1
|
Shubow S, Gunsior M, Rosenberg A, Wang YM, Altepeter T, Guinn D, Rajabiabhari M, Kotarek J, Mould DR, Zhou H, Cheifetz AS, Garces S, Chevalier R, Gavan S, Trusheim MR, Rispens T, Bray K, Partridge MA. Therapeutic Drug Monitoring of Biologics: Current Practice, Challenges and Opportunities - a Workshop Report. AAPS J 2025; 27:62. [PMID: 40087239 DOI: 10.1208/s12248-025-01050-9] [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: 01/02/2025] [Accepted: 02/23/2025] [Indexed: 03/17/2025] Open
Abstract
Therapeutic drug monitoring (TDM) for dose modification of biologics has the potential to improve patient outcomes. The US Food and Drug Administration (FDA) and the American Association of Pharmaceutical Scientists (AAPS) hosted the first US-based public workshop on TDM of biologics with contributions from a broad array of interested parties including healthcare providers, clinical pharmacologists, test developers, bioanalysis and immunogenicity scientists, health economics and outcomes research (HEOR) experts and regulators. The key insight was that despite a body of evidence to support TDM in certain therapeutic areas, there remain substantial challenges to widespread clinical implementation. There is a lack of consensus regarding the integration of TDM in clinical guidelines, and a lack of consensus on the cost-effectiveness of TDM; both factors contribute to the difficulty that healthcare providers face in obtaining reimbursement for TDM (both coverage of testing itself, and coverage of potential dosing modifications). The HEOR experts outlined alternative routes to obtaining reimbursement and suggested advocating for changes in coverage policies to promote TDM use in the clinic. Reaching alignment across policy makers, patients and advocacy groups, payers, and healthcare providers, on specific treatment settings where TDM will be clearly beneficial, was identified as an important step to advancing TDM implementation for the benefit of patients.
Collapse
Affiliation(s)
- Sophie Shubow
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Yow-Ming Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Tara Altepeter
- Division of Gastroenterology, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Daphne Guinn
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Joseph Kotarek
- Office of Health Technology 7, Office of Product Evaluation and Quality, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Diane R Mould
- Projections Research Inc., Phoenixville, Pennsylvania, USA
| | - Honghui Zhou
- Jazz pharmaceuticals, Philadelphia, Pennsylvania, USA
| | - Adam S Cheifetz
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Rachel Chevalier
- Children's Mercy Kansas City, University of Missouri-Kansas City (UMKC), Kansas City, USA
| | - Sean Gavan
- Manchester Centre for Health Economics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | | | - Theo Rispens
- Amsterdam institute for Immunology and Infectious diseases, Immunology, Amsterdam, Netherlands
| | | | | |
Collapse
|
2
|
Kort J, Naleid N, Oley F, Ignatz-Hoover J, Margevicius S, Fu P, Malek E, Cooper B. Melphalan 140 mg/m 2 is Safe and Effective for Frail and Older Multiple Myeloma Patients With Comparable Rates of Minimal Residual Disease Negativity. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025:S2152-2650(25)00068-0. [PMID: 40090795 DOI: 10.1016/j.clml.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/13/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Despite therapeutic advances, multiple myeloma (MM) remains challenging to treat effectively. High-dose melphalan (Mel200) with autologous stem cell transplantation (ASCT) is the standard treatment for transplant-eligible patients. Reduced-dose melphalan (Mel140) is an alternative for older or frail patients, yet its efficacy data remain unclear. METHODS We retrospectively analyzed 233 MM patients undergoing first ASCT between 2014 and 2022, comparing outcomes between Mel140 (n = 82) and Mel200 (n = 151). We assessed patient demographics, disease characteristics, progression-free survival (PFS), and overall survival (OS). In an exploratory subset analysis achievement of MRD from bone marrow samples after ASCT was compared between the 2 groups. RESULTS As expected, patients who received Mel 140 were significantly older with a higher KPS. Median follow-up was 47.7 months. Both groups had similar rates of readmissions and infections within the first 100 days after transplant despite Mel140 group being older with more comorbidities. No significant difference in PFS or OS was observed between Mel140 and Mel200 groups (P > .05). MRD negativity rates at sensitivity levels of 10-5 and 10-6 were comparable (64% vs. 60%, P = .7). Patients achieving sustained MRD negativity demonstrated improved PFS regardless of melphalan dose. CONCLUSION Our findings suggest equivalent efficacy and safety profiles between Mel140 and Mel200, supporting Mel140 as a viable option for older or frail MM patients. In a subset analysis equivalent rates of MRD were achieved between the groups and remained a highly significant predictor of PFS, highlighting its relevance regardless of dosing strategies.
Collapse
Affiliation(s)
- Jeries Kort
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH.
| | - Nikolas Naleid
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Frank Oley
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - James Ignatz-Hoover
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Seunghee Margevicius
- Case Western Reserve University, Department of Population and Quantitative Health Sciences, Cleveland, OH
| | - Pingfu Fu
- Case Western Reserve University, Department of Population and Quantitative Health Sciences, Cleveland, OH
| | - Ehsan Malek
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Brenda Cooper
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
3
|
Samuels A, Irie K, Mizuno T, Reifenberg J, Punt N, Vinks AA, Minar P. Integrating early response biomarkers in pharmacokinetic models: A novel method to individualize the initial infliximab dose in patients with Crohn's disease. Clin Transl Sci 2025; 18:e70086. [PMID: 39985779 PMCID: PMC11846607 DOI: 10.1111/cts.70086] [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: 07/31/2024] [Revised: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 02/24/2025] Open
Abstract
The use of model-informed precision dosing to personalize infliximab has been shown to improve both the acquisition of concentration targets and clinical outcomes during maintenance. Current iterations of infliximab pharmacokinetic models include time-varying covariates of drug clearance, however, not accounting for the expected improvements in the covariates can lead to indiscriminate use of higher infliximab doses and imprecise drug exposure. The aim was to identify changes in the four biomarkers associated with infliximab clearance (Xiong et al. model) and determine if integration of these dynamic changes would improve model performance during induction and early maintenance. We analyzed two cohorts of children receiving infliximab for Crohn's Disease. The Emax method was used to assess time-varying changes in covariates. Model performance (observed vs. predicted infliximab concentrations) was evaluated using median percentage error (bias) and median absolute percentage error (precision). The combined cohorts included 239 Crohn's disease patients. We found from baseline to dose 4, the maximum changes in weight, albumin, erythrocyte sedimentation rate, and neutrophil CD64 were 4.7%, +11.7%, -62.4%, and -26.5%, respectively. We also found the use of baseline covariates alone to forecast future trough concentration was inferior to the Emax time-varying method with a significant improvement observed in bias (doses 2, 3, and 4) and precision (doses 2 and 4). The integration of the four time-varying biomarkers of drug clearance with pharmacokinetic modeling improved the accuracy and precision of the predictions. This novel strategy may be key to improving drug exposure, minimizing indiscriminate dosing strategies, and reducing healthcare costs.
Collapse
Affiliation(s)
- Abigail Samuels
- Department of Internal Medicine, Department of Veterans AffairsUniversity of Cincinnati School of MedicineCincinnatiOhioUSA
| | - Kei Irie
- Division of Translational and Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Tomoyuki Mizuno
- Division of Translational and Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Jack Reifenberg
- University of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Nieko Punt
- MedimaticsMaastrichtThe Netherlands
- Department of Clinical Pharmacy and PharmacologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Alexander A. Vinks
- Division of Translational and Clinical PharmacologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Phillip Minar
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Gastroenterology, Hepatology and NutritionCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
4
|
Mould DR, Upton RN. "Getting the Dose Right"-Revisiting the Topic With Focus on Biologic Agents. Clin Pharmacol Ther 2024; 116:613-618. [PMID: 38680029 DOI: 10.1002/cpt.3285] [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: 01/29/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Nearly two decades after the Peck and Cross article '"Getting the dose right: facts, a blueprint, and encouragements" was published, a review of dose recommendations for biologics shows that the success in getting the dose right appears to have improved given the relatively low incidence of drug withdrawals and dosing/label changes. However, the clinical experience with monoclonal antibodies (MAbs) following approval has been less than perfect. In inflammatory diseases, the disease burden changes with time and high treatment failure rates have been reported. In addition, the use of concomitant steroids and immunosuppressant drugs with MAbs is common. These concomitant agents have their own safety issues and many immunosuppressant agents are not well-tolerated although they have been shown to reduce the incidence of anti-drug antibodies (ADA). This same complexity is seen in MAbs used in oncology as well, although with these agents the doses appear to be higher than needed, which results in high treatment costs and incidence of adverse events. Given the complexity of MAb pharmacokinetics, which makes providing a detailed description of dose options difficult, product labeling should include the options for alternative dose strategies and potentially include the use of therapeutic drug monitoring with dose individualization which have been shown to improve clinical response and reduce the incidence of ADA. So, while the recommended dosing for biologics seems improved over the issues noted 17 years ago, we still have some work to do.
Collapse
Affiliation(s)
- Diane R Mould
- Projections Research Inc, Phoenixville, Pennsylvania, USA
| | - Richard N Upton
- Projections Research Inc, Phoenixville, Pennsylvania, USA
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
5
|
Komenkul V, Sukarnjanaset W, Komolmit P, Wattanavijitkul T. External validation of population pharmacokinetic models of tacrolimus in Thai adult liver transplant recipients. Eur J Clin Pharmacol 2024; 80:1229-1240. [PMID: 38695888 DOI: 10.1007/s00228-024-03692-8] [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: 01/09/2024] [Accepted: 04/17/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Several population pharmacokinetic models of tacrolimus in liver transplant patients were built, and their predictability was evaluated in their settings. However, the extrapolation in the prediction was unclear. This study aimed to evaluate the predictive performance of published tacrolimus models in adult liver transplant recipients using data from the Thai population as an external dataset. METHODS The selected published models were systematically searched and evaluated for their quality. The external dataset of patients who underwent the first liver transplant and received immediate-release tacrolimus was used to assess the predictive performance of each selected model. Trough concentrations between 3 and 6 months were retrospectively collected to evaluate the predictability of each model using prediction-based diagnostics, simulation-based diagnostics, and Bayesian forecasting. RESULTS Sixty-seven patients with 360 trough concentrations and eight selected published models were included in this study. None of the models met the predictive precision criteria in prediction-based diagnostics. Meanwhile, four published population pharmacokinetic models showed a normal distribution in NPDE testing. Regarding Bayesian forecasting, all models improved their forecasts with at least one prior information data point. CONCLUSION Bayesian forecasting is more accurate and precise than other testing methods for predicting drug concentrations. However, none of the evaluated models provides satisfactory predictive performance for generalization to Thai liver transplant patients. This underscores the need for future research to develop population PK models tailored to the Thai population. Such efforts should consider the inclusion of nonlinear pharmacokinetics and region-specific factors, including genetic variability, to improve model accuracy and applicability.
Collapse
Affiliation(s)
- Virunya Komenkul
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Waroonrat Sukarnjanaset
- Department of Pharmaceutical Care, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Piyawat Komolmit
- Division of Gastro-enterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Liver Diseases, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Thitima Wattanavijitkul
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
6
|
Wang W, Zhang Q, Zhao J, Liu T, Yao J, Peng X, Zhi M, Zhang M. HLA-DQA1*05 correlates with increased risk of anti-drug antibody development and reduced response to infliximab in Chinese patients with Crohn's disease. Gastroenterol Rep (Oxf) 2024; 12:goae074. [PMID: 39055374 PMCID: PMC11269678 DOI: 10.1093/gastro/goae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/30/2024] [Accepted: 05/07/2024] [Indexed: 07/27/2024] Open
Abstract
Background The efficacy of anti-TNF therapy in Crohn's disease (CD), such as infliximab, is often compromised by the development of anti-drug antibodies (ADAs). The genetic variation HLA-DQA1*05 has been linked to the immunogenicity of biologics, influencing ADA formation. This study investigates the correlation between HLA-DQA1*05 and ADA formation in CD patients treated with infliximab in a Chinese Han population and assesses clinical outcomes. Methods In this retrospective cohort study, 345 infliximab-exposed CD patients were genotyped for HLADQ A1*05A > G (rs2097432). We evaluated the risk of ADA development, loss of infliximab response, adverse events, and treatment discontinuation among variant and wild-type allele individuals. Results A higher percentage of patients with ADAs formation was observed in HLA-DQA1*05 G variant carriers compared with HLA-DQA1*05 wild-type carriers (58.5% vs 42.9%, P = 0.004). HLA-DQA1*05 carriage significantly increased the risk of ADAs development (adjusted hazard ratio = 1.65, 95% CI 1.18-2.30, P = 0.003) and was associated with a greater likelihood of infliximab response loss (adjusted HR = 2.55, 95% CI 1.78-3.68, P < 0.0001) and treatment discontinuation (adjusted HR = 2.21, 95% CI 1.59-3.06, P < 0.0001). Interestingly, combined therapy with immunomodulators increased the risk of response loss in HLA-DQA1*05 variant carriers. Conclusions HLA-DQA1*05 significantly predicts ADAs formation and impacts treatment outcomes in infliximab-treated CD patients. Pre-treatment screening for this genetic factor could therefore be instrumental in personalizing anti-TNF therapy strategies for these patients.
