51
|
Morris NM, Blee JA, Hauert S. Developing a computational pharmacokinetic model of systemic snakebite envenomation and antivenom treatment. Toxicon 2022; 215:77-90. [PMID: 35716719 DOI: 10.1016/j.toxicon.2022.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/20/2022] [Accepted: 06/09/2022] [Indexed: 11/19/2022]
Abstract
Snakebite envenomation is responsible for over 100,000 deaths and 400,000 cases of disability annually, most of which are preventable through access to safe and effective antivenoms. Snake venom toxins span a wide molecular weight range, influencing their absorption, distribution, and elimination within the body. In recent years, a range of scaffolds have been applied to antivenom development. These scaffolds similarly span a wide molecular weight range and subsequently display diverse pharmacokinetic behaviours. Computational simulations represent a powerful tool to explore the interplay between these varied antivenom scaffolds and venoms, to assess whether a pharmacokinetically optimal antivenom exists. The purpose of this study was to establish a computational model of systemic snakebite envenomation and treatment, for the quantitative assessment and comparison of conventional and next-generation antivenoms. A two-compartment mathematical model of envenomation and treatment was defined and the system was parameterised using existing data from rabbits. Elimination and biodistribution parameters were regressed against molecular weight to predict the dynamics of IgG, F(ab')2, Fab, scFv, and nanobody antivenoms, spanning a size range of 15-150 kDa. As a case study, intramuscular envenomation by Naja sumatrana (equatorial spitting cobra) and its treatment using Fab, F(ab')2, and IgG antivenoms was simulated. Variable venom dose tests were applied to visualise effective antivenom dose levels. Comparisons to existing antivenoms and experimental rescue studies highlight the large dose reductions that could result from recombinant antivenom use. This study represents the first comparative in silico model of snakebite envenomation and treatment.
Collapse
Affiliation(s)
- Natalie M Morris
- Department of Engineering Mathematics, Ada Lovelace Building, University of Bristol, University Walk, Bristol, BS8 1TW, UK.
| | - Johanna A Blee
- Department of Engineering Mathematics, Ada Lovelace Building, University of Bristol, University Walk, Bristol, BS8 1TW, UK.
| | - Sabine Hauert
- Department of Engineering Mathematics, Ada Lovelace Building, University of Bristol, University Walk, Bristol, BS8 1TW, UK.
| |
Collapse
|
52
|
Marcantonio DH, Matteson A, Presler M, Burke JM, Hagen DR, Hua F, Apgar JF. Early Feasibility Assessment: A Method for Accurately Predicting Biotherapeutic Dosing to Inform Early Drug Discovery Decisions. Front Pharmacol 2022; 13:864768. [PMID: 35754500 PMCID: PMC9214263 DOI: 10.3389/fphar.2022.864768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
The application of model-informed drug discovery and development (MID3) approaches in the early stages of drug discovery can help determine feasibility of drugging a target, prioritize between targets, or define optimal drug properties for a target product profile (TPP). However, applying MID3 in early discovery can be challenging due to the lack of pharmacokinetic (PK) and pharmacodynamic (PD) data at this stage. Early Feasibility Assessment (EFA) is the application of mechanistic PKPD models, built from first principles, and parameterized by data that is readily available early in drug discovery to make effective dose predictions. This manuscript demonstrates the ability of EFA to make accurate predictions of clinical effective doses for nine approved biotherapeutics and outlines the potential of extending this approach to novel therapeutics to impact early drug discovery decisions.
Collapse
|
53
|
Morales-Kastresana A, Siegemund M, Haak S, Peper-Gabriel J, Neiens V, Rothe C. Anticalin®-based therapeutics: Expanding new frontiers in drug development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:89-106. [PMID: 35777866 DOI: 10.1016/bs.ircmb.2022.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Anticalin proteins are a novel class of clinical-stage biopharmaceuticals with high potential in various disease areas. Anticalin proteins, derived from extracellular human lipocalins are single-chain proteins, with a highly stable structure that can be engineered to bind with high specificity and potency to targets of therapeutic relevance. The small size and stable structure support their development as inhalable biologics in the field of respiratory diseases as already demonstrated for PRS-060/AZD1402, an Anticalin protein currently undergoing clinical development for the treatment of asthma. Anticalin proteins provide formatting flexibility which allows fusion with the same or other Anticalin proteins, or with other biologics to generate multivalent, multiparatopic or multispecific fusion proteins. The fusion of Anticalin proteins to antibodies allows the generation of potent therapeutic proteins with new modes of action, such as antibody-Anticalin bispecific proteins with tumor-localized activity. Cinrebafusp alfa and PRS-344/S095012 antibody-Anticalin bispecific proteins were designed to reduce potential systemic toxicity by localizing the activity to the tumor, and are currently in clinical development in immuno-oncology. Furthermore, the ease in generating bi- and multispecifics as well as the small and stable structure prompted the investigation of Anticalin proteins for the CAR T space, opening additional potential treatment options based on Anticalin protein therapies.
Collapse
Affiliation(s)
| | | | - Stefan Haak
- Pieris Pharmaceuticals GmbH, Hallbergmoos, Germany
| | | | | | | |
Collapse
|
54
|
van der Straten K, van Gils MJ, de Taeye SW, de Bree GJ. Optimization of Anti-SARS-CoV-2 Neutralizing Antibody Therapies: Roadmap to Improve Clinical Effectiveness and Implementation. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:867982. [PMID: 35419561 PMCID: PMC8996231 DOI: 10.3389/fmedt.2022.867982] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
One of the major breakthroughs to combat the current Coronavirus Disease 2019 (COVID-19) pandemic has been the development of highly effective vaccines against the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Still, alternatives are needed for individuals who are at high risk of developing severe COVID-19 and are not protected by vaccination. Monoclonal antibodies against the spike protein of SARS-CoV-2 have been shown to be effective as prophylaxis and treatment against COVID-19. However, the emergence of variants of concern (VOCs) challenges the efficacy of antibody therapies. This review describes the neutralization resistance of the clinically-approved monoclonal antibody therapies against the Alpha (B.1.1.7), Beta (B.1.351), Gamma (P1), Delta (B.1.617.2), and the Omicron (B.1.1.529) variants. To guide the development of monoclonal antibody therapies and to anticipate on the continuous evolution of SARS-CoV-2, we highlight different strategies to broaden the antibody activity by targeting more conserved epitopes and/or simultaneously targeting multiple sites of vulnerability of the virus. This review further describes the contribution of antibody Fc effector functions to optimize the antibody efficacy. In addition, the main route of SARS-CoV-2 antibody administration is currently intravenously and dictates a monthly injection when used as prophylactic. Therefore, we discusses the concept of long-acting antibodies (LAABs) and non-intravenously routes of antibody administration in order to broaden the clinical applicability of antibody therapies.
