1
|
Perazzolo S. SAAM II: A general mathematical modeling rapid prototyping environment. CPT Pharmacometrics Syst Pharmacol 2024; 13:1088-1102. [PMID: 38863172 PMCID: PMC11247119 DOI: 10.1002/psp4.13181] [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: 02/04/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
Simulation Analysis and Modeling II (SAAM II) is a graphical modeling software used in life sciences for compartmental model analysis, particularly, but not exclusively, appreciated in pharmacokinetics (PK) and pharmacodynamics (PD), metabolism, and tracer modeling. Its intuitive "circles and arrows" visuals allow users to easily build, solve, and fit compartmental models without the need for coding. It is suitable for rapid prototyping of models for complex kinetic analysis or PK/PD problems, and in educating students and non-modelers. Although it is straightforward in design, SAAM II incorporates sophisticated algorithms programmed in C to address ordinary differential equations, deal with complex systems via forcing functions, conduct multivariable regression featuring the Bayesian maximum a posteriori, perform identifiability and sensitivity analyses, and offer reporting functionalities, all within a single package. After 26 years from the last SAAM II tutorial paper, we demonstrate here SAAM II's updated applicability to current life sciences challenges. We review its features and present four contemporary case studies, including examples in target-mediated PK/PD, CAR-T-cell therapy, viral dynamics, and transmission models in epidemiology. Through such examples, we demonstrate that SAAM II provides a suitable interface for rapid model selection and prototyping. By enabling the fast creation of detailed mathematical models, SAAM II addresses a unique requirement within the mathematical modeling community.
Collapse
Affiliation(s)
- Simone Perazzolo
- Nanomath LLC, Spokane, Washington, USA
- University of Washington, Seattle, Washington, USA
| |
Collapse
|
2
|
Cremers S, Tucker GT, Aronson JK, Ritter JM, Cohen AF. The British Journal of Clinical Pharmacology: The first 50 years. Br J Clin Pharmacol 2024; 90:4-11. [PMID: 38153173 DOI: 10.1111/bcp.15952] [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: 10/13/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 12/29/2023] Open
Abstract
The British Journal of Clinical Pharmacology celebrates its 50th anniversary of publication in 2023. Here four previous Editors-in-Chief and the current Editor reflect on the Journal's history and the changes that have occurred during that time.
Collapse
Affiliation(s)
- Serge Cremers
- Departments of Pathology & Cell Biology and Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Geoffrey T Tucker
- School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Jeffrey K Aronson
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Radcliffe Observatory Quarter, Oxford, UK
| | - James M Ritter
- Department of Clinical Pharmacology, Clinical Research Facility, St Thomas' Hospital, London, UK
| | - Adam F Cohen
- Leiden University Medical Centre and Centre for Human Drug Research, Leiden, The Netherlands
| |
Collapse
|
3
|
Chenane HR, Lingas G, Menidjel R, Laouenan C, Tubiana S, Descamps D, Le Hingrat Q, Abel L, Guedj J, Malhotra S, Kumar-Singh S, Visseaux B, Ghosn J, Charpentier C, Lebourgeois S. High sera levels of SARS-CoV-2 N antigen are associated with death in hospitalized COVID-19 patients. J Med Virol 2023; 95:e29247. [PMID: 38009713 DOI: 10.1002/jmv.29247] [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: 04/19/2023] [Revised: 09/05/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
The presence of free severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid-antigen in sera (N-antigenemia) has been shown in COVID-19 patients. However, the link between the quantitative levels of N-antigenemia and COVID-19 disease severity is not entirely understood. To assess the dynamics and clinical association of N-antigen sera levels with disease severity in COVID-19 patients, we analyzed data from patients included in the French COVID cohort, with at least one sera sample between January and September 2020. We assessed N-antigenemia levels and anti-N IgG titers, and patient outcomes was classified in two groups, survival or death. In samples collected within 8 days since symptom onset, we observed that deceased patients had a higher positivity rate (93% vs. 81%; p < 0.001) and higher median levels of predicted N-antigenemia (2500 vs. 1200 pg/mL; p < 0.001) than surviving patients. Predicted time to N-antigen clearance in sera was prolonged in deceased patients compared to survivors (23.3 vs 19.3 days; p < 0.0001). In a subset of patients with both sera and nasopharyngeal (NP) swabs, predicted time to N-antigen clearance in sera was prolonged in deceased patients (p < 0.001), whereas NP viral load clearance did not differ between the groups (p = 0.07). Our results demonstrate a strong relationship between N-antigenemia levels and COVID-19 severity on a prospective cohort.