Collapse
Affiliation(s)
- Wei Wang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| | - Qi Zhang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| | - Junzhang Zhao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| | - Tao Liu
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| | - Jiayin Yao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| | - Xiang Peng
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| | - Min Zhi
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| | - Min Zhang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510655, P. R. China
| |
Collapse
|
7
|
Kisembo HN, Malumba R, Sematimba H, Ankunda R, Nalweyiso ID, Malwadde EK, Rutebemberwa E, Kasasa S, Salama DH, Kawooya MG. Understanding the factors that influence CT utilization for mild traumatic brain injury in a low resource setting - a qualitative study using the Theoretical Domains Framework. Afr J Emerg Med 2024; 14:103-108. [PMID: 38756826 PMCID: PMC11096711 DOI: 10.1016/j.afjem.2024.04.004] [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: 08/17/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction In low resource settings (LRS), utilization of Computed Tomography scan (CTS) for mild traumatic brain injuries (mTBIs) presents unique challenges and considerations given the limited infrastructure, financial resources, and trained personnel. The Theoretical Domains Framework (TDF) offers a comprehensive theoretical lens to explore factors influencing the decision-making to order CTS for mTBI by imaging referrers (IRs). Objectives The primary objective was to explore IRs' beliefs about factors influencing CT utilization in mTBIs using TDF in Uganda.Differences in the factors influencing CTS ordering behavior across specialties, levels of experience, and hospital category were also explored. Materials and Methods In-depth semi-structured interviews guided by TDF were conducted among purposively selected IRs from 6 tertiary public and private hospitals with functional CTS services. A thematic analysis was performed with codes and emerging themes developed based on the TDF. Results Eleven IRs including medical officers, non-neurosurgeon specialists and neurosurgeons aged on average 42 years (SD+/-12.3 years) participated.Identified factors within skills domain involved IRs' clinical assessment and decision-making abilities, while beliefs about capabilities and consequences encompassed their confidence in diagnostic abilities and perceptions of CTS risks and benefits. The environmental context and resources domain addressed the availability of CT scanners and financial constraints. The knowledge domain elicited IRs' understanding of clinical guidelines and evidence-based practices while social influences considered peer influence and institutional culture. For memory, attention & decision processes domain, IRs adherence to guidelines and intentions to order CT scans were cited. Conclusion Using TDF, IRs identified several factors believed to influence decision making to order CTS in mTBI in a LRS. The findings can inform stakeholders to develop targeted strategies and evidence-based interventions to optimize CT utilization in mTBI such as; educational programs, workflow modifications, decision support tools, and infrastructure improvements, among others.
Collapse
Affiliation(s)
- Harriet Nalubega Kisembo
- Makerere University, College of Health Sciences, School of Medicine
- Department of Radiology, Mulago National Referral and Teaching Hospital, Kampala, Uganda
| | - Richard Malumba
- Ernest cook Ultrasound Research and Education Institute, Mengo Hospital, Kampala, Uganda
| | - Henry Sematimba
- Ernest cook Ultrasound Research and Education Institute, Mengo Hospital, Kampala, Uganda
| | - Racheal Ankunda
- Ernest cook Ultrasound Research and Education Institute, Mengo Hospital, Kampala, Uganda
| | | | - Elsie-Kiguli Malwadde
- African Centre for Global Health and Social Transformation (ACHEST), Kampala, Uganda
| | - Elizeus Rutebemberwa
- School of Public Health, Department of Health Policy & Management, Makerere University, Kampala, Uganda
| | - Simon Kasasa
- Department of Epidemiology & Biostatistics, School of Public Health, Makerere University, Kampala, Uganda
| | | | - Michael Grace Kawooya
- Ernest cook Ultrasound Research and Education Institute, Mengo Hospital, Kampala, Uganda
| |
Collapse
|
8
|
Bullock GS, Duncan P, Chandler AM, Aguilar AA, Latham N, Storer T, Alexander N, McDonough CM. Development of an exercise therapy referral clinical support tool for patients with osteoporosis at risk for falls. J Am Geriatr Soc 2024; 72:1810-1816. [PMID: 38344943 DOI: 10.1111/jgs.18796] [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: 08/28/2023] [Revised: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 06/19/2024]
Abstract
BACKGROUND The purpose of this study was to develop a clinical support tool for osteoporosis clinic providers to support risk assessment and referrals for evidence-based exercise therapy programs. METHODS A sequential Delphi method was used with a multidisciplinary group of national falls experts, to provide consensus on referral to exercise therapy for patients at risk for falls. The Delphi study included a primary research team, expert panel, and clinical partners to answer the questions: (1) "What patient characteristics are needed to develop a clinical support tool?"; (2) "What are the recommended exercise referrals for patients with osteoporosis at risk for falls?" The consensus process consisted of two rounds with 8 weeks between meetings. Two qualitative researchers analyzed the data using a modified version of a matrix analysis approach. RESULTS The following were the most important variables to include when determining exercise therapy referrals for patients with osteoporosis: Patient history and demographics, falls history over the last year, current physical function and balance, caregiver and transportation status, socioeconomic and insurance status, and patient preference. Potential exercise therapy referrals included one-on-one physical therapy, group physical therapy, home health, community-based exercise programs, and not acceptable for exercise therapy. CONCLUSIONS Patient characteristics including patient history, physical function and balance performance, socioeconomic and insurance status, and patient preference for exercise therapy are important to inform both the medical provider and patient with osteoporosis to choose the most appropriate exercise therapy referral. Adoption of the algorithmic suggestions may have a significant impact on uptake and adherence to exercise therapy, ultimately improving patient physical function and reducing falls risk.
Collapse
Affiliation(s)
- Garrett S Bullock
- Department of Orthopaedic Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Pamela Duncan
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Alison M Chandler
- Qualitative and Patient Reported Outcomes Shared Resource, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Aylin A Aguilar
- Qualitative and Patient Reported Outcomes Shared Resource, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Nancy Latham
- Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tom Storer
- Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Neil Alexander
- Geriatric and Palliative Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christine M McDonough
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Dako F, Cook T, Zafar H, Schnall M. Population Health Management in Radiology: Economic Considerations. J Am Coll Radiol 2023; 20:962-968. [PMID: 37597716 DOI: 10.1016/j.jacr.2023.07.016] [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: 05/17/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/21/2023]
Abstract
There is a growing emphasis on population health management (PHM) in the United States, in part because it has the worst health outcomes indices among high-income countries despite spending by far the most on health care. Successful PHM is expected to lead to a healthier population with reduced health care utilization and cost. The role of radiology in PHM is increasingly being recognized, including efforts in care coordination, secondary prevention, and appropriate imaging utilization, among others. To further discuss economic considerations for PHM, we must understand the evolving health care payer environment, which combines fee-for-service and increasingly, an alternative payment model framework developed by the Health Care Payment Learning and Action Network. In considering the term "value-based care," perceived value needs to accrue to those who ultimately pay for care, which is more commonly employers and the government. This perspective drives the design of alternative payment models and thus should be taken into consideration to ensure sustainable practice models.
Collapse
Affiliation(s)
- Farouk Dako
- Director of the Center for Global and Population Health Research in Radiology, Department of Radiology, Senior Fellow, Leonard Davis Institute of Health Economics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.
| | - Tessa Cook
- Vice Chair, Practice Transformation, Department of Radiology, Senior Fellow, Leonard Davis Institute of Health Economics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Hanna Zafar
- Vice Chair, Quality, Department of Radiology, Senior Fellow, Leonard Davis Institute of Health Economics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Mitchell Schnall
- Chairman and Eugene P. Pendergrass Professor of Radiology, Department of Radiology, Senior Fellow, Leonard Davis Institute of Health Economics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Nasr A, Minar P. The Role of Therapeutic Drug Monitoring in Children. Gastroenterol Clin North Am 2023; 52:549-563. [PMID: 37543399 PMCID: PMC10865141 DOI: 10.1016/j.gtc.2023.05.002] [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] [Indexed: 08/07/2023]
Abstract
The use of biologic therapies has changed the treatment landscape for children with inflammatory bowel disease. While the novel biologics have improved clinical outcomes, there remains a significant gap in achieving endoscopic remission, prolonged steroid-free remission, and drug durability. Contributing to this gap is the paucity of real-world pharmacokinetic studies in children and a failure to dose optimize therapy during induction. Emerging data from a pediatric clinical trial and several observational studies have shown that the combination of proactive therapeutic drug monitoring and achievement of early therapeutic concentrations is effective in achieving improved outcomes. The next steps will be to leverage these past studies to develop more innovative clinical trials to properly assess the safety and effectiveness of proactive therapeutic drug monitoring in children.
Collapse
Affiliation(s)
- Alexander Nasr
- Division of Pediatric Gastroenterology, Hepatology & Nutrition, Cincinnati Children's Hospital Medical Center, MLC 2010, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Phillip Minar
- Division of Pediatric Gastroenterology, Hepatology & Nutrition, Cincinnati Children's Hospital Medical Center, MLC 2010, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
11
|
Oda K, Saito H, Jono H. Bayesian prediction-based individualized dosing of anti-methicillin-resistant Staphylococcus aureus treatment: Recent advancements and prospects in therapeutic drug monitoring. Pharmacol Ther 2023; 246:108433. [PMID: 37149156 DOI: 10.1016/j.pharmthera.2023.108433] [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: 12/26/2022] [Revised: 04/19/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
As one of the efficient techniques for TDM, the population pharmacokinetic (popPK) model approach for dose individualization has been developed due to the rapidly growing innovative progress in computer technology and has recently been considered as a part of model-informed precision dosing (MIPD). Initial dose individualization and measurement followed by maximum a posteriori (MAP)-Bayesian prediction using a popPK model are the most classical and widely used approach among a class of MIPD strategies. MAP-Bayesian prediction offers the possibility of dose optimization based on measurement even before reaching a pharmacokinetically steady state, such as in an emergency, especially for infectious diseases requiring urgent antimicrobial treatment. As the pharmacokinetic processes in critically ill patients are affected and highly variable due to pathophysiological disturbances, the advantages offered by the popPK model approach make it highly recommended and required for effective and appropriate antimicrobial treatment. In this review, we focus on novel insights and beneficial aspects of the popPK model approach, especially in the treatment of infectious diseases with anti-methicillin-resistant Staphylococcus aureus agents represented by vancomycin, and discuss the recent advancements and prospects in TDM practice.
Collapse
Affiliation(s)
- Kazutaka Oda
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Hideyuki Saito
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan; Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University; 1-1-1, Honjo, Chuo-ku, Kumamoto, Japan
| | - Hirofumi Jono
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan; Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University; 1-1-1, Honjo, Chuo-ku, Kumamoto, Japan.
| |
Collapse
|
12
|
Desai DC, Dherai AJ, Strik A, Mould DR. Personalized Dosing of Infliximab in Patients With Inflammatory Bowel Disease Using a Bayesian Approach: A Next Step in Therapeutic Drug Monitoring. J Clin Pharmacol 2023; 63:480-489. [PMID: 36458468 DOI: 10.1002/jcph.2189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022]
Abstract
Although biological agents have revolutionized the management of inflammatory bowel diseases (IBDs), a significant proportion of patients show primary non-response or develop secondary loss of response. Therapeutic drug monitoring (TDM) is advocated to maintain the efficacy of biologic agents. Reactive TDM can rationalize the management of primary non-response and secondary loss of response and has shown to be more cost-effective compared with empiric dose escalation. Proactive TDM is shown to increase clinical remission and the durability of the response to a biologic agent. However, the efficacy of proactive and reactive TDM has been questioned in recent studies and meta-analyses. Hence, we need a different approach to TDM, which addresses inflammatory burden, the individual patient, and disease factors. Bayesian approaches, which use population pharmacokinetic models, enable clinicians to make better use of TDM for dose adjustment. With rapid improvement in computer technology, these Bayesian model-based software packages are now available for clinical use. Bayesian dashboard systems allow clinicians to apply model-based dosing to understand an individual's pharmacokinetics and achieve a target serum drug concentration. The model is updated using previously measured drug concentrations and relevant patient factors, such as body weight, C-reactive protein, and serum albumin concentration, to maintain effective drug concentrations in the serum. Initial studies have found utility for the Bayesian approach in induction and maintenance, in adult and pediatric patients, in clinical trials, and in real-life situations for patients with IBD treated with infliximab. This needs confirmation in larger studies. This article reviews the Bayesian approach to therapeutic drug monitoring in IBD.
