Hierarchical Mechanistic Modelling of Clinical Pharmacokinetic Data

Total Page:16

File Type:pdf, Size:1020Kb

Hierarchical Mechanistic Modelling of Clinical Pharmacokinetic Data Hierarchical mechanistic modelling of clinical pharmacokinetic data A thesis submitted to the University of Manchester for the degree of Doctor of Philosophy in the Faculty of Medical and Human Sciences 2015 Thierry Wendling Manchester Pharmacy School Contents List of Figures ........................................................................................................ 6 List of Tables .......................................................................................................... 9 List of abbreviations ............................................................................................ 10 Abstract ................................................................................................................ 13 Declaration ........................................................................................................... 14 Copyright ............................................................................................................. 14 Acknowledgements .............................................................................................. 15 Chapter 1: General introduction ...................................................................... 16 1.1. Modelling and simulation in drug development ........................................ 17 1.1.1. The drug development process .......................................................... 17 1.1.2. The role of pharmacokinetic and pharmacodynamic modelling in drug development ................................................................................................... 18 1.2. Mechanistic modelling of clinical pharmacokinetic data .......................... 23 1.2.1. Physiologically-based pharmacokinetic models ................................ 23 1.2.2. Oral absorption models .................................................................... 26 1.2.3. Hierarchical pharmacokinetic modelling .......................................... 29 1.3. Modelling survival data in cancer research ............................................... 32 1.3.1. Pharmacodynamics in oncology........................................................ 32 1.3.2. Survival analysis ............................................................................... 33 1.4. Project objectives ..................................................................................... 38 1.5. List of manuscripts and author contribution statement .............................. 39 1.6. References ............................................................................................... 42 Chapter 2: Model-based evaluation of the impact of formulation and food intake on the complex oral absorption of mavoglurant in healthy subjects ...... 50 Chapter 3: Application of a Bayesian approach to physiological modelling of mavoglurant population pharmacokinetics ........................................................ 51 2 Chapter 4: Reduction of a whole-body physiologically-based pharmacokinetic model to stabilise the Bayesian analysis of clinical data ..................................... 52 Chapter 5: Predicting survival of pancreatic cancer patients treated with gemcitabine using longitudinal tumour size data ............................................... 53 5.1. Abstract ................................................................................................... 54 5.1.1. Background ...................................................................................... 54 5.1.2. Methods ............................................................................................ 54 5.1.3. Results .............................................................................................. 54 5.1.4. Conclusions ...................................................................................... 54 5.2. Introduction ............................................................................................. 55 5.3. Materials and methods ............................................................................. 56 5.3.1. Data ................................................................................................. 56 5.3.2. Statistical analysis ............................................................................ 57 5.3.2.1. Screening risk factors ..................................................................... 57 5.3.2.2. Modelling tumour size time-series ................................................. 58 5.3.2.3. Survival analysis ............................................................................ 59 5.3.2.4. Validation ...................................................................................... 61 5.4. Results ..................................................................................................... 62 5.4.1. Baseline risk factors ......................................................................... 62 5.4.2. Tumour size time-series model .......................................................... 62 5.4.3. Survival analysis ............................................................................... 64 5.4.3.1. The PAR approach ......................................................................... 65 5.4.3.2. The COX approach ........................................................................ 66 5.4.4. Predictive performance ..................................................................... 67 5.5. Discussion ............................................................................................... 69 5.6. Acknowledgements .................................................................................. 