1
|
Zaph S, Leiser RJ, Tao M, Kaddi C, Xu C. Application of Quantitative Systems Pharmacology Approaches to Support Pediatric Labeling in Rare Diseases. Handb Exp Pharmacol 2024. [PMID: 39673036 DOI: 10.1007/164_2024_734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024]
Abstract
Quantitative Systems Pharmacology (QSP) models offer a promising approach to extrapolate drug efficacy across different patient populations, particularly in rare diseases. Unlike conventional empirical models, QSP models can provide a mechanistic understanding of disease progression and therapeutic response by incorporating current disease knowledge into the descriptions of biomarkers and clinical endpoints. This allows for a holistic representation of the disease and drug response. The mechanistic nature of QSP models is well suited to pediatric extrapolation concepts, providing a quantitative method to assess disease and drug response similarity between adults and pediatric patients. The application of a QSP-based assessment of the disease and drug similarity in adult and pediatric patients in the clinical development program of olipudase alfa, a treatment for Acid Sphingomyelinase Deficiency (ASMD), illustrates the potential of this approach.
Collapse
Affiliation(s)
- Susana Zaph
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA.
| | - Randolph J Leiser
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| | - Mengdi Tao
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| | - Chanchala Kaddi
- Translational Disease Modeling - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| | - Christine Xu
- Pharmacokinetics, Dynamics, Metabolism - Translational Medicine, Research & Development, Sanofi-US, Bridgewater, NJ, USA
| |
Collapse
|
2
|
Singh FA, Afzal N, Smithline SJ, Thalhauser CJ. Assessing the performance of QSP models: biology as the driver for validation. J Pharmacokinet Pharmacodyn 2024; 51:533-542. [PMID: 37386340 DOI: 10.1007/s10928-023-09871-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/15/2023] [Indexed: 07/01/2023]
Abstract
Validation of a quantitative model is a critical step in establishing confidence in the model's suitability for whatever analysis it was designed. While processes for validation are well-established in the statistical sciences, the field of quantitative systems pharmacology (QSP) has taken a more piecemeal approach to defining and demonstrating validation. Although classical statistical methods can be used in a QSP context, proper validation of a mechanistic systems model requires a more nuanced approach to what precisely is being validated, and what role said validation plays in the larger context of the analysis. In this review, we summarize current thoughts of QSP validation in the scientific community, contrast the aims of statistical validation from several contexts (including inference, pharmacometrics analysis, and machine learning) with the challenges faced in QSP analysis, and use examples from published QSP models to define different stages or levels of validation, any of which may be sufficient depending on the context at hand.
Collapse
Affiliation(s)
- Fulya Akpinar Singh
- Genmab US, Inc., 777 Scudders Mill Rd Bldg 2 4th Floor, Plainsboro, NJ, 08536, USA
| | - Nasrin Afzal
- Genmab US, Inc., 777 Scudders Mill Rd Bldg 2 4th Floor, Plainsboro, NJ, 08536, USA
| | - Shepard J Smithline
- Genmab US, Inc., 777 Scudders Mill Rd Bldg 2 4th Floor, Plainsboro, NJ, 08536, USA
| | - Craig J Thalhauser
- Genmab US, Inc., 777 Scudders Mill Rd Bldg 2 4th Floor, Plainsboro, NJ, 08536, USA.
| |
Collapse
|
3
|
Decrane R, Stoker T, Murr A, Ford J, El-Masri H. Cross species extrapolation of the disruption of thyroid hormone synthesis by oxyfluorfen using in vitro data, physiologically based pharmacokinetic (PBPK), and thyroid hormone kinetics models. Curr Res Toxicol 2023; 5:100138. [PMID: 38074188 PMCID: PMC10697989 DOI: 10.1016/j.crtox.2023.100138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 03/22/2024] Open
Abstract
The thyroid hormones play key roles in physiological processes such as regulation of the metabolic and cardiac systems as well as the development of the brain and surrounding sympathetic nervous system. Recent efforts to screen environmental chemicals for their ability to alter thyroid hormone synthesis, transport, metabolism and/or function have identified novel chemicals that target key processes in the thyroid pathway. One newly identified chemical, oxyfluorfen, is a diphenyl-ether herbicide used for control of annual broadleaf and grassy weeds in a variety of tree fruit, nut, vine, and field crops. Using in vitro high-throughput screening (HTS) assays, oxyfluorofen was identified to be a potent inhibitor of the thyroidal sodium-iodide symporter (NIS). To quantitatively assess this inhibition mechanism in vivo, we extrapolated in vitro NIS inhibition data to in vivo disruption of thyroid hormones synthesis in rats using physiologically based pharmacokinetic (PBPK) and thyroid hormone kinetics models. The overall computational model (chemical PBPK and THs kinetic sub-models) was calibrated against in vivo data for the levels of oxyfluorfen in thyroid tissue and serum and against serum levels of thyroid hormones triiodothyronine (T3) and thyroxine (T4) in rats. The rat thyroid model was then extrapolated to humans using human in vitro HTS data for NIS inhibition and the chemical specific hepatic clearance rate in humans. The overall species extrapolated PBPK-thyroid kinetics model can be used to predict dose-response (% drop in thyroid serum levels compared to homeostasis) relationships in humans. These relationships can be used to estimate points of departure for health risks related to a drop in serum levels of TH hormones based on HTS assays in vitro to in vivo extrapolation (IVIVE), toxicokinetics, and physiological principles.