Collapse
Affiliation(s)
- Devendra C Desai
- Division of Gastroenterology, PD Hinduja Hospital, Veer Savarkar Marg, Mahim, Mumbai, India
| | - Alpa J Dherai
- Department of Laboratory Medicine, PD Hinduja Hospital, Veer Savarkar Marg, Mahim, Mumbai, India
| | - Anne Strik
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, Amsterdam, the Netherlands
| | - Diane R Mould
- Projections Research Inc., Phoenixville, Pennsylvania, USA
| |
Collapse
|
13
|
Huang H, Liu Q, Zhang X, Xie H, Liu M, Chaphekar N, Wu X. External Evaluation of Population Pharmacokinetic Models of Busulfan in Chinese Adult Hematopoietic Stem Cell Transplantation Recipients. Front Pharmacol 2022; 13:835037. [PMID: 35873594 PMCID: PMC9300831 DOI: 10.3389/fphar.2022.835037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/17/2022] [Indexed: 11/30/2022] Open
Abstract
Objective: Busulfan (BU) is a bi-functional DNA-alkylating agent used in patients undergoing hematopoietic stem cell transplantation (HSCT). Over the last decades, several population pharmacokinetic (pop PK) models of BU have been established, but external evaluation has not been performed for almost all models. The purpose of the study was to evaluate the predictive performance of published pop PK models of intravenous BU in adults using an independent dataset from Chinese HSCT patients, and to identify the best model to guide personalized dosing. Methods: The external evaluation methods included prediction-based diagnostics, simulation-based diagnostics, and Bayesian forecasting. In prediction-based diagnostics, the relative prediction error (PE%) was calculated by comparing the population predicted concentration (PRED) with the observations. Simulation-based diagnostics included the prediction- and variability-corrected visual predictive check (pvcVPC) and the normalized prediction distribution error (NPDE). Bayesian forecasting was executed by giving prior one to four observations. The factors influencing the model predictability, including the impact of structural models, were assessed. Results: A total of 440 concentrations (110 patients) were obtained for analysis. Based on prediction-based diagnostics and Bayesian forecasting, preferable predictive performance was observed in the model developed by Huang et al. The median PE% was -1.44% which was closest to 0, and the maximum F20 of 57.27% and F30 of 72.73% were achieved. Bayesian forecasting demonstrated that prior concentrations remarkably improved the prediction precision and accuracy of all models, even with only one prior concentration. Conclusion: This is the first study to comprehensively evaluate published pop PK models of BU. The model built by Huang et al. had satisfactory predictive performance, which can be used to guide individualized dosage adjustment of BU in Chinese patients.
Collapse
Affiliation(s)
- Huiping Huang
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Qingxia Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiaohan Zhang
- College of Arts and Sciences, University of Virginia, Charlottesville, VA, United States
| | - Helin Xie
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- *Correspondence: Xuemei Wu, ; Maobai Liu,
| | - Nupur Chaphekar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xuemei Wu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- *Correspondence: Xuemei Wu, ; Maobai Liu,
| |
Collapse
|
14
|
Hattingh HL, Michaleff ZA, Fawzy P, Du L, Willcocks K, Tan KM, Keijzers G. Ordering of computed tomography scans for head and cervical spine: a qualitative study exploring influences on doctors' decision-making. BMC Health Serv Res 2022; 22:790. [PMID: 35717206 PMCID: PMC9206095 DOI: 10.1186/s12913-022-08156-2] [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: 09/17/2021] [Accepted: 06/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ordering of computed tomography (CT) scans needs to consideration of diagnostic utility as well as resource utilisation and radiation exposure. Several factors influence ordering decisions, including evidence-based clinical decision support tools to rule out serious disease. The aim of this qualitative study was to explore factors influencing Emergency Department (ED) doctors' decisions to order CT of the head or cervical spine. METHODS In-depth semi-structured interviews were conducted with purposively selected ED doctors from two affiliated public hospitals. An interview tool with 10 questions, including three hypothetical scenarios, was developed and validated to guide discussions. Interviews were audio recorded, transcribed verbatim, and compared with field notes. Transcribed data were imported into NVivo Release 1.3 to facilitate coding and thematic analysis. RESULTS In total 21 doctors participated in semi-structured interviews between February and December 2020; mean interview duration was 35 min. Data saturation was reached. Participants ranged from first-year interns to experienced consultants. Five overarching emerging themes were: 1) health system and local context, 2) work structure and support, 3) professional practices and responsibility, 4) reliable patient information, and 5) holistic patient-centred care. Mapping of themes and sub-themes against a behaviour change model provided a basis for future interventions. CONCLUSIONS CT ordering is complex and multifaceted. Multiple factors are considered by ED doctors during decisions to order CT scans for head or c-spine injuries. Increased education on the use of clinical decision support tools and an overall strategy to improve awareness of low-value care is needed. Strategies to reduce low-yield CT ordering will need to be sustainable, sophisticated and supportive to achieve lasting change.
Collapse
Affiliation(s)
- H Laetitia Hattingh
- Diagnostic and Sub-Specialty Services, Gold Coast Health, Southport, Gold Coast, QLD, 4215, Australia. .,School of Pharmacy and Medical Sciences, Griffith University, Southport, Gold Coast, QLD, 4222, Australia.
| | | | - Peter Fawzy
- Neurosurgery Department, Gold Coast Health, Southport, Gold Coast, QLD, 4215, Australia.,School of Medicine and Health Sciences, Bond University, Gold Coast, QLD, 4226, Australia
| | - Leanne Du
- Medical Imaging, Gold Coast Health, Southport, Gold Coast, QLD, 4215, Australia
| | - Karlene Willcocks
- Diagnostic and Sub-Specialty Services, Gold Coast Health, Southport, Gold Coast, QLD, 4215, Australia
| | - K Meng Tan
- Diagnostic and Sub-Specialty Services, Gold Coast Health, Southport, Gold Coast, QLD, 4215, Australia
| | - Gerben Keijzers
- Department of Emergency Medicine, Gold Coast Health, Southport, Gold Coast, QLD, 4215, Australia.,Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, 4226, Australia.,School of Medicine, Griffith University, Southport, Gold Coast, QLD, 4222, Australia
| |
Collapse
|
15
|
Uster DW, Wicha SG. Optimized sampling to estimate vancomycin drug exposure: Comparison of pharmacometric and equation-based approaches in a simulation-estimation study. CPT Pharmacometrics Syst Pharmacol 2022; 11:711-720. [PMID: 35259285 PMCID: PMC9197536 DOI: 10.1002/psp4.12782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/31/2022] Open
Abstract
Vancomycin dosing should be accompanied by area under the concentration‐time curve (AUC)–guided dosing using model‐informed precision dosing software according to the latest guidelines. Although a peak plus a trough sample is considered the gold standard to determine the AUC, single‐sample strategies might be more economic. Yet, optimal sampling times for AUC determination of vancomycin have not been systematically evaluated. In the present study, automated one‐ or two‐sample strategies were systematically explored to estimate the AUC with a model averaging and a model selection algorithm. Both were compared with a conventional equation‐based approach in a simulation‐estimation study mimicking a heterogenous patient population (n = 6000). The optimal single‐sample timepoints were identified between 2–6.5 h post dose, with varying bias values between −2.9% and 1.0% and an imprecision of 23.3%–24.0% across the population pharmacokinetic approaches. Adding a second sample between 4.5–6.0 h improved the predictive performance (−1.7% to 0.0% bias, 17.6%–18.6% imprecision), although the difference in the two‐sampling strategies were minor. The equation‐based approach was always positively biased and hence inferior to the population pharmacokinetic approaches. In conclusion, the approaches always preferred samples to be drawn early in the profile (<6.5 h), whereas sampling of trough concentrations resulted in a higher imprecision. Furthermore, optimal sampling during the early treatment phase could already give sufficient time to individualize the second dose, which is likely unfeasible using trough sampling.
Collapse
Affiliation(s)
- David W Uster
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| |
Collapse
|
16
|
Lee S, Song M, Lim W, Song E, Han J, Kim BH. Development and Validation of Open-Source R Package HMCtdm for Therapeutic Drug Monitoring. Pharmaceuticals (Basel) 2022; 15:127. [PMID: 35215240 PMCID: PMC8875672 DOI: 10.3390/ph15020127] [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: 11/01/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 12/01/2022] Open
Abstract
Most therapeutic drug monitoring (TDM) packages are based on the maximum a posteriori (MAP) estimation. In this study, HMCtdm, a new TDM package, was developed using a Hamiltonian Monte Carlo (HMC) simulation. The estimation process of HMCtdm for the drugs amikacin, vancomycin, theophylline, and phenytoin was based on the R package Torsten. The prior pharmacokinetic (PK) models of the drugs were derived from the Abbottbase® pharmacokinetics systems (PKS) program. The performance of HMCtdm for each drug was assessed through internal and external validations. The internal validation results of the HMCtdm were compared with those of a MAP-based estimation. The developed open-source HMCtdm package is user friendly. The validation results were reviewed and interpreted using the mean percentage error and root mean squared error. The successful transplantation of the prior PK structures (used in PKS) was confirmed by comparing the validation results with a MAP estimation. An open-source HMC-based TDM package was also successfully developed in this study, and its performance was evaluated. This package can be operated by users unfamiliar with C++ and can be further developed for various applications.
Collapse
Affiliation(s)
- Sooyoung Lee
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - Moonsik Song
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (M.S.); (W.L.)
| | - Woojae Lim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (M.S.); (W.L.)
| | - Eunjung Song
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Medical Center, Seoul 02447, Korea;
| | - Jongdae Han
- Department of Computer Science, Sangmyung University, Seoul 03016, Korea;
| | - Bo-Hyung Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (M.S.); (W.L.)
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Medical Center, Seoul 02447, Korea;
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
17
|
Corral Alaejos Á, Zarzuelo Castañeda A, Jiménez Cabrera S, Sánchez-Guijo F, Otero MJ, Pérez-Blanco JS. External evaluation of population pharmacokinetic models of imatinib in adults diagnosed with chronic myeloid leukaemia. Br J Clin Pharmacol 2021; 88:1913-1924. [PMID: 34705297 DOI: 10.1111/bcp.15122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/29/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
AIMS Imatinib is considered the standard first-line treatment in newly diagnosed patients with chronic-phase myeloid leukaemia (CML). Several imatinib population pharmacokinetic (popPK) models have been developed. However, their predictive performance has not been well established when extrapolated to different populations. Therefore, this study aimed to perform an external evaluation of available imatinib popPK models developed mainly in adult patients, and to evaluate the improvement in individual model-based predictions through Bayesian forecasting computed by each model at different treatment occasions. METHODS A literature review was conducted through PubMed and Scopus to identify popPK models. Therapeutic drug monitoring data collected in adult CML patients treated with imatinib was used for external evaluation, including prediction- and simulated-based diagnostics together with Bayesian forecasting analysis. RESULTS Fourteen imatinib popPK studies were included for model-performance evaluation. A total of 99 imatinib samples were collected from 48 adult CML patients undergoing imatinib treatment with a minimum of one plasma concentration measured at steady-state between January 2016 and December 2020. The model proposed by Petain et al showed the best performance concerning prediction-based diagnostics in the studied population. Bayesian forecasting demonstrated a significant improvement in predictive performance at the second visit. Inter-occasion variability contributed to reducing bias and improving individual model-based predictions. CONCLUSIONS Imatinib popPK studies developed in Caucasian subjects including α1-acid glycoprotein showed the best model performance in terms of overall bias and precision. Moreover, two imatinib samples from different visits appear sufficient to reach an adequate model-based individual prediction performance trough Bayesian forecasting.