72 5.7. Conflict of interest ................................................................................... 72 5.8. References ............................................................................................... 73 3 Chapter 6: General discussion .......................................................................... 76 Appendix A1: Supplementary material for Chapter 2 ....................................... 80 A1.1. Supplementary methods ............................................................................ 81 A1.1.1. Implementation of dose superimposition in NONMEM using a sum of inverse Gaussian functions as an input rate function ...................................... 81 A1.1.1.1. Comparison of the FORTRAN subroutine approach with the reference method, using simulations ........................................................... 81 A1.1.1.2. Verification of the partial derivatives defined in the FORTRAN subroutine .................................................................................................. 82 A1.2. Supplementary results ............................................................................... 83 A1.2.1. Individual goodness-of-fit plots for each formulation-food conditions 83 A1.2.2. Implementation of dose superimposition in NONMEM using a sum of inverse Gaussian functions as an input rate function ...................................... 85 A1.2.2.1. Comparison of the FORTRAN subroutine approach with a reference method using simulations ............................................................ 85 A1.2.2.2. Verification of the partial derivatives .......................................... 86 A1.3. User-defined FORTRAN functions FUNCA, FUNCB and FUNCC (subroutine referred as sumdoset_3IG.f90 in Appendix A1.4) ............................ 87 A1.4. Dose superposition in NONMEM using the user-supplied FORTRAN subroutine (example of the MR-fasted model using a sum of three IG functions as input rate function) ............................................................................................. 90 A1.5. Dose superposition performed solely in a NM-TRAN abbreviated code for mavoglurant MR-fasted model (reference method) ............................................. 92 A1.6. References ................................................................................................ 95 Appendix A2: Supplementary material for Chapter 3 ....................................... 96 A2.1. Prediction of mavoglurant hepatic intrinsic clearance from recombinant human cytochrome P450 enzyme kinetic data .................................................... 97 A2.2. Estimation of the formulation-specific dissolution constant z from in vitro dissolution profiles ............................................................................................. 99 4 A2.3. Prediction of mavoglurant effective permeability in human jejunum ....... 100 A2.4. Calculation of systemic bioavailability ................................................... 101 A2.5. Drug-drug interaction between mavoglurant and ketoconazole in adult subjects ............................................................................................................ 102 A2.6. Results of the Bayesian analysis of Study 1 data when estimating all drug- specific parameters ........................................................................................... 104 A2.7. References .............................................................................................. 104 Appendix A3: Supplementary material for Chapter 4 ..................................... 106 A3.1. Supplementary tables .............................................................................. 107 A3.2. Supplementary figures ............................................................................ 109 Appendix A4: Supplementary material for Chapter 5 ..................................... 112
Recommended publications
  • Metabotropic Glutamate Receptors
    mGluR Metabotropic glutamate receptors mGluR (metabotropic glutamate receptor) is a type of glutamate receptor that are active through an indirect metabotropic process. They are members of thegroup C family of G-protein-coupled receptors, or GPCRs. Like all glutamate receptors, mGluRs bind with glutamate, an amino acid that functions as an excitatoryneurotransmitter. The mGluRs perform a variety of functions in the central and peripheral nervous systems: mGluRs are involved in learning, memory, anxiety, and the perception of pain. mGluRs are found in pre- and postsynaptic neurons in synapses of the hippocampus, cerebellum, and the cerebral cortex, as well as other parts of the brain and in peripheral tissues. Eight different types of mGluRs, labeled mGluR1 to mGluR8, are divided into groups I, II, and III. Receptor types are grouped based on receptor structure and physiological activity. www.MedChemExpress.com 1 mGluR Agonists, Antagonists, Inhibitors, Modulators & Activators (-)-Camphoric acid (1R,2S)-VU0155041 Cat. No.: HY-122808 Cat. No.: HY-14417A (-)-Camphoric acid is the less active enantiomer (1R,2S)-VU0155041, Cis regioisomer of VU0155041, is of Camphoric acid. Camphoric acid stimulates a partial mGluR4 agonist with an EC50 of 2.35 osteoblast differentiation and induces μM. glutamate receptor expression. Camphoric acid also significantly induced the activation of NF-κB and AP-1. Purity: ≥98.0% Purity: ≥98.0% Clinical Data: No Development Reported Clinical Data: No Development Reported Size: 10 mM × 1 mL, 100 mg Size: 10 mM × 1 mL, 5 mg, 10 mg, 25 mg (2R,4R)-APDC (R)-ADX-47273 Cat. No.: HY-102091 Cat. No.: HY-13058B (2R,4R)-APDC is a selective group II metabotropic (R)-ADX-47273 is a potent mGluR5 positive glutamate receptors (mGluRs) agonist.