Collapse
Affiliation(s)
- Karlijn van der Straten
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Internal Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Steven W. de Taeye
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Godelieve J. de Bree
- Department of Internal Medicine, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
55
|
Stüber JC, Rechberger KF, Miladinović SM, Pöschinger T, Zimmermann T, Villenave R, Eigenmann MJ, Kraft TE, Shah DK, Kettenberger H, Richter WF. Impact of charge patches on tumor disposition and biodistribution of therapeutic antibodies. AAPS OPEN 2022. [DOI: 10.1186/s41120-021-00048-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThis study explores the impact of antibody surface charge on tissue distribution into various tissues including tumor. Tumor-bearing mice were dosed intravenously with a mixture comprising three antibodies engineered to carry negative charge patches, a balanced charge distribution, or positive patches, respectively (cassette dosing). Tissue levels were analyzed with a specific LC-MS/MS method. In addition, the antibody mix was administered to non-tumor bearing mice. Muscle and skin interstitial fluid were obtained by centrifugation and analyzed by LC-MS/MS. An in vitro endothelium model was explored for its feasibility to mimic the observed distribution differences.A balanced charge distribution was optimal in terms of total tumor exposure, while in other tissues, negatively charged and balanced charged antibodies gave similar results. In contrast, positive charge patches generally resulted in increased serum clearance but markedly enhanced tumor and organ uptake, leading to higher tissue-to-serum ratios. The uptake and availability in the interstitial space were confirmed by specific assessment of antibody levels in the interstitial fluid of the muscle and skin, with similar charge impact as in total tissue. The in vitro model was able to differentiate the transport propensity of this series of antibody variants. In summary, our results show the differential effects of charge patches on an antibody surface on biodistribution and tumor uptake. These insights may help in the design of molecules with biodistribution properties tailored to their purpose, and an optimized safety profile.
Collapse
|
56
|
Wechalekar A, Antoni G, Al Azzam W, Bergström M, Biswas S, Chen C, Cheriyan J, Cleveland M, Cookson L, Galette P, Janiczek RL, Kwong RY, Lukas MA, Millns H, Richards D, Schneider I, Solomon SD, Sörensen J, Storey J, Thompson D, van Dongen G, Vugts DJ, Wall A, Wikström G, Falk RH. Pharmacodynamic evaluation and safety assessment of treatment with antibodies to serum amyloid P component in patients with cardiac amyloidosis: an open-label Phase 2 study and an adjunctive immuno-PET imaging study. BMC Cardiovasc Disord 2022; 22:49. [PMID: 35152886 PMCID: PMC8843022 DOI: 10.1186/s12872-021-02407-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/26/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND In a Phase I study treatment with the serum amyloid P component (SAP) depleter miridesap followed by monoclonal antibody to SAP (dezamizumab) showed removal of amyloid from liver, spleen and kidney in patients with systemic amyloidosis. We report results from a Phase 2 study and concurrent immuno-positron emission tomography (PET) study assessing efficacy, pharmacodynamics, pharmacokinetics, safety and cardiac uptake (of dezamizumab) following the same intervention in patients with cardiac amyloidosis. METHODS Both were uncontrolled open-label studies. After SAP depletion with miridesap, patients received ≤ 6 monthly doses of dezamizumab in the Phase 2 trial (n = 7), ≤ 2 doses of non-radiolabelled dezamizumab plus [89Zr]Zr-dezamizumab (total mass dose of 80 mg at session 1 and 500 mg at session 2) in the immuno-PET study (n = 2). Primary endpoints of the Phase 2 study were changed from baseline to follow-up (at 8 weeks) in left ventricular mass (LVM) by cardiac magnetic resonance imaging and safety. Primary endpoint of the immuno-PET study was [89Zr]Zr-dezamizumab cardiac uptake assessed via PET. RESULTS Dezamizumab produced no appreciable or consistent reduction in LVM nor improvement in cardiac function in the Phase 2 study. In the immuno-PET study, measurable cardiac uptake of [89Zr]Zr-dezamizumab, although seen in both patients, was moderate to low. Uptake was notably lower in the patient with higher LVM. Treatment-associated rash with cutaneous small-vessel vasculitis was observed in both studies. Abdominal large-vessel vasculitis after initial dezamizumab dosing (300 mg) occurred in the first patient with immunoglobulin light chain amyloidosis enrolled in the Phase 2 study. Symptom resolution was nearly complete within 24 h of intravenous methylprednisolone and dezamizumab discontinuation; abdominal computed tomography imaging showed vasculitis resolution by 8 weeks. CONCLUSIONS Unlike previous observations of visceral amyloid reduction, there was no appreciable evidence of amyloid removal in patients with cardiac amyloidosis in this Phase 2 trial, potentially related to limited cardiac uptake of dezamizumab as demonstrated in the immuno-PET study. The benefit-risk assessment for dezamizumab in cardiac amyloidosis was considered unfavourable after the incidence of large-vessel vasculitis and development for this indication was terminated. Trial registration NCT03044353 (2 February 2017) and NCT03417830 (25 January 2018).
Collapse
Affiliation(s)
| | - Gunnar Antoni
- Institutionen för Medicinska Vetenskaper, Uppsala University, Uppsala, Sweden
| | - Wasfi Al Azzam
- GlaxoSmithKline, Philadelphia, USA
- Takeda, Lexington, MA, USA
| | | | - Swethajit Biswas
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Chao Chen
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Joseph Cheriyan
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | | | | | | | | | | | - Duncan Richards
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Ian Schneider
- GlaxoSmithKline, Cambridge, UK
- Consolidated Consulting LTD, Cambridge, UK
| | | | - Jens Sörensen
- Institutionen för Medicinska Vetenskaper, Uppsala University, Uppsala, Sweden
| | | | | | | | | | - Anders Wall
- Institutionen för Medicinska Vetenskaper, Uppsala University, Uppsala, Sweden
| | - Gerhard Wikström
- Institutionen för Medicinska Vetenskaper, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
57
|
Abstract
Antibodies have been used to prevent or treat viral infections since the nineteenth century, but the full potential to use passive immunization for infectious diseases has yet to be realized. The advent of efficient methods for isolating broad and potently neutralizing human monoclonal antibodies is enabling us to develop antibodies with unprecedented activities. The discovery of IgG Fc region modifications that extend antibody half-life in humans to three months or more suggests that antibodies could become the principal tool with which we manage future viral epidemics. Antibodies for members of most virus families that cause severe disease in humans have been isolated, and many of them are in clinical development, an area that has accelerated during the effort to prevent or treat COVID-19 (coronavirus disease 2019). Broad and potently neutralizing antibodies are also important research reagents for identification of protective epitopes that can be engineered into active vaccines through structure-based reverse vaccinology. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- James E Crowe
- Vanderbilt Vaccine Center, Department of Pediatrics, and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| |
Collapse
|
58
|
Miyano T, Irvine AD, Tanaka RJ. Model-based meta-analysis to optimise S. aureus-targeted therapies for atopic dermatitis. JID INNOVATIONS 2022; 2:100110. [PMID: 35757782 PMCID: PMC9214323 DOI: 10.1016/j.xjidi.2022.100110] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/29/2022] Open
Abstract
Several clinical trials of Staphylococcus aureus (S. aureus)‒targeted therapies for atopic dermatitis (AD) have shown conflicting results about whether they improve AD severity scores. This study performs a model-based meta-analysis to investigate the possible causes of these conflicting results and suggests how to improve the efficacies of S. aureus‒targeted therapies. We developed a mathematical model that describes systems-level AD pathogenesis involving dynamic interactions between S. aureus and coagulase-negative Staphylococcus (CoNS). Our model simulation reproduced the clinically observed detrimental effects of the application of S. hominis A9 and flucloxacillin on AD severity and showed that these effects disappeared if the bactericidal activity against CoNS was removed. A hypothetical (modeled) eradication of S. aureus by 3.0 log10 colony-forming unit per cm2 without killing CoNS achieved Eczema Area and Severity Index 75 comparable with that of dupilumab. This efficacy was potentiated if dupilumab was administered in conjunction with S. aureus eradication (Eczema Area and Severity Index 75 at week 16) (S. aureus eradication: 66.7%, dupilumab 61.6% and combination 87.8%). The improved efficacy was also seen for virtual dupilumab poor responders. Our model simulation suggests that killing CoNS worsens AD severity and that S. aureus‒specific eradication without killing CoNS could be effective for patients with AD, including dupilumab poor responders. This study will contribute to designing promising S. aureus‒targeted therapy.