Collapse
Affiliation(s)
| | | | - Reyene Menidjel
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
| | - Cédric Laouenan
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Centre d'Investigations cliniques-Epidémiologie Clinique 1425, Hôpital Bichat, Paris, France
| | - Sarah Tubiana
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Centre d'Investigations cliniques-Epidémiologie Clinique 1425, Hôpital Bichat, Paris, France
| | - Diane Descamps
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Virologie, Hôpital Bichat, Paris, France
| | - Quentin Le Hingrat
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Virologie, Hôpital Bichat, Paris, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jérémie Guedj
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
| | - Surbhi Malhotra
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Molecular Pathology group, Cell Biology & Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Benoit Visseaux
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
| | - Jade Ghosn
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Maladies Infectieuses et Tropicales, Hôpital Bichat, Paris, France
| | - Charlotte Charpentier
- Inserm, IAME, UMR 1137, Université Paris Cité, Paris, France
- Service de Virologie, Hôpital Bichat, Paris, France
| | | |
Collapse
|
4
|
Beahm DR, Deng Y, DeAngelo TM, Sarpeshkar R. Drug Cocktail Formulation via Circuit Design. IEEE TRANSACTIONS ON MOLECULAR, BIOLOGICAL, AND MULTI-SCALE COMMUNICATIONS 2023; 9:28-48. [PMID: 37397625 PMCID: PMC10312325 DOI: 10.1109/tmbmc.2023.3246928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Electronic circuits intuitively visualize and quantitatively simulate biological systems with nonlinear differential equations that exhibit complicated dynamics. Drug cocktail therapies are a powerful tool against diseases that exhibit such dynamics. We show that just six key states, which are represented in a feedback circuit, enable drug-cocktail formulation: 1) healthy cell number; 2) infected cell number; 3) extracellular pathogen number; 4) intracellular pathogenic molecule number; 5) innate immune system strength; and 6) adaptive immune system strength. To enable drug cocktail formulation, the model represents the effects of the drugs in the circuit. For example, a nonlinear feedback circuit model fits measured clinical data, represents cytokine storm and adaptive autoimmune behavior, and accounts for age, sex, and variant effects for SARS-CoV-2 with few free parameters. The latter circuit model provided three quantitative insights on the optimal timing and dosage of drug components in a cocktail: 1) antipathogenic drugs should be given early in the infection, but immunosuppressant timing involves a tradeoff between controlling pathogen load and mitigating inflammation; 2) both within and across-class combinations of drugs have synergistic effects; 3) if they are administered sufficiently early in the infection, anti-pathogenic drugs are more effective at mitigating autoimmune behavior than immunosuppressant drugs.
Collapse
Affiliation(s)
| | - Yijie Deng
- Thayer School or Engineering, Dartmouth College, Hanover, NH 03755 USA
| | - Thomas M DeAngelo
- Thayer School or Engineering, Dartmouth College, Hanover, NH 03755 USA
| | - Rahul Sarpeshkar
- Departments of Engineering, Physics, Microbiology & Immunobiology, and Molecular & Systems Biology, Dartmouth College, Hanover, NH 03755 USA
| |
Collapse
|
5
|
Agyeman AA, You T, Chan PLS, Lonsdale DO, Hadjichrysanthou C, Mahungu T, Wey EQ, Lowe DM, Lipman MCI, Breuer J, Kloprogge F, Standing JF. Comparative assessment of viral dynamic models for SARS-CoV-2 for pharmacodynamic assessment in early treatment trials. Br J Clin Pharmacol 2022; 88:5428-5433. [PMID: 36040430 PMCID: PMC9538685 DOI: 10.1111/bcp.15518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 08/03/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022] Open
Abstract
Pharmacometric analyses of time series viral load data may detect drug effects with greater power than approaches using single time points. Because SARS-CoV-2 viral load rapidly rises and then falls, viral dynamic models have been used. We compared different modelling approaches when analysing Phase II-type viral dynamic data. Using two SARS-CoV-2 datasets of viral load starting within 7 days of symptoms, we fitted the slope-intercept exponential decay (SI), reduced target cell limited (rTCL), target cell limited (TCL) and TCL with eclipse phase (TCLE) models using nlmixr. Model performance was assessed via Bayesian information criterion (BIC), visual predictive checks (VPCs), goodness-of-fit plots, and parameter precision. The most complex (TCLE) model had the highest BIC for both datasets. The estimated viral decline rate was similar for all models except the TCL model for dataset A with a higher rate (median [range] day-1 : dataset A; 0.63 [0.56-1.84]; dataset B: 0.81 [0.74-0.85]). Our findings suggest simple models should be considered during pharmacodynamic model development.