Collapse
|
4
|
Steffens B, Koch G, Gächter P, Claude F, Gotta V, Bachmann F, Schropp J, Janner M, l'Allemand D, Konrad D, Welzel T, Szinnai G, Pfister M. Clinically practical pharmacometrics computer model to evaluate and personalize pharmacotherapy in pediatric rare diseases: application to Graves' disease. Front Med (Lausanne) 2023; 10:1099470. [PMID: 37206476 PMCID: PMC10188966 DOI: 10.3389/fmed.2023.1099470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/14/2023] [Indexed: 05/21/2023] Open
Abstract
Objectives Graves' disease (GD) with onset in childhood or adolescence is a rare disease (ORPHA:525731). Current pharmacotherapeutic approaches use antithyroid drugs, such as carbimazole, as monotherapy or in combination with thyroxine hormone substitutes, such as levothyroxine, as block-and-replace therapy to normalize thyroid function and improve patients' quality of life. However, in the context of fluctuating disease activity, especially during puberty, a considerable proportion of pediatric patients with GD is suffering from thyroid hormone concentrations outside the therapeutic reference ranges. Our main goal was to develop a clinically practical pharmacometrics computer model that characterizes and predicts individual disease activity in children with various severity of GD under pharmacotherapy. Methods Retrospectively collected clinical data from children and adolescents with GD under up to two years of treatment at four different pediatric hospitals in Switzerland were analyzed. Development of the pharmacometrics computer model is based on the non-linear mixed effects approach accounting for inter-individual variability and incorporating individual patient characteristics. Disease severity groups were defined based on free thyroxine (FT4) measurements at diagnosis. Results Data from 44 children with GD (75% female, median age 11 years, 62% receiving monotherapy) were analyzed. FT4 measurements were collected in 13, 15, and 16 pediatric patients with mild, moderate, or severe GD, with a median FT4 at diagnosis of 59.9 pmol/l (IQR 48.4, 76.8), and a total of 494 FT4 measurements during a median follow-up of 1.89 years (IQR 1.69, 1.97). We observed no notable difference between severity groups in terms of patient characteristics, daily carbimazole starting doses, and patient years. The final pharmacometrics computer model was developed based on FT4 measurements and on carbimazole or on carbimazole and levothyroxine doses involving two clinically relevant covariate effects: age at diagnosis and disease severity. Discussion We present a tailored pharmacometrics computer model that is able to describe individual FT4 dynamics under both, carbimazole monotherapy and carbimazole/levothyroxine block-and-replace therapy accounting for inter-individual disease progression and treatment response in children and adolescents with GD. Such clinically practical and predictive computer model has the potential to facilitate and enhance personalized pharmacotherapy in pediatric GD, reducing over- and underdosing and avoiding negative short- and long-term consequences. Prospective randomized validation trials are warranted to further validate and fine-tune computer-supported personalized dosing in pediatric GD and other rare pediatric diseases.