Collapse
Affiliation(s)
- Takuya Miyano
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alan D. Irvine
- Pediatric Dermatology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, College of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Reiko J. Tanaka
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Correspondence: Reiko J. Tanaka, Department of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| |
Collapse
|
59
|
Chang HP, Li Z, Shah DK. Development of a Physiologically-Based Pharmacokinetic Model for Whole-Body Disposition of MMAE Containing Antibody-Drug Conjugate in Mice. Pharm Res 2022; 39:1-24. [PMID: 35044590 DOI: 10.1007/s11095-021-03162-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE To quantitate and mathematically characterize the whole-body pharmacokinetics (PK) of different ADC analytes following administration of an MMAE-conjugated ADC in tumor-bearing mice. METHODS The PK of different ADC analytes (total antibody, total drug, unconjugated drug) was measured following administration of an MMAE-conjugated ADC in tumor-bearing mice. The PK of ADC analytes was compared with the whole-body PK of the antibody and drug obtained following administration of these molecules alone. An ADC PBPK model was developed by linking antibody PBPK model with small-molecule PBPK model, where the drug was assumed to deconjugate in DAR-dependent manner. RESULTS Comparison of antibody biodistribution coefficient (ABC) values for total antibody suggests that conjugation of drug did not significantly affect the PK of antibody. Comparison of tissue:plasma AUC ratio (T/P) for the conjugated drug and total antibody suggests that in certain tissues (e.g., spleen) ADC may demonstrate higher deconjugation. It was observed that the tissue distribution profile of the drug can be altered following its conjugation to antibody. For example, MMAE distribution to the liver was found to increase while its distribution to the heart was found to decrease upon conjugation to antibody. MMAE exposure in the tumor was found to increase by ~20-fold following administration as conjugate (i.e., ADC). The PBPK model was able to a priori predict the PK of all three ADC analytes in plasma, tissues, and tumor reasonably well. CONCLUSIONS The ADC PBPK model developed here serves as a platform for translational and clinical investigations of MMAE containing ADCs.
Collapse
Affiliation(s)
- Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
60
|
Rafidi H, Rajan S, Urban K, Shatz-Binder W, Hui K, Ferl GZ, Kamath AV, Boswell CA. Effect of molecular size on interstitial pharmacokinetics and tissue catabolism of antibodies. MAbs 2022; 14:2085535. [PMID: 35867780 PMCID: PMC9311319 DOI: 10.1080/19420862.2022.2085535] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Advances in antibody engineering have enabled the construction of novel molecular formats in diverse shapes and sizes, providing new opportunities for biologic therapies and expanding the need to understand how various structural aspects affect their distribution properties. To assess the effect of antibody size on systemic pharmacokinetics (PK) and tissue distribution with or without neonatal Fc receptor (FcRn) binding, we evaluated a series of non-mouse-binding anti-glycoprotein D monoclonal antibody formats, including IgG [~150 kDa], one-armed IgG [~100 kDa], IgG-HAHQ (attenuated FcRn binding) [~150 kDa], F(ab')2 [~100 kDa], and F(ab) [~50 kDa]. Tissue-specific concentration-time profiles were corrected for blood content based on vascular volumes and normalized based on interstitial volumes to allow estimation of interstitial concentrations and interstitial:serum concentration ratios. Blood correction demonstrated that the contribution of circulating antibody on total uptake was greatest at early time points and for highly vascularized tissues. Tissue interstitial PK largely mirrored serum exposure profiles. Similar interstitial:serum ratios were obtained for the two FcRn-binding molecules, IgG and one-armed IgG, which reached pseudo-steady-state kinetics in most tissues. For non-FcRn-binding molecules, interstitial:serum ratios changed over time, suggesting that these molecules did not reach steady-state kinetics during the study. Furthermore, concentration-time profiles of both intact and catabolized molecule were measured by a dual tracer approach, enabling quantification of tissue catabolism and demonstrating that catabolism levels were highest for IgG-HAHQ. Overall, these data sets provide insight into factors affecting preclinical distribution and may be useful in estimating interstitial concentrations and/or catabolism in human tissues.
Collapse
Affiliation(s)
- Hanine Rafidi
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Sharmila Rajan
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Konnie Urban
- Safety Assessment, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Whitney Shatz-Binder
- Protein Chemistry, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Keliana Hui
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Gregory Z Ferl
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA.,Biomedical Imaging, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Amrita V Kamath
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - C Andrew Boswell
- Departments of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA.,Biomedical Imaging, Research and Early Development, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
61
|
Engineered human blood-brain barrier microfluidic model for vascular permeability analyses. Nat Protoc 2022; 17:95-128. [PMID: 34997242 DOI: 10.1038/s41596-021-00635-w] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/15/2021] [Indexed: 01/07/2023]
Abstract
The blood-brain barrier (BBB) greatly restricts the entry of biological and engineered therapeutic molecules into the brain. Due to challenges in translating results from animal models to the clinic, relevant in vitro human BBB models are needed to assess pathophysiological molecular transport mechanisms and enable the design of targeted therapies for neurological disorders. This protocol describes an in vitro model of the human BBB self-assembled within microfluidic devices from stem-cell-derived or primary brain endothelial cells, and primary brain pericytes and astrocytes. This protocol requires 1.5 d for device fabrication, 7 d for device culture and up to 5 d for downstream imaging, protein and gene expression analyses. Methodologies to measure the permeability of any molecule in the BBB model, which take 30 min per device, are also included. Compared with standard 2D assays, the BBB model features relevant cellular organization and morphological characteristics, as well as values of molecular permeability within the range expected in vivo. These properties, coupled with a functional brain endothelial expression profile and the capability to easily test several repeats with low reagent consumption, make this BBB model highly suitable for widespread use in academic and industrial laboratories.