    [Show full text]
  • The Role of Modelling and Simulation in Drug Development
    The Role of Modelling and Simulation in Drug Development Leon Aarons Manchester Pharmacy School The University of Manchester Plan • Overview • Role of modelling in Phase I drug development • Role of modelling in the development of anti- cancer agents • Summary Models The type of model to be developed should be driven by the available information and the goal of the simulations Category Information Goal needed Explanatory Rich Extrapolation in complex and Mechanistic variable environment models Semi-mechanistic models Empirical Interpolation in Descriptive models “Poor” simple and stable environment Days Months Years 100 10 12tt C() t C12 e C e 1 concentration 0.1 0.01 0 2 4 6 8 10 time k12 1 2 k21 k10 dC() t V1 kVCt()()() kVCt kVCt 1dt 1211 1011 2122 dC() t V2 k V C()() t k V C t 2dt 12 1 1 21 2 2 PBPK MODEL dC() t KpV out, H QCtQC ()().() tCLKpC t H Hdt H A H out, H int H out , H Role of Modelling and Simulation in Phase I Drug Development L. Aarons, M. Karlsson, F. Mentré, F. Rombout, J.-L. Steimer, A. van Peer COST B15 Brussels January 10-11, 2000 Eur.J.Pharm.Sci. 13, 115-122 (2001) COST European COoperation in the Field of Scientific and Technical Research Definition of Phase I • Phase I is the transition Phase from preclinical to clinical evaluation •Learning process primarily focused on: - Safety (Tolerability) - PK/PD - Metabolism - Potential interactions (drugs/food) - Dosage form development (equivalence) Phase I Safety ¥ Tolerability Data • FIM and S/MD study wide dose range • Escalation design • Healthy subjects
    [Show full text]
  • Emergency Use Authorization (EUA) for Bamlanivimab 700 Mg and Etesevimab 1400 Mg IV Administered Together Center for Drug Evaluation and Research (CDER) Review
    ________________________________________________________________ Emergency Use Authorization (EUA) for Bamlanivimab 700 mg and Etesevimab 1400 mg IV Administered Together Center for Drug Evaluation and Research (CDER) Review Identifying Information Application Type (EUA or Pre-EUA) EUA If EUA, designate whether pre-event or intra-event EUA request. 1 EUA Application Number(s) EUA 000094 Sponsor (entity requesting EUA or Eli Lilly and Company pre-EUA consideration), point of Christine Phillips, PhD, RAC contact, address, phone number, fax Advisor, Global Regulatory Affairs - NA (b) (6) number, email address Mobile: Email: [email protected] Manufacturer, if different from Eli Lilly and Company Sponsor Submission Date(s) November 16, 2020 Receipt Date(s) November 16, 2020 OND Division / Office Division of Antivirals/Office of Infectious Diseases Reviewer Name(s)/Discipline(s) Natalie Pica, MD, PhD – Clinical Reviewer Wendy Carter, DO – Cross Discipline Team Lead Michael Thomson, PhD – Clinical Virology Reviewer Jules O’Rear, PhD – Clinical Virology Team Lead Daniel Rubin, PhD – Statistics Reviewer Thamban Valappil, PhD – Statistics Team Lead Dionne Price, PhD – Director, Division of Biometrics IV Jason Moore, PharmD – Clinical Pharmacology Reviewer Su-Young Choi PharmD, PhD – Clinical Pharmacology Team Lead Kellie Reynolds, PharmD – Division Director, DIDP Ye Xiong, PhD – Pharmacometrics Reviewer Justin Earp, PhD – Pharmacometrics Team Lead Yaning Wang, PhD – Division Director, DPM 1 If a Pre-EUA is in existence at the time
    [Show full text]
  • Draft COMP Agenda 16-18 January 2018
    12 January 2018 EMA/COMP/818236/2017 Inspections, Human Medicines Pharmacovigilance and Committees Committee for Orphan Medicinal Products (COMP) Draft agenda for the meeting on 16-18 January 2018 Chair: Bruno Sepodes – Vice-Chair: Lesley Greene 16 January 2018, 09:00-19:30, room 2F 17 January 2018, 08:30-19:30, room 2F 18 January 2018, 08:30-18:30, room 2F Health and safety information In accordance with the Agency’s health and safety policy, delegates are to be briefed on health, safety and emergency information and procedures prior to the start of the meeting. Disclaimers Some of the information contained in this agenda is considered commercially confidential or sensitive and therefore not disclosed. With regard to intended therapeutic indications or procedure scopes listed against products, it must be noted that these may not reflect the full wording proposed by applicants and may also vary during the course of the review. Additional details on some of these procedures will be published in the COMP meeting reports once the procedures are finalised. Of note, this agenda is a working document primarily designed for COMP members and the work the Committee undertakes. Note on access to documents Some documents mentioned in the agenda cannot be released at present following a request for access to documents within the framework of Regulation (EC) No 1049/2001 as they are subject to on- going procedures for which a final decision has not yet been adopted. They will become public when adopted or considered public according to the principles stated in the Agency policy on access to documents (EMA/127362/2006).