Collapse
Affiliation(s)
- Akosua A Agyeman
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Tao You
- Beyond Consulting Ltd., Cheshire, UK.,Medical Research Council, UK
| | | | - Dagan O Lonsdale
- Department of Clinical Pharmacology, St George's University of London, London, UK.,Department of Intensive Care, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Christoforos Hadjichrysanthou
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Tabitha Mahungu
- Department of Infectious Diseases, Royal Free Hospital London NHS Foundation Trust, London, UK
| | - Emmanuel Q Wey
- Department of Infectious Diseases, Royal Free Hospital London NHS Foundation Trust, London, UK.,Centre for Clinical Microbiology, Division of Infection and Immunity University College London, London, UK
| | - David M Lowe
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, UK.,Institute of Immunity and Transplantation, University College London, Royal Free Campus, London, UK
| | - Marc C I Lipman
- Department of Respiratory Medicine, Royal Free London NHS Foundation Trust, London, UK.,UCL Respiratory, University College London, Royal Free Campus, London, UK
| | - Judy Breuer
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.,Department of Microbiology, Great Ormond Street Hospital for Children, London, UK
| | - Frank Kloprogge
- Institute for Global Health, University College London, London, UK
| | - Joseph F Standing
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.,Department of Pharmacy, Great Ormond Street Hospital for Children, London, UK
| |
Collapse
|
6
|
Abstract
OBJECTIVE A deep understanding of the relationship between a scarce drug's dose and clinical response is necessary to appropriately distribute a supply-constrained drug along these lines. SUMMARY OF KEY DATA The vast majority of drug development and repurposing during the COVID-19 pandemic - an event that has made clear the ever-present scarcity in healthcare systems -has been ignorant of scarcity and dose optimisation's ability to help address it. CONCLUSIONS Future pandemic clinical trials systems should obtain dose optimisation data, as these appear necessary to enable appropriate scarce resource allocation according to societal values.
Collapse
Affiliation(s)
- Garth Strohbehn
- Center for Clinical Management Research, Veterans Affairs Ann Arbor Health Care, Ann Arbor, Michigan, USA
| | - Govind Persad
- Sturm College of Law, University of Denver, Denver, Colorado, USA
| | - William F Parker
- Maclean Center for Clinical Medical Ethics, University of Chicago, Chicago, Illinois, USA
| | - Srinivas Murthy
- Paediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Arsène S, Couty C, Faddeenkov I, Go N, Granjeon-Noriot S, Šmít D, Kahoul R, Illigens B, Boissel JP, Chevalier A, Lehr L, Pasquali C, Kulesza A. Modeling the disruption of respiratory disease clinical trials by non-pharmaceutical COVID-19 interventions. Nat Commun 2022; 13:1980. [PMID: 35418135 PMCID: PMC9008035 DOI: 10.1038/s41467-022-29534-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/21/2022] [Indexed: 02/07/2023] Open
Abstract
Respiratory disease trials are profoundly affected by non-pharmaceutical interventions (NPIs) against COVID-19 because they perturb existing regular patterns of all seasonal viral epidemics. To address trial design with such uncertainty, we developed an epidemiological model of respiratory tract infection (RTI) coupled to a mechanistic description of viral RTI episodes. We explored the impact of reduced viral transmission (mimicking NPIs) using a virtual population and in silico trials for the bacterial lysate OM-85 as prophylaxis for RTI. Ratio-based efficacy metrics are only impacted under strict lockdown whereas absolute benefit already is with intermediate NPIs (eg. mask-wearing). Consequently, despite NPI, trials may meet their relative efficacy endpoints (provided recruitment hurdles can be overcome) but are difficult to assess with respect to clinical relevance. These results advocate to report a variety of metrics for benefit assessment, to use adaptive trial design and adapted statistical analyses. They also question eligibility criteria misaligned with the actual disease burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ben Illigens
- Novadiscovery SA, Lyon, France
- Dresden International University, Dresden, Germany
| | | | | | | | | | | |
Collapse
|
8
|
Karr J, Malik-Sheriff RS, Osborne J, Gonzalez-Parra G, Forgoston E, Bowness R, Liu Y, Thompson R, Garira W, Barhak J, Rice J, Torres M, Dobrovolny HM, Tang T, Waites W, Glazier JA, Faeder JR, Kulesza A. Model Integration in Computational Biology: The Role of Reproducibility, Credibility and Utility. FRONTIERS IN SYSTEMS BIOLOGY 2022; 2:822606. [PMID: 36909847 PMCID: PMC10002468 DOI: 10.3389/fsysb.2022.822606] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During the COVID-19 pandemic, mathematical modeling of disease transmission has become a cornerstone of key state decisions. To advance the state-of-the-art host viral modeling to handle future pandemics, many scientists working on related issues assembled to discuss the topics. These discussions exposed the reproducibility crisis that leads to inability to reuse and integrate models. This document summarizes these discussions, presents difficulties, and mentions existing efforts towards future solutions that will allow future model utility and integration. We argue that without addressing these challenges, scientists will have diminished ability to build, disseminate, and implement high-impact multi-scale modeling that is needed to understand the health crises we face.