Collapse
Affiliation(s)
- Britta Steffens
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
- *Correspondence: Britta Steffens
| | - Gilbert Koch
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Pascal Gächter
- Pediatric Endocrinology and Diabetology, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Fabien Claude
- Pediatric Endocrinology and Diabetology, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Verena Gotta
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Freya Bachmann
- Department of Mathematics and Statistics, University of Konstanz, Konstanz, Germany
| | - Johannes Schropp
- Department of Mathematics and Statistics, University of Konstanz, Konstanz, Germany
| | - Marco Janner
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Dagmar l'Allemand
- Department of Pediatric Endocrinology and Diabetology, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Tatjana Welzel
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Gabor Szinnai
- Pediatric Endocrinology and Diabetology, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
5
|
Koch G, Steffens B, Leroux S, Gotta V, Schropp J, Gächter P, Bachmann F, Welzel T, Janner M, L'Allemand D, Konrad D, Szinnai G, Pfister M. Modeling of levothyroxine in newborns and infants with congenital hypothyroidism: challenges and opportunities of a rare disease multi-center study. J Pharmacokinet Pharmacodyn 2021; 48:711-723. [PMID: 34117565 PMCID: PMC8405503 DOI: 10.1007/s10928-021-09765-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 05/12/2021] [Indexed: 11/22/2022]
Abstract
Modeling of retrospectively collected multi-center data of a rare disease in pediatrics is challenging because laboratory data can stem from several decades measured with different assays. Here we present a retrospective pharmacometrics (PMX) based data analysis of the rare disease congenital hypothyroidism (CH) in newborns and infants. Our overall aim is to develop a model that can be applied to optimize dosing in this pediatric patient population since suboptimal treatment of CH during the first 2 years of life is associated with a reduced intelligence quotient between 10 and 14 years. The first goal is to describe a retrospectively collected dataset consisting of 61 newborns and infants with CH up to 2 years of age. Overall, 505 measurements of free thyroxine (FT4) and 510 measurements of thyrotropin or thyroid-stimulating hormone were available from patients receiving substitution treatment with levothyroxine (LT4). The second goal is to introduce a scale/location-scale normalization method to merge available FT4 measurements since 34 different postnatal age- and assay-specific laboratory reference ranges were applied. This method takes into account the change of the distribution of FT4 values over time, i.e. a transformation from right-skewed towards normality during LT4 treatment. The third goal is to develop a practical and useful PMX model for LT4 treatment to characterize FT4 measurements, which is applicable within a clinical setting. In summary, a time-dependent normalization method and a practical PMX model are presented. Since there is no on-going or planned development of new pharmacological approaches for CH, PMX based modeling and simulation can be leveraged to personalize dosing with the goal to enhance longer-term neurological outcome in children with the rare disease CH.
Collapse
Affiliation(s)
- Gilbert Koch
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland.
| | - Britta Steffens
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Stephanie Leroux
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Verena Gotta
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Johannes Schropp
- Department of Mathematics and Statistics, University of Konstanz, Konstanz, Germany
| | - Pascal Gächter
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
- Pediatric Endocrinology and Diabetology, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Freya Bachmann
- Department of Mathematics and Statistics, University of Konstanz, Konstanz, Germany
| | - Tatjana Welzel
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
| | - Marco Janner
- Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, Bern University Hospital , University of Bern, Bern, Switzerland
| | - Dagmar L'Allemand
- Department of Pediatric Endocrinology and Diabetology, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Gabor Szinnai
- Pediatric Endocrinology and Diabetology, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel UKBB, University of Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
6
|
Handa S, Hassan I, Gilbert M, El-Masri H. Mechanistic Computational Model for Extrapolating In vitro Thyroid Peroxidase (TPO) Inhibition Data to Predict Serum Thyroid Hormone Levels in Rats. Toxicol Sci 2021; 183:36-48. [PMID: 34117770 DOI: 10.1093/toxsci/kfab074] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
High throughput (HTP) in vitro assays are developed to screen chemicals for their potential to inhibit thyroid hormones (THs) synthesis. Some of these experiments, such as the thyroid peroxidase (TPO) inhibition assay, are based on thyroid microsomal extracts. However, the regulation of thyroid disruption chemicals (TDCs) is based on THs in vivo serum levels. This necessitates the estimation of TDCs in vivo tissue levels in the thyroid where THs synthesis inhibition by TPO takes place. The in vivo tissue levels of chemicals are controlled by pharmacokinetic determinants such as absorption, distribution, metabolism and excretion (ADME), and can be described quantitatively in physiologically based pharmacokinetic (PBPK) models. An integrative computational model including chemical specific PBPK and TH kinetics models provides a mechanistic quantitative approach to translate thyroidal HTP in vitro assays to in vivo measures of circulating THs serum levels. This computational framework is developed to quantitatively establish the linkage between applied dose, chemical thyroid tissue levels, thyroid TPO inhibition potential, and in vivo TH serum levels. Once this link is established quantitively, the overall model is used to calibrate the TH kinetics parameters using experimental data for THs levels in thyroid tissue and serum for the two drugs Propylthiouracil (PTU) and Methimazole (MMI). The calibrated quantitative framework is then evaluated against literature data for the environmental chemical ethylenethiourea (ETU). The linkage of PBPK and TH kinetics models illustrates a computational framework that can be extrapolated to humans to screen chemicals based on their exposure levels and potential to disrupt serum THs levels in vivo.