Collapse
Affiliation(s)
| | | | | | - Fermín Sánchez-Guijo
- Institute for Biomedical Research of Salamanca, Salamanca, Spain.,Haematology Department, University Hospital of Salamanca, Salamanca, Spain.,Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María José Otero
- Pharmacy Service, University Hospital of Salamanca, Salamanca, Spain.,Institute for Biomedical Research of Salamanca, Salamanca, Spain
| | - Jonás Samuel Pérez-Blanco
- Department of Pharmaceutical Sciences, Pharmacy Faculty, University of Salamanca, Salamanca, Spain.,Institute for Biomedical Research of Salamanca, Salamanca, Spain
| |
Collapse
|
18
|
Monoclonal Antibody Monitoring: Clinically Relevant Aspects, A Systematic Critical Review. Ther Drug Monit 2021; 42:45-56. [PMID: 31365482 DOI: 10.1097/ftd.0000000000000681] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Monoclonal antibody (mAb) therapy does not usually lead to a clinical response in all patients and resistance may increase over time after repeated mAb administration. This lack or loss of response to the treatment may originate from different and little-known epigenetic, biomolecular, or pathophysiological mechanisms, although an inadequate serum concentration is perhaps the most likely cause, even if not widely recognized and investigated yet. Patient factors that influence the pharmacokinetics (PK) of a mAb should be taken into account. Multiple analyses of patient-derived PK data have identified various factors influencing the clearance of mAbs. These factors include the presence of antidrug antibodies, low serum albumin, high serum levels of C-reactive protein, high body weight, and gender differences among others. The same clearance processes involved in systemic clearance after intravenous administration are also involved in local first-pass catabolism after subcutaneous administration of mAbs. Therapeutic drug monitoring has been proposed as a way to understand and respond to the variability in clinical response and remission. For both classes of mAbs with anti-inflammatory and antitumor effects, dose-guided optimization based on the measurement of serum concentrations in individual patients could be the next step for a personalized and targeted mAb therapy.
Collapse
|
19
|
Liebchen U, Klose M, Paal M, Vogeser M, Zoller M, Schroeder I, Schmitt L, Huisinga W, Michelet R, Zander J, Scharf C, Weinelt FA, Kloft C. Evaluation of the MeroRisk Calculator, A User-Friendly Tool to Predict the Risk of Meropenem Target Non-Attainment in Critically Ill Patients. Antibiotics (Basel) 2021; 10:468. [PMID: 33924047 PMCID: PMC8074046 DOI: 10.3390/antibiotics10040468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The MeroRisk-calculator, an easy-to-use tool to determine the risk of meropenem target non-attainment after standard dosing (1000 mg; q8h), uses a patient's creatinine clearance and the minimum inhibitory concentration (MIC) of the pathogen. In clinical practice, however, the MIC is rarely available. The objectives were to evaluate the MeroRisk-calculator and to extend risk assessment by including general pathogen sensitivity data. METHODS Using a clinical routine dataset (155 patients, 891 samples), a direct data-based evaluation was not feasible. Thus, in step 1, the performance of a pharmacokinetic model was determined for predicting the measured concentrations. In step 2, the PK model was used for a model-based evaluation of the MeroRisk-calculator: risk of target non-attainment was calculated using the PK model and agreement with the MeroRisk-calculator was determined by a visual and statistical (Lin's concordance correlation coefficient (CCC)) analysis for MIC values 0.125-16 mg/L. The MeroRisk-calculator was extended to include risk assessment based on EUCAST-MIC distributions and cumulative-fraction-of-response analysis. RESULTS Step 1 showed a negligible bias of the PK model to underpredict concentrations (-0.84 mg/L). Step 2 revealed a high level of agreement between risk of target non-attainment predictions for creatinine clearances >50 mL/min (CCC = 0.990), but considerable deviations for patients <50 mL/min. For 27% of EUCAST-listed pathogens the median cumulative-fraction-of-response for the observed patients receiving standard dosing was < 90%. CONCLUSIONS The MeroRisk-calculator was successfully evaluated: For patients with maintained renal function it allows a reliable and user-friendly risk assessment. The integration of pathogen-based risk assessment substantially increases the applicability of the tool.
Collapse
Affiliation(s)
- Uwe Liebchen
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany; (U.L.); (M.K.); (L.S.); (R.M.); (F.A.W.)
- Department of Anaesthesiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (M.Z.); (I.S.); (C.S.)
| | - Marian Klose
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany; (U.L.); (M.K.); (L.S.); (R.M.); (F.A.W.)
| | - Michael Paal
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (M.P.); (M.V.); (J.Z.)
| | - Michael Vogeser
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (M.P.); (M.V.); (J.Z.)
| | - Michael Zoller
- Department of Anaesthesiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (M.Z.); (I.S.); (C.S.)
| | - Ines Schroeder
- Department of Anaesthesiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (M.Z.); (I.S.); (C.S.)
| | - Lisa Schmitt
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany; (U.L.); (M.K.); (L.S.); (R.M.); (F.A.W.)
- Graduate Research Training Program PharMetrX, Freie Universität Berlin, 12169 Berlin, Germany
- Graduate Research Training Program PharMetrX, Universität Potsdam, 14476 Potsdam, Germany
| | - Wilhelm Huisinga
- Institute of Mathematics, Universität Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany;
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany; (U.L.); (M.K.); (L.S.); (R.M.); (F.A.W.)
| | - Johannes Zander
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (M.P.); (M.V.); (J.Z.)
- Laboratory Dr. Brunner, Luisenstr. 7e, 78464 Konstanz, Germany
| | - Christina Scharf
- Department of Anaesthesiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (M.Z.); (I.S.); (C.S.)
| | - Ferdinand A. Weinelt
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany; (U.L.); (M.K.); (L.S.); (R.M.); (F.A.W.)
- Graduate Research Training Program PharMetrX, Freie Universität Berlin, 12169 Berlin, Germany
- Graduate Research Training Program PharMetrX, Universität Potsdam, 14476 Potsdam, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Kelchstr. 31, 12169 Berlin, Germany; (U.L.); (M.K.); (L.S.); (R.M.); (F.A.W.)
| |
Collapse
|
20
|
Ryu S, Jung WJ, Jiao Z, Chae JW, Yun HY. External evaluation of the predictive performance of seven population pharmacokinetic models for phenobarbital in neonates. Br J Clin Pharmacol 2021; 87:3878-3889. [PMID: 33638184 DOI: 10.1111/bcp.14803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/09/2021] [Accepted: 02/13/2021] [Indexed: 02/06/2023] Open
Abstract
AIM Several studies have reported population pharmacokinetic models for phenobarbital (PB), but the predictive performance of these models has not been well documented. This study aims to do external evaluation of the predictive performance in published pharmacokinetic models. METHODS Therapeutic drug monitoring data collected in neonates and young infants treated with PB for seizure control was used for external evaluation. A literature review was conducted through PubMed to identify population pharmacokinetic models. Prediction- and simulation-based diagnostics, and Bayesian forecasting were performed for external evaluation. The incorporation of allometric scaling for body size and maturation factors into the published models was also tested for prediction improvement. RESULTS A total of 79 serum concentrations from 28 subjects were included in the external dataset. Seven population pharmacokinetic studies of PB were identified as relevant in the literature search and included for our evaluation. The model by Voller et al showed the best performance concerning prediction-based evaluation. In simulation-based analyses, the normalized prediction distribution error of two models (those of Shellhaas et al and Marsot et al) obeyed a normal distribution. Bayesian forecasting with more than one observation improved predictive capability. Incorporation of both allometric size scaling and maturation function generally enhanced the predictive performance, with improvement as observed in the model of Vucicevic et al. CONCLUSIONS: The predictive performance of published pharmacokinetic models of PB was diverse. Bayesian forecasting and incorporation of both size and maturation factors could improve the predictability of the models for neonates.
Collapse
Affiliation(s)
- Sunae Ryu
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea.,National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Republic of Korea
| | - Woo Jin Jung
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jung-Woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
21
|
Xiong Y, Mizuno T, Colman R, Hyams J, Noe JD, Boyle B, Tsai YT, Dong M, Jackson K, Punt N, Rosen MJ, Denson LA, Vinks AA, Minar P. Real-World Infliximab Pharmacokinetic Study Informs an Electronic Health Record-Embedded Dashboard to Guide Precision Dosing in Children with Crohn's Disease. Clin Pharmacol Ther 2021; 109:1639-1647. [PMID: 33354765 DOI: 10.1002/cpt.2148] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 12/20/2022]
Abstract
Standard-of-care infliximab dosing regimens were developed prior to the routine use of therapeutic drug monitoring and identification of target concentrations. Not surprisingly, subtherapeutic infliximab concentrations in pediatric Crohn's disease (CD) are common. The primary aim was to conduct a real-world pharmacokinetic (PK) evaluation to discover blood biomarkers of rapid clearance, identify exposure targets, and a secondary aim to translate PK modeling to the clinic. In a multicenter observational study, 671 peak and trough infliximab concentrations from 78 patients with CD were analyzed with a drug-tolerant assay (Esoterix; LabCorp, Calabasas, CA). Individual area under the curve (AUC) estimates were generated as a measure of drug exposure over time. Population PK modeling (nonlinear mixed-effect modeling) identified serum albumin, antibody to infliximab, erythrocyte sedimentation rate (ESR), and neutrophil CD64 as biomarkers for drug clearance. Week 14 and week 52 biochemical remitters (fecal calprotectin < 250 µg/g) had higher infliximab exposure (AUC) throughout induction. The optimal infliximab AUC target during induction for week 14 biochemical remission was 79,348 µg*h/mL (area under the receiver operating characteristic curve (AUROC) 0.77, [0.63-0.90], 85.7% sensitive, and 64.3% specific) with those exceeding the AUC target more likely to achieve a surgery-free week 52 biochemical remission (OR 4.3, [1.2-14.6]). Pretreatment predictors for subtherapeutic week 14 AUC included neutrophil CD64 > 6 (OR 4.5, [1.4-17.8]), ESR > 30 mm/h (OR 3.8, [1.4-11]), age < 10 years old (OR 4.2, [1.2-20]), and weight < 30 kg (OR 6.6, [2.1-25]). We created a decision-support PK dashboard with an iterative process and embedded the modeling program within the electronic health record. Model-informed precision dosing guided by real-world PKs is now available at the bedside in real-time.
Collapse
Affiliation(s)
- Ye Xiong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ruben Colman
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jeffrey Hyams
- Connecticut Children's Medical Center, Hartford, Connecticut, USA
| | - Joshua D Noe
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Yi-Ting Tsai
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kimberly Jackson
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Michael J Rosen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lee A Denson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Phillip Minar
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
22
|
Wojtyniak J, Selzer D, Schwab M, Lehr T. Physiologically Based Precision Dosing Approach for Drug‐Drug‐Gene Interactions: A Simvastatin Network Analysis. Clin Pharmacol Ther 2020; 109:201-211. [DOI: 10.1002/cpt.2111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/07/2020] [Indexed: 12/25/2022]
Affiliation(s)
- Jan‐Georg Wojtyniak
- Clinical Pharmacy Saarland University Saarbrücken Germany
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical Pharmacology Stuttgart Germany
| | - Dominik Selzer
- Clinical Pharmacy Saarland University Saarbrücken Germany
| | - Matthias Schwab
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical Pharmacology Stuttgart Germany
- Departments of Clinical Pharmacology and Pharmacy and Biochemistry University of Tübingen Tübingen Germany
- Cluster of Excellence iFIT (EXC2180) "Image‐guided and Functionally Instructed Tumor Therapies" University of Tübingen Tübingen Germany
| | - Thorsten Lehr
- Clinical Pharmacy Saarland University Saarbrücken Germany
| |
Collapse
|
23
|
Brockmeyer JM, Wise RT, Burgener EB, Milla C, Frymoyer A. Area under the curve achievement of once daily tobramycin in children with cystic fibrosis during clinical care. Pediatr Pulmonol 2020; 55:3343-3350. [PMID: 32827334 DOI: 10.1002/ppul.25037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND The area under the concentration-time curve over 24 hours (AUC24 ) is frequently utilized to monitor tobramycin exposure in children with cystic fibrosis (CF). An understanding of exposure target achievement during clinical implementation of an AUC24 based approach in children is limited. METHODS A retrospective chart review was performed in children with CF treated with once daily tobramycin and drug concentration monitoring at a pediatric CF center. During clinical care AUC24 was estimated using a traditional log-linear regression approach (LLR). AUC24 was also estimated retrospectively using a pharmacokinetic model-based Bayesian forecasting approach (BF). AUC24 achievement after both approaches were compared. RESULTS In 77 treatment courses (mean age, 12.7 ± 5.0 years), a target AUC24 100 to 125 mg h/L was achieved after starting dose in 21 (27%) and after initial dose adjustment in 35 (45%). In the first 7 days of treatment, 24 (32%) required ≥3 dose adjustments, and the mean number of drug concentrations measured was 7.1 ± 3.2. Examination of a BF approach demonstrated adequate prediction of measured tobramycin concentrations (median bias -2.1% [95% CI -3.1 to -1.4]; median precision 7.6% [95% CI, 7.1%-8.2%]). AUC24 estimates utilizing the BF approach were higher than the LLR approach with a mean difference of 6.4 mg h/L (95% CI, 4.8 to 8.0 mg h/L). CONCLUSIONS Achievement of a narrow AUC24 target is challenging during clinical care, and dose individualization is needed in most children with CF. Implementing a BF approach for estimating AUC24 in children with CF is supported.