Collapse
Affiliation(s)
- Jonathan Karr
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rahuman S. Malik-Sheriff
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, United Kingdom
| | - James Osborne
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, Australia
| | | | - Eric Forgoston
- Department of Applied Mathematics and Statistics, Montclair State University, Montclair, NJ, United States
| | - Ruth Bowness
- Department of Mathematical Sciences, University of Bath, Bath, United Kingdom
| | - Yaling Liu
- Department of Mechanical Engineering and Mechanics, Department of Bioengineering, Lehigh University, Bethlehem, PA, United States
| | - Robin Thompson
- Mathematics Institute and the Zeeman Institute for Systems Biology and Infectious Disease Epidemiology Research, University of Warwick, Coventry, United Kingdom
| | - Winston Garira
- Department of Mathematics and Applied Mathematics, Modelling Health and Environmental Linkages Research Group, University of Venda, Limpopo, South Africa
| | - Jacob Barhak
- Jacob Barhak Analytics, Austin, TX, United States
| | - John Rice
- Independent Retired Working Group Volunteer, Virginia Beach, VA, United States
| | - Marcella Torres
- Department of Mathematics and Computer Science, University of Richmond, Richmond, VA, United States
| | - Hana M. Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| | - Tingting Tang
- Department of Mathematics and Statistics in San Diego State University (SDSU) and SDSU Imperial Valley, Calexico, CA, United States
| | - William Waites
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Computer and Information Sciences, University of Strathclyde, Glasgow, Scotland
| | - James A. Glazier
- Biocomplexity Institute, Indiana University, Bloomington, IN, United States
| | - James R. Faeder
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | | |
Collapse
|
9
|
Courcelles E, Boissel JP, Massol J, Klingmann I, Kahoul R, Hommel M, Pham E, Kulesza A. Solving the Evidence Interpretability Crisis in Health Technology Assessment: A Role for Mechanistic Models? FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:810315. [PMID: 35281671 PMCID: PMC8907708 DOI: 10.3389/fmedt.2022.810315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/17/2022] [Indexed: 01/11/2023] Open
Abstract
Health technology assessment (HTA) aims to be a systematic, transparent, unbiased synthesis of clinical efficacy, safety, and value of medical products (MPs) to help policymakers, payers, clinicians, and industry to make informed decisions. The evidence available for HTA has gaps-impeding timely prediction of the individual long-term effect in real clinical practice. Also, appraisal of an MP needs cross-stakeholder communication and engagement. Both aspects may benefit from extended use of modeling and simulation. Modeling is used in HTA for data-synthesis and health-economic projections. In parallel, regulatory consideration of model informed drug development (MIDD) has brought attention to mechanistic modeling techniques that could in fact be relevant for HTA. The ability to extrapolate and generate personalized predictions renders the mechanistic MIDD approaches suitable to support translation between clinical trial data into real-world evidence. In this perspective, we therefore discuss concrete examples of how mechanistic models could address HTA-related questions. We shed light on different stakeholder's contributions and needs in the appraisal phase and suggest how mechanistic modeling strategies and reporting can contribute to this effort. There are still barriers dissecting the HTA space and the clinical development space with regard to modeling: lack of an adapted model validation framework for decision-making process, inconsistent and unclear support by stakeholders, limited generalizable use cases, and absence of appropriate incentives. To address this challenge, we suggest to intensify the collaboration between competent authorities, drug developers and modelers with the aim to implement mechanistic models central in the evidence generation, synthesis, and appraisal of HTA so that the totality of mechanistic and clinical evidence can be leveraged by all relevant stakeholders.