Collapse
Affiliation(s)
- Sakshi Handa
- Center for Computational Toxicology and Exposure, Research Triangle Park, NC
| | - Iman Hassan
- Office of Air Quality Planning and Standards, Research Triangle Park, NC
| | - Mary Gilbert
- Center for Public Health and Environmental Assessment, Research Triangle Park, NC
| | - Hisham El-Masri
- Center for Computational Toxicology and Exposure, Research Triangle Park, NC
| |
Collapse
|
7
|
Bradshaw EL, Spilker ME, Zang R, Bansal L, He H, Jones RD, Le K, Penney M, Schuck E, Topp B, Tsai A, Xu C, Nijsen MJ, Chan JR. Applications of Quantitative Systems Pharmacology in Model-Informed Drug Discovery: Perspective on Impact and Opportunities. CPT Pharmacometrics Syst Pharmacol 2019; 8:777-791. [PMID: 31535440 PMCID: PMC6875708 DOI: 10.1002/psp4.12463] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Quantitative systems pharmacology (QSP) approaches have been increasingly applied in the pharmaceutical since the landmark white paper published in 2011 by a National Institutes of Health working group brought attention to the discipline. In this perspective, we discuss QSP in the context of other modeling approaches and highlight the impact of QSP across various stages of drug development and therapeutic areas. We discuss challenges to the field as well as future opportunities.
Collapse
Affiliation(s)
| | - Mary E. Spilker
- Pfizer Worldwide Research and DevelopmentSan DiegoCaliforniaUSA
| | | | | | - Handan He
- Novartis Institutes for Biomedical ResearchEast HanoverNew JerseyUSA
| | | | - Kha Le
- AgiosCambridgeMassachusettsUSA
| | | | | | | | - Alice Tsai
- Vertex Pharmaceuticals IncorporatedBostonMassachusettsUSA
| | | | | | | |
Collapse
|
8
|
Helmlinger G, Sokolov V, Peskov K, Hallow KM, Kosinsky Y, Voronova V, Chu L, Yakovleva T, Azarov I, Kaschek D, Dolgun A, Schmidt H, Boulton DW, Penland RC. Quantitative Systems Pharmacology: An Exemplar Model-Building Workflow With Applications in Cardiovascular, Metabolic, and Oncology Drug Development. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:380-395. [PMID: 31087533 PMCID: PMC6617832 DOI: 10.1002/psp4.12426] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022]
Abstract
Quantitative systems pharmacology (QSP), a mechanistically oriented form of drug and disease modeling, seeks to address a diverse set of problems in the discovery and development of therapies. These problems bring a considerable amount of variability and uncertainty inherent in the nonclinical and clinical data. Likewise, the available modeling techniques and related software tools are manifold. Appropriately, the development, qualification, application, and impact of QSP models have been similarly varied. In this review, we describe the progressive maturation of a QSP modeling workflow: a necessary step for the efficient, reproducible development and qualification of QSP models, which themselves are highly iterative and evolutive. Furthermore, we describe three applications of QSP to impact drug development; one supporting new indications for an approved antidiabetic clinical asset through mechanistic hypothesis generation, one highlighting efficacy and safety differentiation within the sodium‐glucose cotransporter‐2 inhibitor drug class, and one enabling rational selection of immuno‐oncology drug combinations.