Collapse
Affiliation(s)
- Jake M Brockmeyer
- Department of Pharmacy, Lucile Packard Children's Hospital Stanford, Palo Alto, California
| | - Russell T Wise
- Department of Pharmacy, Lucile Packard Children's Hospital Stanford, Palo Alto, California
| | - Elizabeth B Burgener
- Division of Pediatric Pulmonary Medicine, Stanford University, Stanford, California
| | - Carlos Milla
- Division of Pediatric Pulmonary Medicine, Stanford University, Stanford, California
| | - Adam Frymoyer
- Department of Pediatrics, Stanford University, Stanford, California
| |
Collapse
|
24
|
Uster DW, Stocker SL, Carland JE, Brett J, Marriott DJE, Day RO, Wicha SG. A Model Averaging/Selection Approach Improves the Predictive Performance of Model-Informed Precision Dosing: Vancomycin as a Case Study. Clin Pharmacol Ther 2020; 109:175-183. [PMID: 32996120 DOI: 10.1002/cpt.2065] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/12/2020] [Indexed: 11/10/2022]
Abstract
Many important drugs exhibit substantial variability in pharmacokinetics and pharmacodynamics leading to a loss of the desired clinical outcomes or significant adverse effects. Forecasting drug exposures using pharmacometric models can improve individual target attainment when compared with conventional therapeutic drug monitoring (TDM). However, selecting the "correct" model for this model-informed precision dosing (MIPD) is challenging. We derived and evaluated a model selection algorithm (MSA) and a model averaging algorithm (MAA), which automates model selection and finds the best model or combination of models for each patient using vancomycin as a case study, and implemented both algorithms in the MIPD software "TDMx." The predictive performance (based on accuracy and precision) of the two algorithms was assessed in (i) a simulation study of six distinct populations and (ii) a clinical dataset of 180 patients undergoing TDM during vancomycin treatment and compared with the performance obtained using a single model. Throughout the six virtual populations the MSA and MAA (imprecision: 9.9-24.2%, inaccuracy: less than ± 8.2%) displayed more accurate predictions than the single models (imprecision: 8.9-51.1%; inaccuracy: up to 28.9%). In the clinical dataset, the predictive performance of the single models applying at least one plasma concentration varied substantially (imprecision: 28-62%, inaccuracy: -16 to 25%), whereas the MSA or MAA utilizing these models simultaneously resulted in unbiased and precise predictions (imprecision: 29% and 30%, inaccuracy: -5% and 0%, respectively). MSA and MAA approaches implemented in TDMx might thereby lower the burden of fit-for-purpose validation of individual models and streamline MIPD.
Collapse
Affiliation(s)
- David W Uster
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Sophie L Stocker
- Department of Clinical Pharmacology and Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Jane E Carland
- Department of Clinical Pharmacology and Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Jonathan Brett
- Department of Clinical Pharmacology and Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Deborah J E Marriott
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia.,Department of Clinical Microbiology and Infectious Diseases, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Richard O Day
- Department of Clinical Pharmacology and Toxicology, St. Vincent's Hospital, Sydney, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| |
Collapse
|
25
|
Fused Deposition Modeling (FDM), the new asset for the production of tailored medicines. J Control Release 2020; 330:821-841. [PMID: 33130069 DOI: 10.1016/j.jconrel.2020.10.056] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/22/2020] [Accepted: 10/25/2020] [Indexed: 10/23/2022]
Abstract
Over the last few years, conventional medicine has been increasingly moving towards precision medicine. Today, the production of oral pharmaceutical forms tailored to patients is not achievable by traditional industrial means. A promising solution to customize oral drug delivery has been found in the utilization of 3D Printing and in particular Fused Deposition Modeling (FDM). Thus, the aim of this systematic literature review is to provide a synthesis on the production of pharmaceutical solid oral forms using FDM technology. In total, 72 relevant articles have been identified via two well-known scientific databases (PubMed and ScienceDirect). Overall, three different FDM methods have been reported: "Impregnation-FDM", "Hot Melt Extrusion coupled with FDM" and "Print-fill", which yielded to the formulation of thermoplastic polymers used as main component, five families of other excipients playing different functional roles and 47 active ingredients. Solutions are underway to overcome the high printing temperatures, which was the initial brake on to use thermosensitive ingredients with this technology. Also, the moisture sensitivity shown by a large number of prints in preliminary storage studies is highlighted. FDM seems to be especially fitted for the treatment of rare diseases, and particular populations requiring tailored doses or release kinetics. For future use of FDM in clinical trials, an implication of health regulatory agencies would be necessary. Hence, further efforts would likely be oriented to the use of a quality approach such as "Quality by Design" which could facilitate its approval by the authorities, and also be an aid to the development of this technology for manufacturers.
Collapse
|
26
|
Kluwe F, Michelet R, Mueller‐Schoell A, Maier C, Klopp‐Schulze L, Dyk M, Mikus G, Huisinga W, Kloft C. Perspectives on Model‐Informed Precision Dosing in the Digital Health Era: Challenges, Opportunities, and Recommendations. Clin Pharmacol Ther 2020; 109:29-36. [DOI: 10.1002/cpt.2049] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Franziska Kluwe
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy Freie Universität Berlin Berlin Germany
- Graduate Research Training Program PharMetrX Freie Universität Berlin and Universitaet Potsdam Berlin Germany
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy Freie Universität Berlin Berlin Germany
| | - Anna Mueller‐Schoell
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy Freie Universität Berlin Berlin Germany
- Graduate Research Training Program PharMetrX Freie Universität Berlin and Universitaet Potsdam Berlin Germany
| | - Corinna Maier
- Graduate Research Training Program PharMetrX Freie Universität Berlin and Universitaet Potsdam Berlin Germany
- Institute of Mathematics Universität Potsdam Potsdam Germany
| | - Lena Klopp‐Schulze
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy Freie Universität Berlin Berlin Germany
- Graduate Research Training Program PharMetrX Freie Universität Berlin and Universitaet Potsdam Berlin Germany
| | - Madelé Dyk
- Flinders Centre for Innovation in Cancer, College of Medicine & Public Health Flinders University Adelaide Australia
| | - Gerd Mikus
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy Freie Universität Berlin Berlin Germany
- Department of Clinical Pharmacology and Pharmacoepidemiology University Hospital Heidelberg Heidelberg Germany
| | | | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy Freie Universität Berlin Berlin Germany
| |
Collapse
|
27
|
Cheng Y, Wang CY, Li ZR, Pan Y, Liu MB, Jiao Z. Can Population Pharmacokinetics of Antibiotics be Extrapolated? Implications of External Evaluations. Clin Pharmacokinet 2020; 60:53-68. [PMID: 32960439 DOI: 10.1007/s40262-020-00937-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVE External evaluation is an important issue in the population pharmacokinetic analysis of antibiotics. The purpose of this review was to summarize the current approaches and status of external evaluations and discuss the implications of external evaluation results for the future individualization of dosing regimens. METHODS We systematically searched the PubMed and EMBASE databases for external evaluation studies of population analysis and extracted the relevant information from these articles. A total of 32 studies were included in this review. RESULTS Vancomycin was investigated in 17 (53.1%) articles and was the most studied drug. Other studied drugs included gentamicin, tobramycin, amikacin, amoxicillin, ceftaroline, meropenem, fluconazole, voriconazole, and rifampicin. Nine (28.1%) studies were prospective, and the sample size varied widely between studies. Thirteen (40.6%) studies evaluated the population pharmacokinetic models by systematically searching for previous studies. Seven (21.9%) studies were multicenter studies, and 27 (84.4%) adopted the sparse sampling strategy. Almost all external evaluation studies of antibiotics (93.8%) used metrics for prediction-based diagnostics, while relatively fewer studies were based on simulations (46.9%) and Bayesian forecasting (25.0%). CONCLUSION The results of external evaluations in previous studies revealed the poor extrapolation performance of existing models of prediction- and simulation-based diagnostics, whereas the posterior Bayesian method could improve predictive performance. There is an urgent need for the development of standards and guidelines for external evaluation studies.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200040, China.,Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Road, Gulou, Fuzhou, 350001, China
| | - Chen-Yu Wang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200040, China
| | - Zi-Ran Li
- College of Pharmacy, Fudan University, Shanghai, China
| | - Yan Pan
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200040, China
| | - Mao-Bai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, 29 Xin Quan Road, Gulou, Fuzhou, 350001, China.
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai, 200040, China.
| |
Collapse
|
28
|
Dave MB, Dherai AJ, Desai DC, Mould DR, Ashavaid TF. Optimization of infliximab therapy in inflammatory bowel disease using a dashboard approach-an Indian experience. Eur J Clin Pharmacol 2020; 77:55-62. [PMID: 32803288 DOI: 10.1007/s00228-020-02975-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/01/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Infliximab (IFX) therapy in inflammatory bowel disease (IBD) is associated with loss of response in half the patients, due to complex pharmacokinetic and immunological factors. Dashboard's Bayesian algorithms use information from model and individual multivariate determinants of IFX concentration and can predict dose and dosing interval. AIM To compare measured IFX concentrations in our laboratory with values predicted by iDose dashboard system and report its efficacy in managing patients not responding to conventional dosing schedule. METHOD Clinical history, demographic details, and laboratory findings such as albumin and C-reactive protein (CRP) data of IBD patients (n = 30; median age 23 years (IQR: 14.25 - 33.5)) referred for IFX drug monitoring in our laboratory from November 2017 to November 2019 were entered in iDose software. The IFX concentration predicted by iDose based on this information was compared with that measured in our laboratory. In addition, a prospective dashboard-guided dosing was prescribed in 11 of these 30 patients not responding to conventional dosing and was followed to assess their clinical outcome. RESULT IFX monitoring in our 30 patients had shown therapeutic concentration in 12, supratherapeutic in 2 and subtherapeutic concentration in 16 patients. The iDose predicted concentration showed concordance in 21 of these 30 patients. Of 11 patients managed with iDose-assisted prospective dosing, 8 achieved clinical remission, 2 showed partial response, and one developed antibodies. CONCLUSION Retrospective data analysis showed concordance between laboratory measured and iDose-predicted IFX level in 70% of patients. iDose-assisted management achieved clinical remission and cost reduction.