Collapse
|
62
|
Ménochet K, Yu H, Wang B, Tibbitts J, Hsu CP, Kamath AV, Richter WF, Baumann A. Non-human primates in the PKPD evaluation of biologics: Needs and options to reduce, refine, and replace. A BioSafe White Paper. MAbs 2022; 14:2145997. [PMID: 36418217 PMCID: PMC9704389 DOI: 10.1080/19420862.2022.2145997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Monoclonal antibodies (mAbs) deliver great benefits to patients with chronic and/or severe diseases thanks to their strong specificity to the therapeutic target. As a result of this specificity, non-human primates (NHP) are often the only preclinical species in which therapeutic antibodies cross-react with the target. Here, we highlight the value and limitations that NHP studies bring to the design of safe and efficient early clinical trials. Indeed, data generated in NHPs are integrated with in vitro information to predict the concentration/effect relationship in human, and therefore the doses to be tested in first-in-human trials. The similarities and differences in the systems defining the pharmacokinetics and pharmacodynamics (PKPD) of mAbs in NHP and human define the nature and the potential of the preclinical investigations performed in NHPs. Examples have been collated where the use of NHP was either pivotal to the design of the first-in-human trial or, inversely, led to the termination of a project prior to clinical development. The potential impact of immunogenicity on the results generated in NHPs is discussed. Strategies to optimize the use of NHPs for PKPD purposes include the addition of PD endpoints in safety assessment studies and the potential re-use of NHPs after non-terminal studies or cassette dosing several therapeutic agents of interest. Efforts are also made to reduce the use of NHPs in the industry through the use of in vitro systems, alternative in vivo models, and in silico approaches. In the case of prediction of ocular PK, the body of evidence gathered over the last two decades renders the use of NHPs obsolete. Expert perspectives, advantages, and pitfalls with these alternative approaches are shared in this review.
Collapse
Affiliation(s)
| | - Hongbin Yu
- R&D Project Management and Development Strategies, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Bonnie Wang
- Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Inc, Princeton, NJ, USA
| | - Jay Tibbitts
- Nonclinical Development, South San Francisco, CA, USA
| | - Cheng-Pang Hsu
- Preclinical Development and Clinical Pharmacology, AskGene Pharma Inc, Camarillo, CA, USA
| | - Amrita V. Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, South San Francisco, CA, USA
| | - Wolfgang F. Richter
- Roche Pharma Research and Early Development, Roche Innovation, Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Andreas Baumann
- R&D, Bayer Pharma AG, Berlin, Germany & Non-clinical Biotech Consulting, Potsdam, Germany °(° present affiliation)
| |
Collapse
|
63
|
Abstract
Mycoplasmas are small, genome-reduced bacteria. They are obligate parasites that can be found in a wide range of host species, including the majority of livestock animals and humans. Colonization of the host can result in a wide spectrum of outcomes. In many cases, these successful parasites are considered commensal, as they are found in the microbiota of asymptomatic carriers. Conversely, mycoplasmas can also be pathogenic, as they are associated with a range of both acute and chronic inflammatory diseases which are problematic in veterinary and human medicine. The chronicity of mycoplasma infections and the ability of these bacteria to infect even recently vaccinated individuals clearly indicate that they are able to successfully evade their host’s humoral immune response. Over the years, multiple strategies of immune evasion have been identified in mycoplasmas, with a number of them aimed at generating important antigenic diversity. More recently, mycoplasma-specific anti-immunoglobulin strategies have also been characterized. Through the expression of the immunoglobulin-binding proteins protein M or mycoplasma immunoglobulin binding (MIB), mycoplasmas have the ability to target the host’s antibodies and to prevent them from interacting with their cognate antigens. In this review, we discuss how these discoveries shed new light on the relationship between mycoplasmas and their host’s immune system. We also propose that these strategies should be taken into consideration for future studies, as they are key to our understanding of mycoplasma diseases' chronic and inflammatory nature and are probably a contributing factor to reduce vaccine efficacy.
Collapse
|
64
|
Tang L, Feng Y, Gao S, Mu Q, Liu C. Nanotherapeutics Overcoming the Blood-Brain Barrier for Glioblastoma Treatment. Front Pharmacol 2021; 12:786700. [PMID: 34899350 PMCID: PMC8655904 DOI: 10.3389/fphar.2021.786700] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/10/2021] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor with a poor prognosis. The current standard treatment regimen represented by temozolomide/radiotherapy has an average survival time of 14.6 months, while the 5-year survival rate is still less than 5%. New therapeutics are still highly needed to improve the therapeutic outcome of GBM treatment. The blood-brain barrier (BBB) is the main barrier that prevents therapeutic drugs from reaching the brain. Nanotechnologies that enable drug delivery across the BBB hold great promise for the treatment of GBM. This review summarizes various drug delivery systems used to treat glioma and focuses on their approaches for overcoming the BBB to enhance the accumulation of small molecules, protein and gene drugs, etc. in the brain.
Collapse
Affiliation(s)
- Lin Tang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yicheng Feng
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Sai Gao
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Qingchun Mu
- The People’s Hospital of Gaozhou, Gaozhou, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
65
|
Gevenois PJLY, De Pauw P, Schoonooghe S, Delporte C, Sebti T, Amighi K, Muyldermans S, Wauthoz N. Development of Neutralizing Multimeric Nanobody Constructs Directed against IL-13: From Immunization to Lead Optimization. THE JOURNAL OF IMMUNOLOGY 2021; 207:2608-2620. [PMID: 34645688 DOI: 10.4049/jimmunol.2100250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/16/2021] [Indexed: 11/19/2022]
Abstract
IL-13 is a pleiotropic cytokine mainly secreted by Th2 cells. It reacts with many different types of cells involved in allergy, inflammation, and fibrosis, e.g., mastocytes, B cells, and fibroblasts. The role of IL-13 in conditions involving one or several of these phenotypes has therefore been extensively investigated. The inhibition of this cytokine in animal models for various pathologies yielded highly promising results. However, most human trials relying on anti-IL-13 conventional mAbs have failed to achieve a significant improvement of the envisaged disorders. Where some studies might have suffered from several weaknesses, the strategies themselves, such as targeting only IL-13 using conventional mAbs or employing a systemic administration, could be questioned. Nanobodies are recombinant Ag-binding fragments derived from the variable part of H chain-only Abs occurring in Camelidae. Thanks to their single-domain structure, small size (≈15 kDa), good stability, and solubility, they can be engineered into multispecific constructs for combined therapies or for use in new strategies such as formulations for local administration, e.g., pulmonary administration. In this study, we describe the generation of 38 nanobodies that can be subdivided into five CDR3 families. Nine nanobodies were found to have a good affinity profile (KD = 1-200 nM), but none were able to strongly inhibit IL-13 biological activity in vitro (IC50 > 50 µM: HEK-Blue IL-13/IL-4 cells). Multimeric constructs were therefore designed from these inhibitors and resulted in an up to 36-fold improvement in affinity and up to 300-fold enhancement of the biological activity while conserving a high specificity toward IL-13.
