 Lodz, Poland; (M.K.-M.); (P.K.)
| | - Przemysław Kardas
- Department of Family Medicine, Medical University of Lodz, 60, Narutowicza St., 90-136 Lodz, Poland; (M.K.-M.); (P.K.)
| | - Elísio Costa
- UCIBIO/REQUIMTE, Faculty of Pharmacy and Porto4Ageing, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
| | - José Manuel Sousa Lobo
- UCIBIO/REQUIMTE, MedTech-Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
| | - Isabel F Almeida
- UCIBIO/REQUIMTE, MedTech-Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
| |
Collapse
|
27
|
Liu T, Lewis TR, Moore JN, Kraft WK, Gauda EB, Sartori D, Moody DE, Gobburu JVS, Ivaturi V. Could Postnatal Age-Related Uridine Diphosphate Glucuronic Acid Be a Rate-Limiting Factor in the Metabolism of Morphine During the First Week of Life? CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:469-477. [PMID: 31044547 PMCID: PMC6656938 DOI: 10.1002/psp4.12407] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/13/2019] [Indexed: 02/06/2023]
Abstract
Neonates experience dramatic changes in the disposition of drugs after birth as a result of enzyme maturation and environmental adjustment, challenging therapeutic decision making. In this research, we establish postnatal age, postmenstrual age, and body weight as physiologically reasonable predictors of morphine's clearance in neonates. By integrating knowledge of bilirubin, morphine, and other drugs metabolized by glucuronidation pathways from previously published studies, we hypothesize that uridine diphosphate glucuronic acid, a postnatal age-dependent sugar, plays an important role in the metabolism of morphine during the first week of life. This finding can be extended to other drugs metabolized by uridine diphosphate glucuronosyltransferase pathways in neonates and thus has important clinical implications for the use of drugs in this population.
Collapse
Affiliation(s)
- Tao Liu
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Tamorah R Lewis
- Division of Neonatology, Department of Pediatrics, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Jason N Moore
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Walter K Kraft
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Estelle B Gauda
- Division of Neonatology, Department of Pediatrics, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - David Sartori
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David E Moody
- Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Jogarao V S Gobburu
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Vijay Ivaturi
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Ollivier C, Thomson A, Manolis E, Blake K, Karlsson KE, Knibbe CA, Pons G, Hemmings R. Commentary on the EMA Reflection Paper on the use of extrapolation in the development of medicines for paediatrics. Br J Clin Pharmacol 2019; 85:659-668. [PMID: 30707770 PMCID: PMC6422728 DOI: 10.1111/bcp.13883] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/12/2022] Open
Abstract
Adopted guidelines reflect a harmonised European approach to a specific scientific issue and should reflect the most recent scientific knowledge. However, whilst EU regulations are mandatory for all member states and EU directives must be followed by national laws in line with the directive, EMA guidelines do not have legal force and alternative approaches may be taken, but these obviously require more justification. This new series of the BJCP, developed in collaboration with the EMA, aims to address this issue by providing an annotated version of some relevant EMA guidelines and regulatory documents by experts. Hopefully, this will help in promoting their diffusion and in opening a forum for discussion with our readers.
Collapse
Affiliation(s)
- Cécile Ollivier
- Human Medicines Research & Development Support DivisionEuropean Medicines AgencyLondonUK
| | - Andrew Thomson
- Human Medicines Research & Development Support DivisionEuropean Medicines AgencyLondonUK
| | - Efthymios Manolis
- Human Medicines Research & Development Support DivisionEuropean Medicines AgencyLondonUK
| | - Kevin Blake
- Human Medicines Research & Development Support DivisionEuropean Medicines AgencyLondonUK
| | - Kristin E. Karlsson
- Department of Efficacy and SafetySwedish Medicinal Products AgencyUppsalaSweden
| | - Catherijne A.J. Knibbe
- Department of Clinical PharmacySt. Antonius HospitalNieuwegeinThe Netherlands
- Faculty of Science, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | | | - Robert Hemmings
- Licensing DivisionMedicines and Healthcare products Regulatory AgencyLondonUK
| |
Collapse
|