Collapse
Affiliation(s)
- Mihika B Dave
- Department of Biochemistry, P. D. Hinduja Hospital & MRC, Veer Savarkar Marg, Mahim, Mumbai, 400016, India
| | - Alpa J Dherai
- Department of Biochemistry, P. D. Hinduja Hospital & MRC, Veer Savarkar Marg, Mahim, Mumbai, 400016, India.
| | - Devendra C Desai
- Department of Gastroenterology, P. D. Hinduja Hospital & MRC, Veer Savarkar Marg, Mahim, Mumbai, 400016, India
| | | | - Tester F Ashavaid
- Department of Biochemistry, P. D. Hinduja Hospital & MRC, Veer Savarkar Marg, Mahim, Mumbai, 400016, India
| |
Collapse
|
29
|
Taylor ZL, Mizuno T, Punt NC, Baskaran B, Navarro Sainz A, Shuman W, Felicelli N, Vinks AA, Heldrup J, Ramsey LB. MTXPK.org: A Clinical Decision Support Tool Evaluating High-Dose Methotrexate Pharmacokinetics to Inform Post-Infusion Care and Use of Glucarpidase. Clin Pharmacol Ther 2020; 108:635-643. [PMID: 32558929 PMCID: PMC7484917 DOI: 10.1002/cpt.1957] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/03/2020] [Indexed: 12/30/2022]
Abstract
Methotrexate (MTX), an antifolate, is administered at high doses to treat malignancies in children and adults. However, there is considerable interpatient variability in clearance of high‐dose (HD) MTX. Patients with delayed clearance are at an increased risk for severe nephrotoxicity and life‐threatening systemic MTX exposure. Glucarpidase is a rescue agent for severe MTX toxicity that reduces plasma MTX levels via hydrolysis of MTX into inactive metabolites, but is only indicated when MTX concentrations are > 2 SDs above the mean excretion curve specific for the given dose together with a significant creatinine increase (> 50%). Appropriate administration of glucarpidase is challenging due to the ambiguity in the labeled indication. A recent consensus guideline was published with an algorithm to provide clarity in when to administer glucarpidase, yet clinical interpretation of laboratory results that do not directly correspond to the algorithm prove to be a limitation of its use. The goal of our study was to develop a clinical decision support tool to optimize the administration of glucarpidase for patients receiving HD MTX. Here, we describe the development of a novel 3‐compartment MTX population pharmacokinetic (PK) model using 31,672 MTX plasma concentrations from 772 pediatric patients receiving HD MTX for the treatment of acute lymphoblastic leukemia and its integration into the online clinical decision support tool, MTXPK.org. This web‐based tool has the functionality to utilize individualized demographics, serum creatinine, and real‐time drug concentrations to predict the elimination profile and facilitate model‐informed administration of glucarpidase.
Collapse
Affiliation(s)
- Zachary L Taylor
- Department of Molecular, Cellular, and Biochemical Pharmacology, University of Cincinnati, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Balaji Baskaran
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Adriana Navarro Sainz
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - William Shuman
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nicholas Felicelli
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alexander A Vinks
- Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jesper Heldrup
- Childhood Cancer and Research Unit, University Children's Hospital, Lund, Sweden
| | - Laura B Ramsey
- Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
30
|
Mizuno T, Dong M, Taylor ZL, Ramsey LB, Vinks AA. Clinical implementation of pharmacogenetics and model-informed precision dosing to improve patient care. Br J Clin Pharmacol 2020; 88:1418-1426. [PMID: 32529759 DOI: 10.1111/bcp.14426] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Providing maximal therapeutic efficacy without toxicity is a universal goal of rational drug therapy. However, substantial between-patient variability in drug response often impedes such successful treatments and brings the necessity of tailoring drug dose to individual needs for more precise therapy. In many cases plenty of patient characteristics, such as body size, genetic makeup and environmental factors, need to be taken into consideration to find the optimal dose in clinical practice. A pharmacokinetics and pharmacodynamics (PK/PD) model-informed approach offers integration of various patient information to provide an expectation of drug response and derive practical dose estimates to support clinicians' dosing decisions. Such an approach was pioneered in the late 1970s, but its broad clinical acceptance and implementation have been hampered by the lack of widespread computer technology, including user-friendly software tools. This has significantly changed in recent years. With the advent of electronic health records (EHRs) and the ubiquity of user-friendly software tools, we now experience a convergence of clinical information, pharmacogenetics, systems pharmacology and pharmacometrics, and technology. Advanced pharmacometrics research is now more appliable and implementable to improve health care. This article presents examples of successful development and implementation of pharmacogenetics-guided and PK/PD model-informed decision support to facilitate precision dosing, including the development of an EHR-embedded decision support tool. Through the integration of clinical decision support tools in EHRs, clinical pharmacometrics support can be brought directly to the clinical team and the bedside.
Collapse
Affiliation(s)
- Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Min Dong
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Zachary L Taylor
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Molecular, Cellular, and Biochemical Pharmacology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Laura B Ramsey
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Research in Patient Services, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
31
|
Frymoyer A, Schwenk HT, Zorn Y, Bio L, Moss JD, Chasmawala B, Faulkenberry J, Goswami S, Keizer RJ, Ghaskari S. Model-Informed Precision Dosing of Vancomycin in Hospitalized Children: Implementation and Adoption at an Academic Children's Hospital. Front Pharmacol 2020; 11:551. [PMID: 32411000 PMCID: PMC7201037 DOI: 10.3389/fphar.2020.00551] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/09/2020] [Indexed: 02/03/2023] Open
Abstract
Background Model-informed precision dosing (MIPD) can serve as a powerful tool during therapeutic drug monitoring (TDM) to help individualize dosing in populations with large pharmacokinetic variation. Yet, adoption of MIPD in the clinical setting has been limited. Overcoming technologic hurdles that allow access to MIPD at the point-of-care and placing it in the hands of clinical specialists focused on medication dosing may encourage adoption. Objective To describe the hospital implementation and usage of a MIPD clinical decision support (CDS) tool for vancomycin in a pediatric population. Methods Within an academic children’s hospital, MIPD for vancomycin was implemented via a commercial cloud-based CDS tool that utilized Bayesian forecasting. Clinical pharmacists were recognized as local champions to facilitate adoption of the tool and operated as end-users. Integration within the electronic health record (EHR) and automatic transmission of patient data to the tool were identified as important requirements. A web-link icon was developed within the EHR which when clicked sends users and needed patient-level clinical data to the CDS platform. Individualized pharmacokinetic predictions and exposure metrics for vancomycin are then presented in the form of a web-based dashboard. Use of the CDS tool as part of TDM was tracked and users were surveyed on their experience. Results After a successful pilot phase in the neonatal intensive care unit, implementation of MIPD was expanded to the pediatric intensive care unit, followed by availability to the entire hospital. During the first 2+ years since implementation, a total of 853 patient-courses (n = 96 neonates, n = 757 children) and 2,148 TDM levels were evaluated using the CDS tool. For the most recent 6 months, the CDS tool was utilized to support 79% (181/230) of patient-courses in which TDM was performed. Of 26 users surveyed, > 96% agreed or strongly agreed that automatic transmission of patient data to the tool was a feature that helped them complete tasks more efficiently; 81% agreed or strongly agreed that they were satisfied with the CDS tool. Conclusions Integration of a vancomycin CDS tool within the EHR, along with leveraging the expertise of clinical pharmacists, allowed for successful adoption of MIPD in clinical care.
Collapse
Affiliation(s)
- Adam Frymoyer
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Hayden T Schwenk
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Yvonne Zorn
- Department of Clinical Pharmacy, Lucile Packard Children's Hospital Stanford, Palo Alto, CA, United States
| | - Laura Bio
- Department of Clinical Pharmacy, Lucile Packard Children's Hospital Stanford, Palo Alto, CA, United States
| | - Jeffrey D Moss
- Department of Clinical Pharmacy, Lucile Packard Children's Hospital Stanford, Palo Alto, CA, United States
| | - Bhavin Chasmawala
- Information Services, Lucile Packard Children's Hospital Stanford, Palo Alto, CA, United States
| | - Joshua Faulkenberry
- Information Services, Lucile Packard Children's Hospital Stanford, Palo Alto, CA, United States
| | | | | | - Shabnam Ghaskari
- Information Services, Lucile Packard Children's Hospital Stanford, Palo Alto, CA, United States
| |
Collapse
|
32
|
Buclin T, Thoma Y, Widmer N, André P, Guidi M, Csajka C, Decosterd LA. The Steps to Therapeutic Drug Monitoring: A Structured Approach Illustrated With Imatinib. Front Pharmacol 2020; 11:177. [PMID: 32194413 PMCID: PMC7062864 DOI: 10.3389/fphar.2020.00177] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/07/2020] [Indexed: 01/07/2023] Open
Abstract
Pharmacometric methods have hugely benefited from progress in analytical and computer sciences during the past decades, and play nowadays a central role in the clinical development of new medicinal drugs. It is time that these methods translate into patient care through therapeutic drug monitoring (TDM), due to become a mainstay of precision medicine no less than genomic approaches to control variability in drug response and improve the efficacy and safety of treatments. In this review, we make the case for structuring TDM development along five generic questions: 1) Is the concerned drug a candidate to TDM? 2) What is the normal range for the drug's concentration? 3) What is the therapeutic target for the drug's concentration? 4) How to adjust the dosage of the drug to drive concentrations close to target? 5) Does evidence support the usefulness of TDM for this drug? We exemplify this approach through an overview of our development of the TDM of imatinib, the very first targeted anticancer agent. We express our position that a similar story shall apply to other drugs in this class, as well as to a wide range of treatments critical for the control of various life-threatening conditions. Despite hurdles that still jeopardize progress in TDM, there is no doubt that upcoming technological advances will shape and foster many innovative therapeutic monitoring methods.
Collapse
Affiliation(s)
- Thierry Buclin
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Yann Thoma
- School of Management and Engineering Vaud (HEIG-VD), University of Applied Science Western Switzerland (HES-SO), Yverdon-les-Bains, Switzerland
| | - Nicolas Widmer
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Pharmacy of Eastern Vaud Hospitals, Rennaz, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Pascal André
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Monia Guidi
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Chantal Csajka
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,Center for Research and Innovation in Clinical Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Laurent A Decosterd
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
33
|
Berends SE, Strik AS, Löwenberg M, D'Haens GR, Mathôt RAA. Clinical Pharmacokinetic and Pharmacodynamic Considerations in the Treatment of Ulcerative Colitis. Clin Pharmacokinet 2020; 58:15-37. [PMID: 29752633 PMCID: PMC6326086 DOI: 10.1007/s40262-018-0676-z] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) of unknown etiology, probably caused by a combination of genetic and environmental factors. The treatment of patients with active UC depends on the severity, localization and history of IBD medication. According to the classic step-up approach, treatment with 5-aminosalicylic acid compounds is the first step in the treatment of mild to moderately active UC. Corticosteroids, such as prednisolone are used in UC patients with moderate to severe disease activity, but only for remission induction therapy because of side effects associated with long-term use. Thiopurines are the next step in the treatment of active UC but monotherapy during induction therapy in UC patients is not preferred because of their slow onset. Therapeutic drug monitoring (TDM) of the pharmacologically active metabolites of thiopurines, 6-thioguanine nucleotide (6-TGN), has proven to be beneficial. Thiopurine S-methyltransferase (TMPT) plays a role in the metabolic conversion pathway of thiopurines and exhibits genetic polymorphism; however, the clinical benefit and relevance of TPMT genotyping is not well established. In patients with severely active UC refractory to corticosteroids, calcineurin inhibitors such as ciclosporin A (CsA) and tacrolimus are potential therapeutic options. These agents usually have a rather rapid onset of action. Monoclonal antibodies (anti-tumor necrosis factor [TNF] agents, vedolizumab) are the last pharmacotherapeutic option for UC patients before surgery becomes inevitable. Body weight, albumin status and antidrug antibodies contribute to the variability in the pharmacokinetics of anti-TNF agents. Additionally, the use of concomitant immunomodulators (thiopurines/methotrexate) lowers the rate of immunogenicity, and therefore the concomitant use of anti-TNF therapy with an immunomodulator may confer some advantage compared with monotherapy in certain patients. TDM of anti-TNF agents could be beneficial in patients with primary nonresponse and secondary loss of response. The potential benefit of applying TDM during vedolizumab treatment has yet to be determined.
Collapse
Affiliation(s)
- Sophie E Berends
- Department Hospital Pharmacy, Academic Medical Center, Amsterdam, The Netherlands.
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| | - Anne S Strik
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Geert R D'Haens
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Ron A A Mathôt
- Department Hospital Pharmacy, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Huang L, Liu Y, Jiao Z, Wang J, Fang L, Mao J. Population pharmacokinetic study of tacrolimus in pediatric patients with primary nephrotic syndrome: A comparison of linear and nonlinear Michaelis–Menten pharmacokinetic model. Eur J Pharm Sci 2020; 143:105199. [DOI: 10.1016/j.ejps.2019.105199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/25/2022]
|
35
|
Admiraal R, Jol-van der Zijde CM, Furtado Silva JM, Knibbe CAJ, Lankester AC, Boelens JJ, Hale G, Etuk A, Wilson M, Adams S, Veys P, van Kesteren C, Bredius RGM. Population Pharmacokinetics of Alemtuzumab (Campath) in Pediatric Hematopoietic Cell Transplantation: Towards Individualized Dosing to Improve Outcome. Clin Pharmacokinet 2019; 58:1609-1620. [PMID: 31131436 PMCID: PMC6885503 DOI: 10.1007/s40262-019-00782-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Alemtuzumab (Campath®) is used to prevent graft-versus-host disease and graft failure following pediatric allogeneic hematopoietic cell transplantation. The main toxicity includes delayed immune reconstitution, subsequent viral reactivations, and leukemia relapse. Exposure to alemtuzumab is highly variable upon empirical milligram/kilogram dosing. METHODS A population pharmacokinetic (PK) model for alemtuzumab was developed based on a total of 1146 concentration samples from 206 patients, aged 0.2-19 years, receiving a cumulative intravenous dose of 0.2-1.5 mg/kg, and treated between 2003 and 2015 in two centers. RESULTS Alemtuzumab PK were best described using a two-compartment model with a parallel saturable and linear elimination pathway. The linear clearance pathway, central volume of distribution, and intercompartmental distribution increased with body weight. Blood lymphocyte counts, a potential substrate for alemtuzumab, did not impact clearance. CONCLUSION The current practice with uniform milligram/kilogram doses leads to highly variable exposures in children due to the non-linear relationship between body weight and alemtuzumab PK. This model may be used for individualized dosing of alemtuzumab.