Collapse
Affiliation(s)
- Philippe J-L Y Gevenois
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium;
| | - Pieter De Pauw
- Laboratory of Cellular and Molecular Immunology, Free University of Brussels, Ixelles, Belgium
| | - Steve Schoonooghe
- Flemish Institute for Biotechnology Nanobody Core, Free University of Brussels, Brussels, Belgium
| | - Cédric Delporte
- Unit of Pharmacognosy, Bioanalysis and Drug Discovery, RD3 and Analytical Platform of the Faculty of Pharmacy, Free University of Brussels, Brussels, Belgium; and
| | | | - Karim Amighi
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Free University of Brussels, Ixelles, Belgium
| | - Nathalie Wauthoz
- Unit of Pharmaceutics and Biopharmaceutics, Free University of Brussels, Faculty of Pharmacy, Brussels, Belgium
| |
Collapse
|
66
|
The in vitro and in vivo efficacy of CT-P59 against Gamma, Delta and its associated variants of SARS-CoV-2. Biochem Biophys Res Commun 2021; 578:91-96. [PMID: 34547629 PMCID: PMC8436435 DOI: 10.1016/j.bbrc.2021.09.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022]
Abstract
The SARS-CoV-2 variant is rapidly spreading across the world and causes to resurge infections. We previously reported that CT-P59 presented its in vivo potency against Beta variants, despite its reduced activity in cell experiments. Yet, it remains uncertain to exert the antiviral effect of CT-P59 on Gamma, Delta and its associated variants (L452R). To tackle this question, we carried out cell tests and animal studies. CT-P59 showed neutralization against Gamma, Delta, Epsilon, and Kappa variants in cells, with reduced susceptibility. The mouse challenge experiments with Gamma and Delta variants substantiated in vivo potency of CT-P59 showing symptom remission and virus abrogation in the respiratory tract. Collectively, cell and animal studies showed that CT-P59 is effective against Gamma and Delta variants infection, hinting that CT-P59 has therapeutic potential for patients infected with Gamma, Delta and its associated variants.
Collapse
|
67
|
Bussing D, Li Z, Li Y, Chang HP, Chang HY, Guo L, Verma A, Shah DK. Pharmacokinetics of Monoclonal Antibody and Antibody Fragments in the Mouse Eye Following Systemic Administration. AAPS JOURNAL 2021; 23:116. [PMID: 34750690 PMCID: PMC8575492 DOI: 10.1208/s12248-021-00647-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022]
Abstract
The ocular pharmacokinetics (PK) of antibody-based therapies are infrequently studied in mice due to the technical difficulties in working with the small murine eye. This study is the first of its kind to quantitatively measure the PK of variously sized proteins in the plasma, cornea/ICB, vitreous humor, retina, and posterior cup (including choroid) of the mouse and to evaluate the relationship between molecular weight (MW) and antibody biodistribution coefficient (BC) to the eye. Proteins analyzed include trastuzumab (150 kDa), trastuzumab-vc-MMAE (T-vc-MMAE, 155 kDa), F(ab)2 (100 kDa), Fab (50 kDa), and scFv (27 kDa). As expected, ocular PK mirrored the systemic PK as plasma was the driving force for ocular exposure. For trastuzumab, T-vc-MMAE, F(ab)2, Fab, and scFv, respectively, the BCs in the cornea/ICB were 0.610%, 0.475%, 1.74%, 3.39%, and 13.7%; the BCs in the vitreous humor were 0.0198%, 0.0427%, 0.0934%, 0.234%, and 5.56%; the BCs for the retina were 0.539%, 0.230%, 0.704%, 2.44%, and 20.4%; the BCs for the posterior cup were 0.557%, 0.650%, 1.47%, 4.06%, and 13.9%. The relationship between BC and MW was best characterized by a log–log regression in which BC decreased as MW increased, with every doubling in MW leading to a decrease in BC by a factor of 3.44 × , 6.76 × , 4.74 × , and 3.43 × in cornea/ICB, vitreous humor, retina, and posterior cup, respectively. In analyzing the disposition of protein therapeutics to the eye, these findings enhance our understanding of the potential for ocular toxicity of systemically administered protein therapeutics and may aid in the discovery of systemically administered protein therapeutics for ocular disorders.
Collapse
Affiliation(s)
- David Bussing
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Yingyi Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Hsueh-Yuan Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Leiming Guo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Ashwni Verma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
68
|
Chigutsa E, Jordie E, Riggs M, Nirula A, Elmokadem A, Knab T, Chien JY. A Quantitative Modeling and Simulation Framework to Support Candidate and Dose Selection of Anti-SARS-CoV-2 Monoclonal antibodies to Advance Bamlanivimab into a First-in-Human Clinical Trial. Clin Pharmacol Ther 2021; 111:595-604. [PMID: 34687040 PMCID: PMC8653169 DOI: 10.1002/cpt.2459] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022]
Abstract
Neutralizing monoclonal antibodies (mAb), novel therapeutics for the treatment of coronavirus disease 2019 (COVID‐19) caused by severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2), have been urgently researched from the start of the pandemic. The selection of the optimal mAb candidate and therapeutic dose were expedited using open‐access in silico models. The maximally effective therapeutic mAb dose was determined through two approaches; both expanded on innovative, open‐science initiatives. A physiologically‐based pharmacokinetic (PBPK) model, incorporating physicochemical properties predictive of mAb clearance and tissue distribution, was used to estimate mAb exposure that maintained concentrations above 90% inhibitory concentration of in vitro neutralization in lung tissue for up to 4 weeks in 90% of patients. To achieve fastest viral clearance following onset of symptoms, a longitudinal SARS‐CoV‐2 viral dynamic model was applied to estimate viral clearance as a function of drug concentration and dose. The PBPK model‐based approach suggested that a clinical dose between 175 and 500 mg of bamlanivimab would maintain target mAb concentrations in the lung tissue over 28 days in 90% of patients. The viral dynamic model suggested a 700 mg dose would achieve maximum viral elimination. Taken together, the first‐in‐human trial (NCT04411628) conservatively proceeded with a starting therapeutic dose of 700 mg and escalated to higher doses to evaluate the upper limit of safety and tolerability. Availability of open‐access codes and application of novel in silico model‐based approaches supported the selection of bamlanivimab and identified the lowest dose evaluated in this study that was expected to result in the maximum therapeutic effect before the first‐in‐human clinical trial.