    [Show full text]
  • Real Projects with Real Impacts Dave Handelsman, Project Data Sphere, LLC, Cary, NC, USA
    PhUSE US Connect 2019 Paper AB06 The Power of Data Sharing: Real Projects with Real Impacts Dave Handelsman, Project Data Sphere, LLC, Cary, NC, USA ABSTRACT Data sharing leading to improved cancer patient outcomes provides exciting examples of applying Big Data and Analytics to clinical research. Since 2014, Project Data Sphere has aggregated patient-level data from more than 135,000 patients spanning 150 trials, and made this data available for exploration and investigation to more than 2,000 researchers. This successful data sharing program has enabled medically relevant insights to be discovered and published in leading peer-accepted journals, and new research programs are underway, including: • Development of an external control arm for small cell lung cancer that may eliminate the need to enroll cancer patients in standard-of-care comparator treatment arms • Development of machine learning tools for tumor image processing • Investigations into rare but serious adverse events associated with new of immuno-oncology therapies In this session, the opportunities and challenges of clinical trial data sharing will be discussed as they relate to Big Data and Analytics. INTRODUCTION Data sharing has broad meaning, even when discussed in the context of clinical research. Depending on the context, this concept can refer to the sharing of patient-level data traditionally associated with clinical trials, or it can alternatively (or additionally) refer to less traditional data types such as genomics and images, and can also refer to data extracted from electronic health records. For the purposes of this paper, data sharing will be discussed in the context of data made available from completed clinical trials.
    [Show full text]
  • Patent Application Publication ( 10 ) Pub . No . : US 2019 / 0192440 A1
    US 20190192440A1 (19 ) United States (12 ) Patent Application Publication ( 10) Pub . No. : US 2019 /0192440 A1 LI (43 ) Pub . Date : Jun . 27 , 2019 ( 54 ) ORAL DRUG DOSAGE FORM COMPRISING Publication Classification DRUG IN THE FORM OF NANOPARTICLES (51 ) Int . CI. A61K 9 / 20 (2006 .01 ) ( 71 ) Applicant: Triastek , Inc. , Nanjing ( CN ) A61K 9 /00 ( 2006 . 01) A61K 31/ 192 ( 2006 .01 ) (72 ) Inventor : Xiaoling LI , Dublin , CA (US ) A61K 9 / 24 ( 2006 .01 ) ( 52 ) U . S . CI. ( 21 ) Appl. No. : 16 /289 ,499 CPC . .. .. A61K 9 /2031 (2013 . 01 ) ; A61K 9 /0065 ( 22 ) Filed : Feb . 28 , 2019 (2013 .01 ) ; A61K 9 / 209 ( 2013 .01 ) ; A61K 9 /2027 ( 2013 .01 ) ; A61K 31/ 192 ( 2013. 01 ) ; Related U . S . Application Data A61K 9 /2072 ( 2013 .01 ) (63 ) Continuation of application No. 16 /028 ,305 , filed on Jul. 5 , 2018 , now Pat . No . 10 , 258 ,575 , which is a (57 ) ABSTRACT continuation of application No . 15 / 173 ,596 , filed on The present disclosure provides a stable solid pharmaceuti Jun . 3 , 2016 . cal dosage form for oral administration . The dosage form (60 ) Provisional application No . 62 /313 ,092 , filed on Mar. includes a substrate that forms at least one compartment and 24 , 2016 , provisional application No . 62 / 296 , 087 , a drug content loaded into the compartment. The dosage filed on Feb . 17 , 2016 , provisional application No . form is so designed that the active pharmaceutical ingredient 62 / 170, 645 , filed on Jun . 3 , 2015 . of the drug content is released in a controlled manner. Patent Application Publication Jun . 27 , 2019 Sheet 1 of 20 US 2019 /0192440 A1 FIG .