Collapse
Affiliation(s)
- Rick Admiraal
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
| | - Cornelia M Jol-van der Zijde
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Catherijne A J Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Arjan C Lankester
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap Jan Boelens
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
- Stem Cell Transplant and Cellular Therapies, Memorial Sloane Kettering Cancer Center, New York, NY, USA
| | | | - Aniekan Etuk
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Melanie Wilson
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Stuart Adams
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Paul Veys
- Bone Marrow Transplantation Department, Great Ormond Street Hospital, London, UK
| | - Charlotte van Kesteren
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
| | - Robbert G M Bredius
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
36
|
Vinks AA, Peck RW, Neely M, Mould DR. Development and Implementation of Electronic Health Record–Integrated Model‐Informed Clinical Decision Support Tools for the Precision Dosing of Drugs. Clin Pharmacol Ther 2019; 107:129-135. [DOI: 10.1002/cpt.1679] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Alexander A. Vinks
- Division of Clinical Pharmacology Cincinnati Children's Hospital Medical Center Cincinnati Ohio USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati Ohio USA
| | - Richard W. Peck
- Pharma Research and Exploratory Development Roche Innovation Center Basel Basel Switzerland
| | - Michael Neely
- Children's Hospital Los Angeles University of Southern California Los Angeles California USA
| | | |
Collapse
|
37
|
Fidler M, Wilkins JJ, Hooijmaijers R, Post TM, Schoemaker R, Trame MN, Xiong Y, Wang W. Nonlinear Mixed-Effects Model Development and Simulation Using nlmixr and Related R Open-Source Packages. CPT Pharmacometrics Syst Pharmacol 2019; 8:621-633. [PMID: 31207186 PMCID: PMC6765694 DOI: 10.1002/psp4.12445] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/29/2019] [Indexed: 12/15/2022] Open
Abstract
nlmixr is a free and open-source R package for fitting nonlinear pharmacokinetic (PK), pharmacodynamic (PD), joint PK-PD, and quantitative systems pharmacology mixed-effects models. Currently, nlmixr is capable of fitting both traditional compartmental PK models as well as more complex models implemented using ordinary differential equations. We believe that, over time, it will become a capable, credible alternative to commercial software tools, such as NONMEM, Monolix, and Phoenix NLME.
Collapse
Affiliation(s)
| | | | | | | | | | - Mirjam N. Trame
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | | | - Wenping Wang
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| |
Collapse
|
38
|
Berends SE, D'Haens GRAM, Schaap T, de Vries A, Rispens T, Bloem K, Mathôt RAA. Dried blood samples can support monitoring of infliximab concentrations in patients with inflammatory bowel disease: A clinical validation. Br J Clin Pharmacol 2019; 85:1544-1551. [PMID: 30927375 PMCID: PMC6595298 DOI: 10.1111/bcp.13939] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/12/2019] [Accepted: 03/21/2019] [Indexed: 01/11/2023] Open
Abstract
Aims Therapeutic drug monitoring (TDM) can optimize the efficacy of infliximab (IFX) in patients with inflammatory bowel disease (IBD). Because of the delay between blood samples taken at trough and availability of results, dose adjustments can only be carried out at the next infusion, typically 8 weeks later. Dried blood samples (DBS) performed at home to measure IFX concentrations can reduce the time to adapt dose/dosing interval. Here, we aimed to validate the clinical application of DBS for IFX in IBD patients and to evaluate the feasibility of home sampling. Methods DBS results from 40 IBD patients on IFX treatment were compared to serum sample results at trough, peak, and 3–5 weeks after IFX infusion. Subsequently, patients performed DBS home sampling one week before the next IFX infusion. These were compared to serum concentrations as predicted by Bayesian analysis. Results IFX concentrations from finger prick and venous puncture correlate well. DBS IFX concentrations showed high correlation with serum IFX concentrations (Spearman correlation: ≥0.965), without bias. Passing‐Bablok regression for IFX concentrations in DBS from home sampling also showed no bias (intercept: 1.02 mg L−1 (95% CI −1.77–2.04 mg L−1), slope: 0.82 (95% CI 0.63–1.40)), with reasonable correlation (Spearman correlation: 0.671). Conclusions Timely adjustment of IFX dose/dosing interval can be facilitated by IFX concentration measurement in home‐sampled DBS. DBS is a reliable method to measure IFX and can be used to predict IFX trough concentrations.
Collapse
Affiliation(s)
- Sophie E. Berends
- Department Hospital PharmacyAmsterdam University Medical CentresAmsterdamThe Netherlands
- Department of Gastroenterology and HepatologyAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Geert R. A. M. D'Haens
- Department of Gastroenterology and HepatologyAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Tiny Schaap
- Biologics Lab, BioanalysisSanquin Diagnostic ServicesAmsterdamThe Netherlands
| | - Annick de Vries
- Biologics Lab, BioanalysisSanquin Diagnostic ServicesAmsterdamThe Netherlands
| | - Theo Rispens
- Department of ImmunopathologySanquin Research and Landsteiner LaboratoryAmsterdamThe Netherlands
| | - Karien Bloem
- Biologics Lab, BioanalysisSanquin Diagnostic ServicesAmsterdamThe Netherlands
| | - Ron A. A. Mathôt
- Department Hospital PharmacyAmsterdam University Medical CentresAmsterdamThe Netherlands
| |
Collapse
|
39
|
Dreesen E, Faelens R, Van Assche G, Ferrante M, Vermeire S, Gils A, Bouillon T. Optimising infliximab induction dosing for patients with ulcerative colitis. Br J Clin Pharmacol 2019; 85:782-795. [PMID: 30634202 DOI: 10.1111/bcp.13859] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/11/2018] [Accepted: 12/21/2018] [Indexed: 12/24/2022] Open
Abstract
AIMS The therapeutic failure of infliximab therapy in patients with ulcerative colitis remains a challenge even 2 decades after its approval. Therapeutic drug monitoring (TDM) has shown value during maintenance therapy, but induction therapy has still not been explored. Patients may be primary nonresponders or underexposed with the standard dosing regimen. We aimed to: (i) develop a population pharmacokinetic-pharmacodynamic model; (ii) identify the best exposure metric that predicts mucosal healing; and (iii) build an exposure-response (ER) model to demonstrate model-based dose finding during induction therapy with infliximab. METHODS Data were retrospectively collected from a clinical database. A total of 583 samples, from 204 patients, was used to develop a population pharmacokinetic model to generate exposure metrics for subsequent ER modelling. A subset of 159 patients was used to develop a logistic regression ER model, describing the relationship between infliximab exposure and ordered transitions between Mayo endoscopic subscore (MES) 3, 2 and ≤1 (baseline to post-induction). RESULTS A 1-compartment population pharmacokinetic model with interindividual and interoccasion variability was found to fit the data best. Covariates influencing exposure were C-reactive protein, albumin, baseline MES, fat-free mass, concomitant corticosteroid use and pancolitis. The cumulative area under the infliximab concentration-time curve until endoscopy (CAUCendoscopy ) was found to be the best exposure metric for predicting mucosal healing (baseline MES >1 and post-induction MES ≤1). The model predicted that 70% of patients will attain mucosal healing with infliximab administered at days 0, 14 and 42 and a target CAUCendoscopy of 3752 mg/L*day at day 84. CONCLUSIONS TDM-based dose individualisation targeting CAUCendoscopy has the potential to improve the effectiveness of infliximab during induction therapy.
Collapse
Affiliation(s)
- Erwin Dreesen
- Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Ruben Faelens
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Gert Van Assche
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Ann Gils
- Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Thomas Bouillon
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
40
|
Lau MK, Bounthavong M, Kay CL, Harvey MA, Christopher MLD. Clinical dashboard development and use for academic detailing in the U.S. Department of Veterans Affairs. J Am Pharm Assoc (2003) 2019; 59:S96-S103.e3. [PMID: 30713078 DOI: 10.1016/j.japh.2018.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/19/2018] [Accepted: 12/04/2018] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To describe the U.S. Department of Veterans Affairs (VA) Academic Detailing Service's (ADS) experience with the development and use of clinical dashboards across the VA's national clinical campaigns. We focused only on dashboards developed by the VA ADS national clinical program managers. SETTING U.S. Department of Veterans Affairs Pharmacy Benefits Management National Academic Detailing Service. PRACTICE DESCRIPTION Academic detailing is a multifaceted, educational outreach intervention that services providers through interactions with academic detailers (at the VA, these are specially trained clinical pharmacy specialists) using evidence-based research, educational brochures, and clinical dashboards to align prescribing behavior with best practices. The VA ADS developed clinical dashboards to benchmark and monitor academic detailing activities and performance and to identify opportunities for redistributing resources. We used the opioid crisis as an example to highlight key steps in the development of a clinical dashboard. EVALUATION Testing is an important part of clinical dashboard development. Evaluations of the users perceptions contributed to lessons learned. RESULTS Data validation, missing data, data availability, standardization, user engagement, and technical limitations were among several challenges the VA ADS encountered during dashboard development. Stakeholder engagement, communication, and flexibility with development time allowed us to develop efficient dashboards. CONCLUSIONS Health care data and health analytics have transformed the type of clinical care that can be practiced by creating the ability to implement system-wide processes for both population management and quality improvement processes. End users of these VA ADS clinical dashboards can generate priority panel reports and data visualization of key performance indicators to identify areas for improvement or action.
Collapse
|
41
|
Polasek TM, Rostami-Hodjegan A, Yim DS, Jamei M, Lee H, Kimko H, Kim JK, Nguyen PTT, Darwich AS, Shin JG. What Does it Take to Make Model-Informed Precision Dosing Common Practice? Report from the 1st Asian Symposium on Precision Dosing. AAPS JOURNAL 2019; 21:17. [PMID: 30627939 DOI: 10.1208/s12248-018-0286-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022]
Abstract
Model-informed precision dosing (MIPD) is modeling and simulation in healthcare to predict the drug dose for a given patient based on their individual characteristics that is most likely to improve efficacy and/or lower toxicity in comparison to traditional dosing. This paper describes the background and status of MIPD and the activities at the 1st Asian Symposium of Precision Dosing. The theme of the meeting was the question, "What does it take to make MIPD common practice?" Formal presentations highlighted the distinction between genetic and non-genetic sources of variability in drug exposure and response, the use of modeling and simulation as decision support tools, and the facilitators to MIPD implementation. A panel discussion addressed the types of models used for MIPD, how the pharmaceutical industry views MIPD, ways to upscale MIPD beyond academic hospital centers, and the essential role of healthcare professional education as a way to progress. The meeting concluded with an ongoing commitment to use MIPD to improve patient care.