Collapse
Affiliation(s)
- Gabriel Helmlinger
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Boston, Massachusetts, USA
| | | | - Kirill Peskov
- M&S Decisions LLC, Moscow, Russia.,Computational Oncology Group, I.M. Sechenov First Moscow State Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Karen M Hallow
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, Georgia, USA.,Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia, USA
| | | | | | - Lulu Chu
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Boston, Massachusetts, USA
| | | | | | | | | | | | - David W Boulton
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Gaithersburg, Maryland, USA
| | - Robert C Penland
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Collins T, Gray K, Bista M, Skinner M, Hardy C, Wang H, Mettetal JT, Harmer AR. Quantifying the relationship between inhibition of VEGF receptor 2, drug-induced blood pressure elevation and hypertension. Br J Pharmacol 2018; 175:618-630. [PMID: 29161763 DOI: 10.1111/bph.14103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/20/2017] [Accepted: 11/11/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Several anti-angiogenic cancer drugs that inhibit VEGF receptor (VEGFR) signalling for efficacy are associated with a 15-60% incidence of hypertension. Tyrosine kinase inhibitors (TKIs) that have off-target activity at VEGFR-2 may also cause blood pressure elevation as an undesirable side effect. Therefore, the ability to translate VEGFR-2 off-target potency into blood pressure elevation would be useful in development of novel TKIs. Here, we have sought to quantify the relationship between VEGFR-2 inhibition and blood pressure elevation for a range of kinase inhibitors. EXPERIMENTAL APPROACH Porcine aortic endothelial cells overexpressing VEGFR-2 (PAE) were used to determine IC50 for VEGFR-2 phosphorylation. These IC50 values were compared with published reports of exposure attained during clinical use and the corresponding incidence of all-grade hypertension. Unbound average plasma concentration (Cav,u ) was selected to be the most appropriate pharmacokinetic parameter. The pharmacokinetic-pharmacodynamic (PKPD) relationship for blood pressure elevation was investigated for selected kinase inhibitors, using data derived either from clinical papers or from rat telemetry experiments. KEY RESULTS All-grade hypertension was predominantly observed when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . Furthermore, based on the PKPD analysis, an exposure-dependent blood pressure elevation >1 mmHg was observed only when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . CONCLUSIONS AND IMPLICATIONS Taken together, these data show that the risk of blood pressure elevation is proportional to the amount of VEGFR-2 inhibition, and a margin of >10-fold between VEGFR-2 IC50 and Cav,u appears to confer a minimal risk of hypertension.
Collapse
Affiliation(s)
- Teresa Collins
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Kelly Gray
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Michal Bista
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Matt Skinner
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Christopher Hardy
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| | - Haiyun Wang
- AstraZeneca, Gatehouse Park, Waltham, MA, 02451, USA
| | | | - Alexander R Harmer
- AstraZeneca, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4, 0WG, UK
| |
Collapse
|
10
|
Helmlinger G, Al-Huniti N, Aksenov S, Peskov K, Hallow KM, Chu L, Boulton D, Eriksson U, Hamrén B, Lambert C, Masson E, Tomkinson H, Stanski D. Drug-disease modeling in the pharmaceutical industry - where mechanistic systems pharmacology and statistical pharmacometrics meet. Eur J Pharm Sci 2017; 109S:S39-S46. [PMID: 28506868 DOI: 10.1016/j.ejps.2017.05.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 10/19/2022]
Abstract
Modeling & simulation (M&S) methodologies are established quantitative tools, which have proven to be useful in supporting the research, development (R&D), regulatory approval, and marketing of novel therapeutics. Applications of M&S help design efficient studies and interpret their results in context of all available data and knowledge to enable effective decision-making during the R&D process. In this mini-review, we focus on two sets of modeling approaches: population-based models, which are well-established within the pharmaceutical industry today, and fall under the discipline of clinical pharmacometrics (PMX); and systems dynamics models, which encompass a range of models of (patho-)physiology amenable to pharmacological intervention, of signaling pathways in biology, and of substance distribution in the body (today known as physiologically-based pharmacokinetic models) - which today may be collectively referred to as quantitative systems pharmacology models (QSP). We next describe the convergence - or rather selected integration - of PMX and QSP approaches into 'middle-out' drug-disease models, which retain selected mechanistic aspects, while remaining parsimonious, fit-for-purpose, and able to address variability and the testing of covariates. We further propose development opportunities for drug-disease systems models, to increase their utility and applicability throughout the preclinical and clinical spectrum of pharmaceutical R&D.
Collapse
Affiliation(s)
- Gabriel Helmlinger
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Waltham, MA, USA.