    [Show full text]
  • Title Page Metabolism and Disposition of the Metabotropic Glutamate
    DMD Fast Forward. Published on June 17, 2013 as DOI: 10.1124/dmd.112.050716 DMD FastThis article Forward. has not been Published copyedited onand Juneformatted. 17, The 2013 final versionas doi:10.1124/dmd.112.050716 may differ from this version. DMD#50716 Title Page Metabolism and disposition of the metabotropic glutamate receptor 5 antagonist (mGluR5) mavoglurant (AFQ056) in healthy subjects Downloaded from Markus Walles, Thierry Wolf, Yi Jin, Michael Ritzau, Luc Alexis Leuthold, Joel Krauser, dmd.aspetjournals.org Hans-Peter Gschwind, David Carcache, Matthias Kittelmann, Magdalena Ocwieja, Mike Ufer, Ralph Woessner, Abhijit Chakraborty and Piet Swart at ASPET Journals on September 27, 2021 Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, Basel Switzerland (M.W., T.W., Y.J., L.A.L., J. K. H.-P.G, R.W., A.C., P.S.) Analytical Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.R.) Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.K., D.C.) Clinical Science & Innovation Novartis Institutes for Biomedical Research, Basel, Switzerland (M.O.) Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.U.) 1 Copyright 2013 by the American Society for Pharmacology and Experimental Therapeutics. DMD Fast Forward. Published on June 17, 2013 as DOI: 10.1124/dmd.112.050716 This article has not been copyedited and formatted. The final version may differ from this version. DMD#50716 Running Title Page Running title:
    [Show full text]
  • Application of Pharmacometric Methods to Improve Pediatric Drug Development
    Virginia Commonwealth University VCU Scholars Compass Theses and Dissertations Graduate School 2011 Application of Pharmacometric Methods to Improve Pediatric Drug Development Mallika Lala Virginia Commonwealth University Follow this and additional works at: https://scholarscompass.vcu.edu/etd Part of the Pharmacy and Pharmaceutical Sciences Commons © The Author Downloaded from https://scholarscompass.vcu.edu/etd/2467 This Dissertation is brought to you for free and open access by the Graduate School at VCU Scholars Compass. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of VCU Scholars Compass. For more information, please contact [email protected]. School of Pharmacy Virginia Commonwealth University This is to certify that the dissertation prepared by Mallika A. Lala entitled APPLICATION OF PHARMACOMETRIC METHODS TO IMPROVE PEDIATRIC DRUG DEVELOPMENT has been approved by her committee as satisfactory completion of the dissertation requirement for the degree of Doctor of Philosophy (Ph.D.) Jogarao V.S. Gobburu, FDA and F. Douglas Boudinot, School of Pharmacy Jurgen Venitz, School of Pharmacy Patricia W. Slattum, School of Pharmacy D’arcy P. Mays, College of Humanities and Sciences Susanna Wu-Pong, Graduate Program Director, School of Pharmacy Victor A. Yanchick, Dean, School of Pharmacy Dr. F. Douglas Boudinot, Dean of the Graduate School May 9th, 2011 i © Mallika A. Lala 2011 All Rights Reserved ii APPLICATION OF PHARMACOMETRIC METHODS TO IMPROVE PEDIATRIC DRUG DEVELOPMENT A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy at Virginia Commonwealth University. by MALLIKA A. LALA Bachelor of Pharmaceutical Science, UICT, India, 2006 Ph.D.
    [Show full text]
  • Emerging Preclinical Pharmacological Targets for Parkinson's Disease
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 20 Emerging preclinical pharmacological targets for Parkinson’s disease Sandeep Vasant More1 and Dong-Kug Choi1 1 Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea Correspondence to: Dong-Kug Choi, email: [email protected] Keywords: dopaminergic, Parkinson’s disease, pharmacological targets, neuroprotection, preclinical Received: October 08, 2015 Accepted: February 08, 2016 Published: March 15, 2016 ABSTRACT Parkinson’s disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT- 1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD.
    [Show full text]
  • Drug Structure–Transport Relationships
    J Pharmacokinet Pharmacodyn (2010) 37:541–573 DOI 10.1007/s10928-010-9174-0 Drug structure–transport relationships Michael S. Roberts Received: 10 August 2010 / Accepted: 22 October 2010 / Published online: 24 November 2010 Ó The Author(s) 2010. This article is published with open access at Springerlink.com Abstract Malcolm Rowland has greatly facilitated an understanding of drug structure–pharmacokinetic relationships using a physiological perspective. His view points, covering a wide range of activities, have impacted on my own work and on my appreciation and understanding of our science. This overview summarises some of our parallel activities, beginning with Malcolm’s work on the pH control of amphetamine excretion, his work on the disposition of aspirin and on the application of clearance concepts in describing the disposition of lidocaine. Malcolm also spent a considerable amount of time developing principles that define solute structure and transport/pharmacokinetic relationships using in situ organ studies, which he then extended to involve the whole body. Together, we developed a physiological approach to studying hepatic clearance, introducing the convection–dispersion model in which there was a spread in blood transit times through the liver accompanied by permeation into hepatocytes and removal by metabolism or excretion into the bile. With a range of colleagues, we then further developed the model and applied it to various organs in the body. One of Malcolm’s special interests was in being able to apply this knowledge, together with an understanding of physiological differences in scaling up pharmacokinetics from animals to man. The description of his many other activities, such as the development of clearance concepts, application of pharmacokinetics to the clinical situation and using Based on an invited presentation at the European Pharmaceutical Sciences Meeting: Pharmacokinetics: Spearheading advances and delivering the science (on the occasion of Professor Malcolm Rowland’s 70th birthday) London, October 5, 2009.