Collapse
Affiliation(s)
| | - Eric Jordie
- Metrum Research Group, Inc., Tariffville, Connecticut, USA
| | - Matthew Riggs
- Metrum Research Group, Inc., Tariffville, Connecticut, USA
| | - Ajay Nirula
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Tim Knab
- Metrum Research Group, Inc., Tariffville, Connecticut, USA
| | | |
Collapse
|
69
|
Sjöström EO, Culot M, Leickt L, Åstrand M, Nordling E, Gosselet F, Kaiser C. Transport study of interleukin-1 inhibitors using a human in vitro model of the blood-brain barrier. Brain Behav Immun Health 2021; 16:100307. [PMID: 34589799 PMCID: PMC8474601 DOI: 10.1016/j.bbih.2021.100307] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/24/2021] [Indexed: 01/15/2023] Open
Abstract
The proinflammatory cytokine Interleukin-1 (IL-1), with its two isoforms α and β, has important roles in multiple pathogenic processes in the central nervous system. The present study aimed to evaluate and compare the blood-to-brain distribution of anakinra (IL-1 receptor antagonist), bermekimab (IL-1α antagonist) and canakinumab (IL-1β antagonist). A human in vitro model of the blood-brain barrier derived from human umbilical cord blood stem cells was used, where isolated CD34+ cells co-cultured with bovine pericytes were matured into polarized brain-like endothelial cells. Transport rates of the three test items were evaluated after 180 min incubation at concentrations 50, 250 and 1250 nM in a transwell system. We report herein that anakinra passes the human brain-like endothelial monolayer at a 4-7-fold higher rate than the monoclonal antibodies tested. Both antibodies had similar transport rates at all concentrations. No dose-dependent effects in transport rates were observed, nor any saturation effects at supraphysiological concentrations. The larger propensity of anakinra to pass this model of the human blood-brain barrier supports existing data and confirms that anakinra can reach the brain compartment at clinically relevant concentrations. As anakinra inhibits the actions of both IL-1α and IL-1β, it blocks all effects of IL-1 downstream signaling. The results herein further add to the growing body of evidence of the potential utility of anakinra to treat neuroinflammatory disorders. Anakinra has a larger propensity to pass the in vitro BBB than monoclonal antibodies targeting the IL-1 system. Implications for targeting inflammation in cerebral ischemia and neurological sequelae of autoinflammatory diseases. Novel and comparative study of biologics in a human in vitro BBB model shows relevance and validity.
Collapse
Affiliation(s)
| | - Maxime Culot
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Lisa Leickt
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Mikael Åstrand
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Erik Nordling
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| | - Fabien Gosselet
- Univ. Artois, UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300, Lens, France
| | - Christina Kaiser
- Swedish Orphan Biovitrum AB (publ), SE-112 76, Stockholm, Sweden
| |
Collapse
|
70
|
Wang L, Chen Y, Zhou W, Miao X, Zhou H. Utilization of physiologically-based pharmacokinetic model to assess disease-mediated therapeutic protein-disease-drug interaction in immune-mediated inflammatory diseases. Clin Transl Sci 2021; 15:464-476. [PMID: 34581012 PMCID: PMC8841519 DOI: 10.1111/cts.13164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 11/30/2022] Open
Abstract
It is known that interleukin-6 (IL-6) can significantly modulate some key drug-metabolizing enzymes, such as phase I cytochrome P450s (CYPs). In this study, a physiologically-based pharmacokinetic (PBPK) model was developed to assess CYPs mediated therapeutic protein drug interactions (TP-DIs) in patients with immune-mediated inflammatory diseases (IMIDs) with elevated systemic IL-6 levels when treated by anti-IL-6 therapies. Literature data of IL-6 levels in various diseases were incorporated in SimCYP to construct respective virtual patient populations. The modulation effects of systemic IL-6 level and local IL-6 level in the gastrointestinal tract (GI) on CYPs activities were assessed. Upon blockade of the IL-6 signaling pathway by an anti-IL-6 treatment, the area under plasma concentration versus time curves (AUCs) of S-warfarin, omeprazole, and midazolam were predicted to decrease by up to 40%, 42%, and 46%, respectively. In patients with Crohn's disease and ulcerative colitis treated with an anti-IL-6 therapy, the lowering of the elevated IL-6 levels in the local GI tissue were predicted to result in further decreases in AUCs of those CYP substrates. The propensity of TP-DIs under comorbidity conditions, such as in patients with cancer with IMID, were also explored. With further validation with relevant clinical data, this PBPK model may provide an in silico way to quantify the magnitude of potential TP-DI in patients with elevated IL-6 levels when an anti-IL-6 therapeutic is used with concomitant small-molecule drugs. This model may be further adapted to evaluate the CYP modulation effect by other therapeutic modalities, which would significantly alter levels of proinflammatory cytokines during the treatment period.
Collapse
Affiliation(s)
- Lujing Wang
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Yang Chen
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Wangda Zhou
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Xin Miao
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Honghui Zhou
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| |
Collapse
|
71
|
Chigutsa E, O'Brien L, Ferguson-Sells L, Long A, Chien J. Population Pharmacokinetics and Pharmacodynamics of the Neutralizing Antibodies Bamlanivimab and Etesevimab in Patients With Mild to Moderate COVID-19 Infection. Clin Pharmacol Ther 2021; 110:1302-1310. [PMID: 34514598 PMCID: PMC8652670 DOI: 10.1002/cpt.2420] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022]
Abstract
Bamlanivimab and etesevimab are neutralizing antibodies indicated for treatment of coronavirus disease 2019 (COVID-19) in patients with early mild or moderate disease. We present the use of pharmacokinetic/pharmacodynamic (PK/PD) modeling that characterizes the timecourse of viral load obtained from 2,970 patients from 2 phase II clinical trials. The model was used for identification of optimal doses that would result in at least 90% of patients achieving serum drug concentrations that result in 90% of maximum drug effect (IC90) for at least 28 days. The serum IC90 (95% confidence interval) was estimated to be 4.2 (3.2-4.3) µg/mL for bamlanivimab and 12.6 (9.7-12.8) µg/mL for etesevimab. Observed clinical trial data confirmed PK and PK/PD model predictions that doses of 700 mg bamlanivimab and 1,400 mg etesevimab would result in maximum reduction in viral load, with no additional effect seen at higher doses. No dose adjustment is recommended as age, sex, race, baseline viral load, and hepatic impairment did not have a significant impact on the PK of the antibodies. Earlier drug administration resulted in greater reductions in viral load, demonstrating the importance of receiving treatment as soon as possible. Relative to placebo, typical reduction in viral load over a 7-day period was estimated to be 80 or 93% (drug administered 4 days or 1 day after the onset of symptoms, respectively), P < 0.0001. PK/PD modeling and simulation was pivotal throughout the drug development and emergency use authorization process.