Collapse
Affiliation(s)
| | | | - Jacques Massol
- Phisquare Institute, Transplantation Foundation, Paris, France
| | | | | | | | | | | |
Collapse
|
10
|
Alexander P, Dobrovolny HM. Treatment of Respiratory Viral Coinfections. EPIDEMIOLOGIA 2022; 3:81-96. [PMID: 36417269 PMCID: PMC9620919 DOI: 10.3390/epidemiologia3010008] [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: 11/29/2021] [Revised: 01/18/2022] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
With the advent of rapid multiplex PCR, physicians have been able to test for multiple viral pathogens when a patient presents with influenza-like illness. This has led to the discovery that many respiratory infections are caused by more than one virus. Antiviral treatment of viral coinfections can be complex because treatment of one virus will affect the time course of the other virus. Since effective antivirals are only available for some respiratory viruses, careful consideration needs to be given on the effect treating one virus will have on the dynamics of the other virus, which might not have available antiviral treatment. In this study, we use mathematical models of viral coinfections to assess the effect of antiviral treatment on coinfections. We examine the effect of the mechanism of action, relative growth rates of the viruses, and the assumptions underlying the interaction of the viruses. We find that high antiviral efficacy is needed to suppress both infections. If high doses of both antivirals are not achieved, then we run the risk of lengthening the duration of coinfection or even of allowing a suppressed virus to replicate to higher viral titers.
Collapse
Affiliation(s)
| | - Hana M. Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX 76129, USA;
| |
Collapse
|
11
|
Dodds MG, Krishna R, Goncalves A, Rayner CR. Model-informed drug repurposing: Viral kinetic modelling to prioritize rational drug combinations for COVID-19. Br J Clin Pharmacol 2021; 87:3439-3450. [PMID: 32693436 PMCID: PMC8451752 DOI: 10.1111/bcp.14486] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
AIM We hypothesized that viral kinetic modelling could be helpful to prioritize rational drug combinations for COVID-19. The aim of this research was to use a viral cell cycle model of SARS-CoV-2 to explore the potential impact drugs, or combinations of drugs, that act at different stages in the viral life cycle might have on various metrics of infection outcome relevant in the early stages of COVID-19 disease. METHODS Using a target-cell limited model structure that has been used to characterize viral load dynamics from COVID-19 patients, we performed simulations to inform on the combinations of therapeutics targeting specific rate constants. The endpoints and metrics included viral load area under the curve (AUC), duration of viral shedding and epithelial cells infected. Based on the known kinetics of the SARS-CoV-2 life cycle, we rank ordered potential targeted approaches involving repurposed, low-potency agents. RESULTS Our simulations suggest that targeting multiple points central to viral replication within infected host cells or release from those cells is a viable strategy for reducing both viral load and host cell infection. In addition, we observed that the time-window opportunity for a therapeutic intervention to effect duration of viral shedding exceeds the effect on sparing epithelial cells from infection or impact on viral load AUC. Furthermore, the impact on reduction on duration of shedding may extend further in patients who exhibit a prolonged shedder phenotype. CONCLUSIONS Our work highlights the use of model-informed drug repurposing approaches to better rationalize effective treatments for COVID-19.
Collapse
Affiliation(s)
| | | | | | - Craig R. Rayner
- Certara, USA Inc.PrincetonNJ
- Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneAustralia
| |
Collapse
|
12
|
Wright DFB, Kirkpatrick CMJ. Science fiction has become reality: Best practice for future viral pandemics. Br J Clin Pharmacol 2021; 87:3385-3387. [PMID: 34296460 PMCID: PMC8444651 DOI: 10.1111/bcp.14997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/02/2022] Open
|
13
|
Lim XY, Teh BP, Tan TYC. Medicinal Plants in COVID-19: Potential and Limitations. Front Pharmacol 2021; 12:611408. [PMID: 33841143 PMCID: PMC8025226 DOI: 10.3389/fphar.2021.611408] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Currently, the search to identify treatments and vaccines for novel coronavirus disease (COVID-19) are ongoing. Desperation within the community, especially among the middle-and low-income groups acutely affected by the economic impact of forced lockdowns, has driven increased interest in exploring alternative choices of medicinal plant-based therapeutics. This is evident with the rise in unsubstantiated efficacy claims of these interventions circulating on social media. Based on enquiries received, our team of researchers was given the chance to produce evidence summaries evaluating the potential of complementary interventions in COVID-19 management. Here, we present and discuss the findings of four selected medicinal plants (Nigella sativa, Vernonia amygdalina, Azadirachta indica, Eurycoma longifolia), with reported antiviral, anti-inflammatory, and immunomodulatory effects that might be interesting for further investigation. Our findings showed that only A. indica reported positive antiviral evidence specific to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) based on preliminary in silico data while all four medicinal plants demonstrated differential anti-inflammatory or immunomodulatory effects. The definitive roles of these medicinal plants in cytokine storms and post-infection complications remains to be further investigated. Quality control and standardisation of medicinal plant-based products also needs to be emphasized. However, given the unprecedented challenges faced, ethnopharmacological research should be given a fair amount of consideration for contribution in this pandemic.