    [Show full text]
  • Rescue of Fmr1ko Phenotypes with Mglur5 Inhibitors MRZ-8456
    Neurobiology of Disease 119 (2018) 190–198 Contents lists available at ScienceDirect Neurobiology of Disease journal homepage: www.elsevier.com/locate/ynbdi KO Rescue of Fmr1 phenotypes with mGluR5 inhibitors: MRZ-8456 versus AFQ-056 T Pamela R. Westmarka,b, Andrzej Dekundyc, Andreas Graviusc, Wojciech Danyszc, ⁎ Cara J. Westmarka, a University of Wisconsin-Madison, Department of Neurology, Madison, WI, USA b University of Wisconsin-Madison, Department of Medicine, Madison, WI, USA c Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany ARTICLE INFO ABSTRACT Keywords: Metabotropic glutamate receptor 5 (mGluR5) is a drug target for central nervous system disorders such as fragile mGluR5 X syndrome that involve excessive glutamate-induced excitation. We tested the efficacy of a novel negative Fmr1 allosteric modulator of mGluR5 developed by Merz Pharmaceuticals, MRZ-8456, in comparison to MPEP and MRZ-8456 AFQ-056 (Novartis, a.k.a. mavoglurant) in both in vivo and in vitro assays in a mouse model of fragile X syn- AFQ-056 drome, Fmr1KO mice. The in vivo assays included susceptibility to audiogenic-induced seizures and pharmaco- MPEP kinetic measurements of drug availability. The in vitro assays included dose response assessments of biomarker Amyloid-beta precursor protein expression and dendritic spine length and density in cultured primary neurons. Both MRZ-8456 and AFQ-056 attenuated wild running and audiogenic-induced seizures in Fmr1KO mice with similar pharmacokinetic profiles. Both drugs significantly reduced dendritic expression of amyloid-beta protein precursor (APP) and rescued the ratio of mature to immature dendritic spines. These findings demonstrate that MRZ-8456, a drug being devel- oped for the treatment of motor complications of L-DOPA in Parkinson’s disease and which completed a phase I clinical trial, is effective in attenuating both well-established (seizures and dendritic spine maturity) and ex- ploratory biomarker (APP expression) phenotypes in a mouse model of fragile X syndrome.
    [Show full text]
  • Understanding Exposure-Receptor Occupancy Relationships For
    1521-0103/377/1/157–168$35.00 https://doi.org/10.1124/jpet.120.000371 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 377:157–168, April 2021 Copyright ª 2021 by The Author(s) This is an open access article distributed under the CC BY Attribution 4.0 International license. Understanding Exposure-Receptor Occupancy Relationships for Metabotropic Glutamate Receptor 5 Negative Allosteric Modulators across a Range of Preclinical and Clinical Studies Kirstie A. Bennett, Eugenia Sergeev, Cliona P. MacSweeney, Geor Bakker, and Anne E. Cooper Sosei Heptares, Cambridge, CB21 6DG, United Kingdom Received October 9, 2020; accepted January 26, 2021 Downloaded from ABSTRACT The metabotropic glutamate receptor 5 (mGlu5) is a recog- a unified exposure-response relationship was observed when nized central nervous system therapeutic target for which both unbound brain concentration and mGlu5 affinity were several negative allosteric modulator (NAM) drug candidates considered. This relationship showed ,10-fold overall differ- have or are continuing to be investigated for various disease ence, was fitted with a Hill slope that was not significantly indications in clinical development. Direct measurement of different from 1, and appeared consistent with a simple Emax jpet.aspetjournals.org target receptor occupancy (RO) is extremely useful to help model. This is the first time this type of comparison has been design and interpret efficacy and safety in nonclinical and conducted, demonstrating a unified brain exposure-RO re- clinical studies. In the mGlu5 field, this has been success- lationship across several species and mGlu5 NAMs with fully achieved by monitoring displacement of radiolabeled diverse properties.
    [Show full text]