Collapse
Affiliation(s)
- Emmanuel Chigutsa
- Global PK/PD & Pharmacometrics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Lisa O'Brien
- Global PK/PD & Pharmacometrics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Lisa Ferguson-Sells
- Global PK/PD & Pharmacometrics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Amanda Long
- Global PK/PD & Pharmacometrics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jenny Chien
- Global PK/PD & Pharmacometrics, Eli Lilly and Company, Indianapolis, Indiana, USA
| |
Collapse
|
72
|
Kapitanov GI, Chabot JR, Narula J, Roy M, Neubert H, Palandra J, Farrokhi V, Johnson JS, Webster R, Jones HM. A Mechanistic Site-Of-Action Model: A Tool for Informing Right Target, Right Compound, And Right Dose for Therapeutic Antagonistic Antibody Programs. FRONTIERS IN BIOINFORMATICS 2021; 1:731340. [DOI: 10.3389/fbinf.2021.731340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Quantitative modeling is increasingly utilized in the drug discovery and development process, from the initial stages of target selection, through clinical studies. The modeling can provide guidance on three major questions–is this the right target, what are the right compound properties, and what is the right dose for moving the best possible candidate forward. In this manuscript, we present a site-of-action modeling framework which we apply to monoclonal antibodies against soluble targets. We give a comprehensive overview of how we construct the model and how we parametrize it and include several examples of how to apply this framework for answering the questions postulated above. The utilities and limitations of this approach are discussed.
Collapse
|
73
|
Mao Y, Evans EE, Mishra V, Balch L, Eberhardt A, Zauderer M, Gold WA. Anti-Semaphorin 4D Rescues Motor, Cognitive, and Respiratory Phenotypes in a Rett Syndrome Mouse Model. Int J Mol Sci 2021; 22:ijms22179465. [PMID: 34502373 PMCID: PMC8431088 DOI: 10.3390/ijms22179465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 01/09/2023] Open
Abstract
Rett syndrome is a neurodevelopmental disorder caused by mutations of the methyl-CpG binding protein 2 gene. Abnormal physiological functions of glial cells contribute to pathogenesis of Rett syndrome. Semaphorin 4D (SEMA4D) regulates processes central to neuroinflammation and neurodegeneration including cytoskeletal structures required for process extension, communication, and migration of glial cells. Blocking SEMA4D-induced gliosis may preserve normal glial and neuronal function and rescue neurological dysfunction in Rett syndrome. We evaluated the pre-clinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody in the Rett syndrome Mecp2T158A transgenic mouse model and investigated the contribution of glial cells as a proposed mechanism of action in treated mice and in primary glial cultures isolated from Mecp2T158A/y mutant mice. SEMA4D is upregulated in neurons while glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1-positive cells are upregulated in Mecp2T158A/y mice. Anti-SEMA4D treatment ameliorates Rett syndrome-specific symptoms and improves behavioural functions in both pre-symptomatic and symptomatic cohorts of hemizygous Mecp2T158A/y male mice. Anti-SEMA4D also reduces astrocyte and microglia activation in vivo. In vitro experiments demonstrate an abnormal cytoskeletal structure in mutant astrocytes in the presence of SEMA4D, while anti-SEMA4D antibody treatment blocks SEMA4D–Plexin B1 signaling and mitigates these abnormalities. These results suggest that anti-SEMA4D immunotherapy may be an effective treatment option to alleviate symptoms and improve cognitive and motor function in Rett syndrome.
Collapse
Affiliation(s)
- Yilin Mao
- Molecular Neurobiology Research Laboratory, Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia;
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Elizabeth E. Evans
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Vikas Mishra
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Leslie Balch
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Allison Eberhardt
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Maurice Zauderer
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Wendy A. Gold
- Molecular Neurobiology Research Laboratory, Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia;
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Molecular Neurobiology Research Laboratory, The Children’s Medical Research Institute, Westmead, NSW 2145, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
74
|
Tolerability, Safety, Pharmacokinetics, and Immunogenicity of a Novel SARS-CoV-2 Neutralizing Antibody, Etesevimab, in Chinese Healthy Adults: a Randomized, Double-Blind, Placebo-Controlled, First-in-Human Phase 1 Study. Antimicrob Agents Chemother 2021; 65:e0035021. [PMID: 33972256 DOI: 10.1128/aac.00350-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread rapidly worldwide. This study is the first to report the tolerability, safety, pharmacokinetics (PK), and immunogenicity of a recombinant human anti-SARS-CoV-2 monoclonal antibody, etesevimab (CB6, JS016, LY3832479, or LY-CoV016), in healthy adults. This paper describes a randomized, double-blind, placebo-controlled, phase 1 study. A total of 40 participants were enrolled to receive a single intravenous dose of either etesevimab or placebo in one of four sequential ascending intravenous dose cohorts. All 40 participants completed the study. Seventeen (42.5%) participants experienced 22 treatment emergent adverse events (TEAEs) that were drug-related, and the rates of these TEAEs among different dose cohorts were numerically comparable. No difference was observed between the combined etesevimab group and the placebo group. The exposure after etesevimab infusion increased in an approximately proportional manner as the dose increased from 2.5 to 50 mg/kg. The elimination half-life (t1/2) value did not differ among different dose cohorts and was estimated to be around 4 weeks. Etesevimab was well tolerated after administration of a single dose at a range of 2.5 mg/kg to 50 mg/kg in healthy Chinese adults. The PK profiles of etesevimab in healthy volunteers showed typical monoclonal antibody distribution and elimination characteristics. (This study has been registered at ClinicalTrials.gov under identifier NCT04441918.).
Collapse
|
75
|
Stacey HD, Golubeva D, Posca A, Ang JC, Novakowski KE, Zahoor MA, Kaushic C, Cairns E, Bowdish DME, Mullarkey CE, Miller MS. IgA potentiates NETosis in response to viral infection. Proc Natl Acad Sci U S A 2021; 118:e2101497118. [PMID: 34183391 PMCID: PMC8271757 DOI: 10.1073/pnas.2101497118] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IgA is the second most abundant antibody present in circulation and is enriched at mucosal surfaces. As such, IgA plays a key role in protection against a variety of mucosal pathogens including viruses. In addition to neutralizing viruses directly, IgA can also stimulate Fc-dependent effector functions via engagement of Fc alpha receptors (Fc-αRI) expressed on the surface of certain immune effector cells. Neutrophils are the most abundant leukocyte, express Fc-αRI, and are often the first to respond to sites of injury and infection. Here, we describe a function for IgA-virus immune complexes (ICs) during viral infections. We show that IgA-virus ICs potentiate NETosis-the programmed cell-death pathway through which neutrophils release neutrophil extracellular traps (NETs). Mechanistically, IgA-virus ICs potentiated a suicidal NETosis pathway via engagement of Fc-αRI on neutrophils through a toll-like receptor-independent, NADPH oxidase complex-dependent pathway. NETs also were capable of trapping and inactivating viruses, consistent with an antiviral function.