Collapse
Affiliation(s)
- Thomas M Polasek
- Certara, 100 Overlook Center, Suite 101, Princeton, New Jersey, 08540, USA. .,Centre for Medicines Use and Safety, Monash University, Melbourne, Australia.
| | - Amin Rostami-Hodjegan
- Certara, 100 Overlook Center, Suite 101, Princeton, New Jersey, 08540, USA.,Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Dong-Seok Yim
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Masoud Jamei
- Certara, 100 Overlook Center, Suite 101, Princeton, New Jersey, 08540, USA
| | - Howard Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Holly Kimko
- Janssen Research and Development, Lower Gwynedd Township, Pennsylvania, USA
| | - Jae Kyoung Kim
- Korea Advanced Institute of Advanced Technology, Daedoek Innopolis, Daejeon, South Korea
| | - Phuong Thi Thu Nguyen
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea.,Faculty of Pharmacy, Haiphong University of Medicine and Pharmacy, Haiphong, Vietnam
| | - Adam S Darwich
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Jae-Gook Shin
- Department of Pharmacology and Clinical Pharmacology, Pharmacogenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
42
|
Pearce NF, Giblin EM, Buckthal C, Ferrari A, Powell JR, Cao Y, Patterson JH. Precision drug dosing: A major opportunity for patients and pharmacists. JOURNAL OF THE AMERICAN COLLEGE OF CLINICAL PHARMACY 2018. [DOI: 10.1002/jac5.1017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Natalie F. Pearce
- Division of Pharmacotherapy and Experimental Therapeutics UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina
| | - Erika M. Giblin
- Division of Pharmacotherapy and Experimental Therapeutics UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina
| | - Catherine Buckthal
- Division of Pharmacotherapy and Experimental Therapeutics UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina
| | - Alana Ferrari
- Division of Pharmacotherapy and Experimental Therapeutics UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina
| | - J. Robert Powell
- Division of Pharmacotherapy and Experimental Therapeutics UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina
| | - J. Herbert Patterson
- Division of Pharmacotherapy and Experimental Therapeutics UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina
| |
Collapse
|
43
|
Ramsey LB, Mizuno T, Vinks AA, Margolis PA. Learning Health Systems as Facilitators of Precision Medicine. Clin Pharmacol Ther 2018; 101:359-367. [PMID: 27984650 DOI: 10.1002/cpt.594] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 12/24/2022]
Affiliation(s)
- L B Ramsey
- Division of Research in Patient Services, Pharmacy Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - T Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - A A Vinks
- Division of Research in Patient Services, Pharmacy Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - P A Margolis
- James M. Anderson Center for Health Systems Excellence, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
44
|
Euteneuer JC, Kamatkar S, Fukuda T, Vinks AA, Akinbi HT. Suggestions for Model-Informed Precision Dosing to Optimize Neonatal Drug Therapy. J Clin Pharmacol 2018; 59:168-176. [PMID: 30204236 DOI: 10.1002/jcph.1315] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/17/2018] [Indexed: 12/19/2022]
Abstract
Evidence for dosing, efficacy, and safety of most medications used to treat neonates is sparse. Thus, dosing is usually derived by extrapolation from adult and pediatric pharmacologic data with scaling by body weight or body surface area. This may lead to drug dosing that is unsafe or ineffective. However, new strategies are being developed and studied to dose medications in critically ill neonates. Mass spectroscopy technology capable of quickly analyzing drug levels is readily available. Software that integrates population pharmacokinetics and pharmacodynamics with data from sparse samples from neonates allows for timely adjustments of dosing to achieve the desired effect while minimizing adverse outcomes. Some genetic polymorphisms that affect drug response in neonates have also been reported. This review highlights aspects of drug response and how it is impacted by prematurity, assesses pharmacogenomic studies in neonates, and offers suggestions for innovative pharmacokinetic/pharmacodynamic model-based approaches that combine population- or physiology-based pharmacology data, Bayesian analysis, and electronic decision support tools for precision dosing in neonates while illustrating examples where this approach can be used to optimize medical therapy in neonates. Barriers to implementing precision dosing in neonates and how to overcome them are also discussed.
Collapse
Affiliation(s)
- Joshua C Euteneuer
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA.,Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Suyog Kamatkar
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tsuyoshi Fukuda
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Henry T Akinbi
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
45
|
Strik AS, Wang YMC, Ruff LE, Yashar W, Messmer BT, Mould DR. Individualized Dosing of Therapeutic Monoclonal Antibodies-a Changing Treatment Paradigm? AAPS JOURNAL 2018; 20:99. [PMID: 30187153 PMCID: PMC8364290 DOI: 10.1208/s12248-018-0257-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
The introduction of monoclonal antibodies (mAbs) to the treatment of inflammatory bowel disease (IBD) was an important medical milestone. MAbs have been demonstrated as safe and efficacious treatments of IBD. However, a large percentage of patients either fail to respond initially or lose response to therapy after a period of treatment. Although there are factors associated with poor treatment outcomes in IBD, one cause for treatment failure may be low mAb exposure. Consequently, gastroenterologists have begun using therapeutic drug monitoring (TDM) to guide dose adjustment. However, while beneficial, TDM does not provide sufficient information to effectively adjust doses. The pharmacokinetics (PK) and pharmacodynamics (PD) of mAbs are complex, with numerous factors impacting on mAb PK and PD. The concept of dashboard-guided dosing based on Bayesian PK models allows physicians to combine TDM with factors influencing mAb PK to individualize therapy more effectively. One issue with TDM has been the slow turnaround of assay results, either necessitating an additional clinic visit for a sample or reacting to TDM results at a subsequent, rather than the current, dose. New point-of-care (POC) assays for mAbs are being developed that would potentially allow physicians to determine drug concentration quickly. However, work remains to understand how to determine what target exposure is needed for an individual patient, and whether the combination of POC assays and dashboards presents a safe approach with substantial outcome benefit over the current standard of care.
Collapse
Affiliation(s)
- Anne S Strik
- Academic Medical Center Division of Gastroenterology, Amsterdam, Netherlands
| | - Yow-Ming C Wang
- Therapeutic Biologics Program, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | | | - Diane R Mould
- Projections Research Inc., 535 Springview Lane, Phoenixville, Pennsylvania, 19460, USA.
| |
Collapse
|
46
|
Mulugeta LY, Yao L, Mould D, Jacobs B, Florian J, Smith B, Sinha V, Barrett JS. Leveraging Big Data in Pediatric Development Programs: Proceedings From the 2016 American College of Clinical Pharmacology Annual Meeting Symposium. Clin Pharmacol Ther 2018; 104:81-87. [PMID: 29319159 DOI: 10.1002/cpt.975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/21/2017] [Accepted: 12/01/2017] [Indexed: 12/26/2022]
Abstract
This article discusses the use of big data in pediatric drug development. The article covers key topics discussed at the ACCP annual meeting symposium in 2016 including the extent to which big data or real-world data can inform clinical trial design and substitute for efficacy and safety data typically obtained in clinical trials. The current states of use, opportunities, and challenges with the use of big data in future pediatric drug development are discussed.
Collapse
Affiliation(s)
| | - Lynne Yao
- U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Diane Mould
- Projections Research Inc, Phoenixville, Pennsylvania, USA
| | - Brian Jacobs
- Children's National Medical Center, Washington, DC; George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Jeffrey Florian
- U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Brian Smith
- Duke University Medical Center, Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Vikram Sinha
- Quantitative Pharmacology and Pharmacometrics, Merck and Co, North Wales, Pennsylvania, USA
| | | |
Collapse
|
47
|
Peck RW. Precision Medicine Is Not Just Genomics: The Right Dose for Every Patient. Annu Rev Pharmacol Toxicol 2018; 58:105-122. [DOI: 10.1146/annurev-pharmtox-010617-052446] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Richard W. Peck
- Pharma Research and Exploratory Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| |
Collapse
|
48
|
Application of Population Pharmacokinetic Modeling for Individualized Infliximab Dosing Strategies in Crohn Disease. J Pediatr Gastroenterol Nutr 2017; 65:639-645. [PMID: 28471911 PMCID: PMC5670026 DOI: 10.1097/mpg.0000000000001620] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES The pharmacokinetics of infliximab (IFX) is highly variable in children with Crohn disease (CD), and a one-size-fits-all approach to dosing is inadequate. Model-based drug dosing can help individualize dosing strategies. We evaluated the predictive performance and clinical utility of a published population pharmacokinetic model of IFX in children with CD. METHODS Within a cohort of 34 children with CD who had IFX trough concentrations measured, the pharmacokinetics of each patient was estimated in NONMEM using a published population pharmacokinetic model. Infliximab concentrations were then predicted based on each patient's dosing history and compared with actual measured concentrations (n = 59). In addition, doses 5 to 10 mg/kg and dosing intervals every 4 to 8 weeks were simulated in each patient to examine dose-trough relationships. RESULTS Predicted concentrations were within ±1.0 μg/mL of actual measured concentrations for 88% of measurements. The median prediction error (ie, measure of bias) was -0.15 μg/mL (95% confidence interval -0.37 to -0.05 μg/mL) and absolute prediction error (ie, measure of precision) was 0.26 μg/mL (95% confidence interval 0.15 to 0.40 μg/mL). At standard maintenance dosing of 5 mg/kg every 8 weeks, a trough >3 μg/mL was predicted to be achieved in 32% of patients. To achieve a trough >3 μg/mL, a dosing interval ≤every 6 weeks was predicted to be required in 29% of patients. CONCLUSIONS A published IFX population pharmacokinetic model demonstrated accurate predictive performance in a pediatric CD population. Individualized IFX dosing strategies in children with CD will be critical to consistently achieve trough concentrations associated with optimal outcomes.
Collapse
|
49
|
Mao JJ, Jiao Z, Yun HY, Zhao CY, Chen HC, Qiu XY, Zhong MK. External evaluation of population pharmacokinetic models for ciclosporin in adult renal transplant recipients. Br J Clin Pharmacol 2017; 84:153-171. [PMID: 28891596 DOI: 10.1111/bcp.13431] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/08/2017] [Accepted: 09/01/2017] [Indexed: 02/03/2023] Open
Abstract
AIMS Several population pharmacokinetic (popPK) models for ciclosporin (CsA) in adult renal transplant recipients have been constructed to optimize the therapeutic regimen of CsA. However, little is known about their predictabilities when extrapolated to different clinical centres. Therefore, this study aimed to externally evaluate the predictive ability of CsA popPK models and determine the potential influencing factors. METHODS A literature search was conducted and the predictive performance was determined for each selected model using an independent data set of 62 patients (471 predose and 500 2-h postdose concentrations) from our hospital. Prediction-based diagnostics and simulation-based normalized prediction distribution error were used to evaluate model predictability. The influence of prior information was assessed using Bayesian forecasting. Additionally, potential factors influencing model predictability were investigated. RESULTS Seventeen models extracted from 17 published popPK studies were assessed. Prediction-based diagnostics showed that ethnicity potentially influenced model transferability. Simulation-based normalized prediction distribution error analyses indicated misspecification in most of the models, especially regarding variance. Bayesian forecasting demonstrated that the predictive performance of the models substantially improved with 2-3 prior observations. The predictability of nonlinear Michaelis-Menten models was superior to that of linear compartmental models when evaluating the impact of structural models, indicating the underlying nonlinear kinetics of CsA. Structural model, ethnicity, covariates and prior observations potentially affected model predictability. CONCLUSIONS Structural model is the predominant factor influencing model predictability. Incorporation of nonlinear kinetics in CsA popPK modelling should be considered. Moreover, Bayesian forecasting substantially improved model predictability.
Collapse
Affiliation(s)
- Jun-Jun Mao
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Zheng Jiao
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | - Chen-Yan Zhao
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Han-Chao Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiao-Yan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming-Kang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Moyer AM, Caraballo PJ. The challenges of implementing pharmacogenomic testing in the clinic. Expert Rev Pharmacoecon Outcomes Res 2017; 17:567-577. [PMID: 28949250 DOI: 10.1080/14737167.2017.1385395] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Pharmacogenomic testing has the potential to greatly benefit patients by enabling personalization of medication management, ensuring better efficacy and decreasing the risk of side effects. However, to fully realize the potential of pharmacogenomic testing, there are several important issues that must be addressed. Areas covered: In this expert review we discuss current challenges impacting the implementation of pharmacogenomic testing in the clinical practice. We emphasize issues related to testing methods, reporting of the results, test selection, clinical interpretation of the results, cost-effectiveness, and the long-term use of pharmacogenomic results in clinical practice. We identify opportunities and future directions to facilitate clinical implementation. Expert commentary: Several key elements are necessary to optimally integrate pharmacogenomic testing into clinical practice. Collaborative efforts among laboratories are needed to improve standardization of testing and reporting of the results. Clinicians need educational opportunities to improve understanding of which test to order and how to interpret the results. The electronic health records and other clinical systems need to improve their storage of the pharmacogenomics test results and interoperability to facilitate the use of clinically actionable results to improve patient care.
Collapse
Affiliation(s)
- Ann M Moyer
- a Department of Laboratory Medicine and Pathology , Mayo Clinic , Rochester , MN , USA
| | - Pedro J Caraballo
- b Department of Medicine , Mayo Clinic , Rochester , MN , USA.,c Center for Translational Informatics and Knowledge Management, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|