| | - Nidal Al-Huniti
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Waltham, MA, USA
| | - Sergey Aksenov
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Waltham, MA, USA
| | | | - Karen M Hallow
- College of Public Health, University of Georgia, Athens, GA, USA; College of Engineering, University of Georgia, Athens, GA, USA
| | - Lulu Chu
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Waltham, MA, USA
| | - David Boulton
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gaithersburg, MD, USA
| | - Ulf Eriksson
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Bengt Hamrén
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Craig Lambert
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Eric Masson
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Waltham, MA, USA
| | - Helen Tomkinson
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Donald Stanski
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
11
|
Leonard JA, Tan YM, Gilbert M, Isaacs K, El-Masri H. Estimating Margin of Exposure to Thyroid Peroxidase Inhibitors Using High-Throughput in vitro Data, High-Throughput Exposure Modeling, and Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling. Toxicol Sci 2016; 151:57-70. [PMID: 26865668 PMCID: PMC4914794 DOI: 10.1093/toxsci/kfw022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Some pharmaceuticals and environmental chemicals bind the thyroid peroxidase (TPO) enzyme and disrupt thyroid hormone production. The potential for TPO inhibition is a function of both the binding affinity and concentration of the chemical within the thyroid gland. The former can be determined through in vitro assays, and the latter is influenced by pharmacokinetic properties, along with environmental exposure levels. In this study, a physiologically based pharmacokinetic (PBPK) model was integrated with a pharmacodynamic (PD) model to establish internal doses capable of inhibiting TPO in relation to external exposure levels predicted through exposure modeling. The PBPK/PD model was evaluated using published serum or thyroid gland chemical concentrations or circulating thyroxine (T4) and triiodothyronine (T3) hormone levels measured in rats and humans. After evaluation, the model was used to estimate human equivalent intake doses resulting in reduction of T4 and T3 levels by 10% (ED10) for 6 chemicals of varying TPO-inhibiting potencies. These chemicals were methimazole, 6-propylthiouracil, resorcinol, benzophenone-2, 2-mercaptobenzothiazole, and triclosan. Margin of exposure values were estimated for these chemicals using the ED10 and predicted population exposure levels for females of child-bearing age. The modeling approach presented here revealed that examining hazard or exposure alone when prioritizing chemicals for risk assessment may be insufficient, and that consideration of pharmacokinetic properties is warranted. This approach also provides a mechanism for integrating in vitro data, pharmacokinetic properties, and exposure levels predicted through high-throughput means when interpreting adverse outcome pathways based on biological responses.
Collapse
Affiliation(s)
- Jeremy A Leonard
- *Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, 37831; National Exposure Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, North Carolina, 27709
| | - Yu-Mei Tan
- National Exposure Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, North Carolina, 27709
| | - Mary Gilbert
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, North Carolina, 27709
| | - Kristin Isaacs
- National Exposure Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, North Carolina, 27709
| | - Hisham El-Masri
- National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Research Triangle Park, North Carolina, 27709
| |
Collapse
|
12
|
Visser SAG, de Alwis DP, Kerbusch T, Stone JA, Allerheiligen SRB. Implementation of quantitative and systems pharmacology in large pharma. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e142. [PMID: 25338195 PMCID: PMC4474169 DOI: 10.1038/psp.2014.40] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/30/2014] [Indexed: 02/04/2023]
Abstract
Quantitative and systems pharmacology concepts and tools are the foundation of the model-informed drug development paradigm at Merck for integrating knowledge, enabling decisions, and enhancing submissions. Rigorous prioritization of modeling and simulation activities has enabled key drug development decisions and led to a high return on investment through significant cost avoidance. Critical factors for the successful implementation, examples on impact on decision making with associated return of investment, and drivers for continued success are discussed.
Collapse
Affiliation(s)
- S A G Visser
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| | - D P de Alwis
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| | - T Kerbusch
- Quantitive Pharmacology and Pharmacometrics, MSD, Oss, The Netherlands
| | - J A Stone
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| | - S R B Allerheiligen
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| |
Collapse
|
13
|
The hypothalamic-pituitary-thyroid axis in infants and children: protection from radioiodines. J Thyroid Res 2014; 2014:710178. [PMID: 24971190 PMCID: PMC4058186 DOI: 10.1155/2014/710178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/07/2014] [Indexed: 11/17/2022] Open
Abstract
Potassium iodide (KI) is recommended as an emergency treatment for exposure to radioiodines, most commonly associated with nuclear detonation or mishaps at nuclear power plants. Protecting the thyroid gland of infants and children remains a priority because of increased incidence of thyroid cancer in the young exposed to radioiodines (such as (131)I and (133)I). There is a lack of clinical studies for KI and radioiodines in children or infants to draw definitive conclusions about the effectiveness and safety of KI administration in the young. In this paper, we compare functional aspects of the hypothalamic-pituitary-thyroid (HPT) axis in the young and adults and review the limited studies of KI in children. The HPT axis in the infant and child is hyperactive and therefore will respond less effectively to KI treatment compared to adults. Research on the safety and efficacy of KI in infants and children is needed.
Collapse
|