Collapse
Affiliation(s)
- Xin Yi Lim
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Malaysia
| | - Bee Ping Teh
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Malaysia
| | - Terence Yew Chin Tan
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam, Malaysia
| |
Collapse
|
14
|
Rayner CR, Smith PF, Andes D, Andrews K, Derendorf H, Friberg LE, Hanna D, Lepak A, Mills E, Polasek TM, Roberts JA, Schuck V, Shelton MJ, Wesche D, Rowland‐Yeo K. Model-Informed Drug Development for Anti-Infectives: State of the Art and Future. Clin Pharmacol Ther 2021; 109:867-891. [PMID: 33555032 PMCID: PMC8014105 DOI: 10.1002/cpt.2198] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
Model-informed drug development (MIDD) has a long and rich history in infectious diseases. This review describes foundational principles of translational anti-infective pharmacology, including choice of appropriate measures of exposure and pharmacodynamic (PD) measures, patient subpopulations, and drug-drug interactions. Examples are presented for state-of-the-art, empiric, mechanistic, interdisciplinary, and real-world evidence MIDD applications in the development of antibacterials (review of minimum inhibitory concentration-based models, mechanism-based pharmacokinetic/PD (PK/PD) models, PK/PD models of resistance, and immune response), antifungals, antivirals, drugs for the treatment of global health infectious diseases, and medical countermeasures. The degree of adoption of MIDD practices across the infectious diseases field is also summarized. The future application of MIDD in infectious diseases will progress along two planes; "depth" and "breadth" of MIDD methods. "MIDD depth" refers to deeper incorporation of the specific pathogen biology and intrinsic and acquired-resistance mechanisms; host factors, such as immunologic response and infection site, to enable deeper interrogation of pharmacological impact on pathogen clearance; clinical outcome and emergence of resistance from a pathogen; and patient and population perspective. In particular, improved early assessment of the emergence of resistance potential will become a greater focus in MIDD, as this is poorly mitigated by current development approaches. "MIDD breadth" refers to greater adoption of model-centered approaches to anti-infective development. Specifically, this means how various MIDD approaches and translational tools can be integrated or connected in a systematic way that supports decision making by key stakeholders (sponsors, regulators, and payers) across the entire development pathway.
Collapse
Affiliation(s)
- Craig R. Rayner
- CertaraPrincetonNew JerseyUSA
- Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | | | - David Andes
- University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kayla Andrews
- Bill & Melinda Gates Medical Research InstituteCambridgeMassachusettsUSA
| | | | | | - Debra Hanna
- Bill & Melinda Gates FoundationSeattleWashingtonUSA
| | - Alex Lepak
- University of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | - Thomas M. Polasek
- CertaraPrincetonNew JerseyUSA
- Centre for Medicines Use and SafetyMonash UniversityMelbourneVictoriaAustralia
- Department of Clinical PharmacologyRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
| | - Jason A. Roberts
- Faculty of MedicineUniversity of Queensland Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Departments of Pharmacy and Intensive Care MedicineRoyal Brisbane and Women’s HospitalBrisbaneQueenslandAustralia
- Division of Anaesthesiology Critical Care Emergency and Pain MedicineNîmes University HospitalUniversity of MontpellierMontpellierFrance
| | | | | | | | | |
Collapse
|
15
|
Perazzolo S, Zhu L, Lin W, Nguyen A, Ho RJY. Systems and Clinical Pharmacology of COVID-19 Therapeutic Candidates: A Clinical and Translational Medicine Perspective. J Pharm Sci 2021; 110:1002-1017. [PMID: 33248057 PMCID: PMC7689305 DOI: 10.1016/j.xphs.2020.11.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
Over 50 million people have been infected with the SARS-CoV-2 virus, while around 1 million have died due to COVID-19 disease progression. COVID-19 presents flu-like symptoms that can escalate, in about 7-10 days from onset, into a cytokine storm causing respiratory failure and death. Although social distancing reduces transmissibility, COVID-19 vaccines and therapeutics are essential to regain socioeconomic normalcy. Even if effective and safe vaccines are found, pharmacological interventions are still needed to limit disease severity and mortality. Integrating current knowledge and drug candidates (approved drugs for repositioning among >35 candidates) undergoing clinical studies (>3000 registered in ClinicalTrials.gov), we employed Systems Pharmacology approaches to project how antivirals and immunoregulatory agents could be optimally evaluated for use. Antivirals are likely to be effective only at the early stage of infection, soon after exposure and before hospitalization, while immunomodulatory agents should be effective in the later-stage cytokine storm. As current antiviral candidates are administered in hospitals over 5-7 days, a long-acting combination that targets multiple SARS-CoV-2 lifecycle steps may provide a long-lasting, single-dose treatment in outpatient settings. Long-acting therapeutics may still be needed even when vaccines become available as vaccines are likely to be approved based on a 50% efficacy target.
Collapse
Affiliation(s)
- Simone Perazzolo
- Department of Pharmaceutics, School of Pharmacy, Seattle, WA 98195, USA; Targeted and Long-Acting Drug Combination Anti-Retroviral Therapeutic (TLC-ART) Program, University of Washington, Seattle, WA 98195, USA; NanoMath, Seattle, WA 98115, USA.
| | - Linxi Zhu
- Department of Pharmaceutics, School of Pharmacy, Seattle, WA 98195, USA; Targeted and Long-Acting Drug Combination Anti-Retroviral Therapeutic (TLC-ART) Program, University of Washington, Seattle, WA 98195, USA
| | - Weixian Lin
- Department of Pharmaceutics, School of Pharmacy, Seattle, WA 98195, USA; First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Alexander Nguyen
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, USA
| | - Rodney J Y Ho
- Department of Pharmaceutics, School of Pharmacy, Seattle, WA 98195, USA; Targeted and Long-Acting Drug Combination Anti-Retroviral Therapeutic (TLC-ART) Program, University of Washington, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
16
|
Dodds M, Xiong Y, Mouksassi S, Kirkpatrick CM, Hui K, Doyle E, Patel K, Cox E, Wesche D, Brown F, Rayner CR. Model-informed drug repurposing: A pharmacometric approach to novel pathogen preparedness, response and retrospection. Br J Clin Pharmacol 2021; 87:3388-3397. [PMID: 33534138 PMCID: PMC8013376 DOI: 10.1111/bcp.14760] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
During a pandemic caused by a novel pathogen (NP), drug repurposing offers the potential of a rapid treatment response via a repurposed drug (RD) while more targeted treatments are developed. Five steps of model‐informed drug repurposing (MIDR) are discussed: (i) utilize RD product label and in vitro NP data to determine initial proof of potential, (ii) optimize potential posology using clinical pharmacokinetics (PK) considering both efficacy and safety, (iii) link events in the viral life cycle to RD PK, (iv) link RD PK to clinical and virologic outcomes, and optimize clinical trial design, and (v) assess RD treatment effects from trials using model‐based meta‐analysis. Activities which fall under these five steps are categorized into three stages: what can be accomplished prior to an NP emergence (preparatory stage), during the NP pandemic (responsive stage) and once the crisis has subsided (retrospective stage). MIDR allows for extraction of a greater amount of information from emerging data and integration of disparate data into actionable insight.
Collapse
Affiliation(s)
| | | | | | - Carl M Kirkpatrick
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Katrina Hui
- Certara, Princeton, NJ, USA.,Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | | | - Kashyap Patel
- Certara, Princeton, NJ, USA.,Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | | | | | | | - Craig R Rayner
- Certara, Princeton, NJ, USA.,Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
17
|
Forrest JI, Rayner CR, Park JJH, Mills EJ. Early Treatment of COVID-19 Disease: A Missed Opportunity. Infect Dis Ther 2020; 9:715-720. [PMID: 33051827 PMCID: PMC7553378 DOI: 10.1007/s40121-020-00349-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Antivirals have demonstrated efficacy in treating other infectious diseases in early stages of disease, reducing morbidity, mortality, and the likelihood of onward transmission. At the time of writing, more than 1900 clinical trials are registered globally to assess the efficacy and safety of candidate therapeutics for COVID-19. The majority of these trials are designed to evaluate the comparative efficacy and safety of candidate therapeutics for the treatment of COVID-19 to prevent death among populations of hospitalized patients with advanced disease. Yet, emerging epidemiological evidence now indicates that the majority of those infected with the SARS-CoV-2, while still infectious, experience minimal or mild disease symptomology. Like HIV and hepatitis C that pioneered treatment as prevention, there is a missed opportunity for trials of early pharmaceutical intervention for COVID-19 disease evaluating not only reductions in morbidity and mortality but also transmissibility. We discuss this clinical research gap within an historical context of viral treatment as prevention for HIV and hepatitis C, and comment on the challenges and opportunities for clinical research of candidate therapeutics for early COVID-19 disease.
Collapse
Affiliation(s)
- Jamie I Forrest
- Cytel Canada Health Inc., Vancouver, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Craig R Rayner
- , Certara, NJ, USA
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Jay J H Park
- Cytel Canada Health Inc., Vancouver, BC, Canada
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edward J Mills
- Cytel Canada Health Inc., Vancouver, BC, Canada.
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
18
|
Rannard SP, McDonald TO, Owen A. Chasing COVID-19 chemotherapeutics without putting the cart before the horse. Br J Clin Pharmacol 2020; 89:421-423. [PMID: 33217038 PMCID: PMC7753793 DOI: 10.1111/bcp.14575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 01/02/2023] Open
Affiliation(s)
- Steven P. Rannard
- Department of ChemistryUniversity of LiverpoolLiverpoolUK,Centre of Excellence in Long‐acting Therapeutics (CELT)University of LiverpoolLiverpoolUK
| | - Tom O. McDonald
- Department of ChemistryUniversity of LiverpoolLiverpoolUK,Centre of Excellence in Long‐acting Therapeutics (CELT)University of LiverpoolLiverpoolUK
| | - Andrew Owen
- Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK,Centre of Excellence in Long‐acting Therapeutics (CELT)University of LiverpoolLiverpoolUK
| |
Collapse
|
19
|
Rayner CR, Dron L, Park JJH, Decloedt EH, Cotton MF, Niranjan V, Smith PF, Dodds MG, Brown F, Reis G, Wesche D, Mills EJ. Accelerating Clinical Evaluation of Repurposed Combination Therapies for COVID-19. Am J Trop Med Hyg 2020; 103:1364-1366. [PMID: 32828137 PMCID: PMC7543863 DOI: 10.4269/ajtmh.20-0995] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022] Open
Abstract
As the global COVID-19 pandemic continues, unabated and clinical trials demonstrate limited effective pharmaceutical interventions, there is a pressing need to accelerate treatment evaluations. Among options for accelerated development is the evaluation of drug combinations in the absence of prior monotherapy data. This approach is appealing for a number of reasons. First, combining two or more drugs with related or complementary therapeutic effects permits a multipronged approach addressing the variable pathways of the disease. Second, if an individual component of a combination offers a therapeutic effect, then in the absence of antagonism, a trial of combination therapy should still detect individual efficacy. Third, this strategy is time saving. Rather than taking a stepwise approach to evaluating monotherapies, this strategy begins with testing all relevant therapeutic options. Finally, given the severity of the current pandemic and the absence of treatment options, the likelihood of detecting a treatment effect with combination therapy maintains scientific enthusiasm for evaluating repurposed treatments. Antiviral combination selection can be facilitated by insights regarding SARS-CoV-2 pathophysiology and cell cycle dynamics, supported by infectious disease and clinical pharmacology expert advice. We describe a clinical evaluation strategy using adaptive combination platform trials to rapidly test combination therapies to treat COVID-19.
Collapse
Affiliation(s)
- Craig R. Rayner
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
- Certara, Princeton, New Jersey
| | - Louis Dron
- Cytel Inc., Vancouver, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Jay J. H. Park
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
- Department of Medicine, Experimental Medicine, University of British Columbia, Vancouver, Canada
| | - Eric H. Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Mark F. Cotton
- Family Clinical Research Unit, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | | | | | | | - Gilmar Reis
- PUC Minas Medical School of Medicine at Contagem, Belo Horizonte, Brazil
| | | | - Edward J. Mills
- Cytel Inc., Vancouver, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| |
Collapse
|
20
|
Clinical trials of disease stages in COVID 19: complicated and often misinterpreted. LANCET GLOBAL HEALTH 2020; 8:e1249-e1250. [PMID: 32828171 PMCID: PMC7440858 DOI: 10.1016/s2214-109x(20)30365-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 12/30/2022]
|