Collapse
Affiliation(s)
- Hannah D Stacey
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Diana Golubeva
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Alyssa Posca
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Jann C Ang
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Kyle E Novakowski
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Muhammad Atif Zahoor
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Charu Kaushic
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Ewa Cairns
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 3K7
- Department of Medicine, Division of Rheumatology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 3K7
| | - Dawn M E Bowdish
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Caitlin E Mullarkey
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Matthew S Miller
- Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada, L8S 4K1;
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada, L8S 4K1
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada, L8S 4K1
| |
Collapse
|
76
|
Gao F, Zhang X. Pharmacokinetic profiles of a SARS-COV-2 neutralizing antibody BD-604 in cynomolgus monkeys. Drug Test Anal 2021; 13:1727-1734. [PMID: 34215019 PMCID: PMC8427073 DOI: 10.1002/dta.3122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Currently, severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) has spread worldwide as a severe pandemic and effective therapeutic medications are urgently needed. As reported previously, BD-604 is a fully human monoclonal antibody with strong in vitro and in vivo neutralizing activity to SARS-COV-2. OBJECTIVE The purpose of this study was to characterize the pharmacokinetic propertie of BD-604 in cynomolgus monkeys. METHODS To analyze the concentration of BD-604 in cynomolgus monkey serum, an ELISA assay was established, and a systemic validation was performed including accuracy and precision, dilution linearity and hook effect, selectivity, specificity, stability, and parallelism tests. Then, six naïve cynomolgus monkeys (3/sex) were administered BD-604 at a single dose of 10 mg/kg via intravenous infusion (60 min). Blood samples were collected at various time points (0-672 h) and analyzed for serum concentrations of BD-604. RESULTS The data from validation experiments assure the reproducibility and reliability of the established ELISA assay. Then, the validated method was used to measure BD-604 concentration in cynomolgus monkey serum. The pharmacokinetics parameters including terminal half-life (t1/2 ), peak serum concentration (Cmax ), area under curve from time zero to last timepoint or infinity (AUClast /AUCinf ), apparent volume of distribution (Vz ), clearance rate (CL), and mean residence time (MRT) were calculated and reported. BD-604 showed no marked sex differences at the dose of 10 mg/kg when comparing the AUC0-last and Cmax between female and male cynomolgus monkeys. CONCLUSION In cynomolgus monkeys, BD-604 possesses pharmacokinetic properties similar to natural IgGs.
Collapse
Affiliation(s)
- Feng Gao
- Singlomics (Beijing DanXu Biopharmaceuticals) Co., Ltd., Beijing, China
| | - Xu Zhang
- Singlomics (Beijing DanXu Biopharmaceuticals) Co., Ltd., Beijing, China
| |
Collapse
|
77
|
Two-pore physiologically based pharmacokinetic model validation using whole-body biodistribution of trastuzumab and different-size fragments in mice. J Pharmacokinet Pharmacodyn 2021; 48:743-762. [PMID: 34146191 DOI: 10.1007/s10928-021-09772-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
In the past, our lab proposed a two-pore PBPK model for different-size protein therapeutics using de novo derived parameters and the model was validated using plasma PK data of different-size antibody fragments digitized from the literature (Li Z, Shah DK, J Pharmacokinet Pharmacodynam 46(3):305-318, 2009). To further validate the model using tissue distribution data, whole-body biodistribution study of 6 different-size proteins in mice were conducted. Studied molecules covered a wide MW range (13-150 kDa). Plasma PK and tissue distribution profiles is 9 tissues were measured, including heart, lung, liver, spleen, kidney, skin, muscle, small intestine, large intestine. Tumor exposure of different-size proteins were also evaluated. The PBPK model was validated by comparing percentage predictive errors (%PE) between observed and model predicted results for each type of molecule in each tissue. Model validation showed that the two-pore PBPK model was able to predict plasma, tissues and tumor PK of all studied molecules relatively well. This model could serve as a platform for developing a generic PBPK model for protein therapeutics in the future.
Collapse
|
78
|
Fernandes CFC, Pereira SS, Luiz MB, Silva NKRL, Silva MCS, Marinho ACM, Fonseca MHG, Furtado GP, Trevizani R, Nicolete R, Soares AM, Zuliani JP, Stabeli RG. Engineering of single-domain antibodies for next-generation snakebite antivenoms. Int J Biol Macromol 2021; 185:240-250. [PMID: 34118288 DOI: 10.1016/j.ijbiomac.2021.06.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/04/2021] [Accepted: 06/06/2021] [Indexed: 12/29/2022]
Abstract
Given the magnitude of the global snakebite crisis, strategies to ensure the quality of antivenom, as well as the availability and sustainability of its supply are under development by several research groups. Recombinant DNA technology has allowed the engineering of monoclonal antibodies and recombinant fragments as alternatives to conventional antivenoms. Besides having higher therapeutic efficacy, with broad neutralization capacity against local and systemic toxicity, novel antivenoms need to be safe and cost-effective. Due to the biological and physical chemical properties of camelid single-domain antibodies, with high volume of distribution to distal tissue, their modular format, and their versatility, their biotechnological application has grown considerably in recent decades. This article presents the most up-to-date developments concerning camelid single-domain-based antibodies against major toxins from snake venoms, the main venomous animals responsible for reported envenoming cases and related human deaths. A brief discussion on the composition, challenges, and perspectives of antivenoms is presented, as well as the road ahead for next-generation antivenoms based on single-domain antibodies.
Collapse
Affiliation(s)
| | - Soraya S Pereira
- Fundação Oswaldo Cruz, Fiocruz Rondônia, and Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Marcos B Luiz
- Fundação Oswaldo Cruz, Fiocruz Rondônia, and Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Nauanny K R L Silva
- Fundação Oswaldo Cruz, Fiocruz Rondônia, and Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Marcela Cristina S Silva
- Fundação Oswaldo Cruz, Fiocruz Rondônia, and Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | | | | | | | | | | | - Andreimar M Soares
- Fundação Oswaldo Cruz, Fiocruz Rondônia, and Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Juliana P Zuliani
- Fundação Oswaldo Cruz, Fiocruz Rondônia, and Instituto Nacional de Ciência e Tecnologia em Epidemiologia da Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil; Universidade Federal de Rondônia, UNIR, Porto Velho, Rondônia, Brazil
| | - Rodrigo G Stabeli
- Plataforma Bi-Institucional de Medicina Translacional (Fiocruz-USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
79
|
Ye Q, Wang Y, Shen S, Xu C, Wang J. Biomaterials-Based Delivery of Therapeutic Antibodies for Cancer Therapy. Adv Healthc Mater 2021; 10:e2002139. [PMID: 33870637 DOI: 10.1002/adhm.202002139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/05/2021] [Indexed: 12/19/2022]
Abstract
Considerable breakthroughs in the treatment of malignant tumors using antibody drugs, especially immunomodulating monoclonal antibodies (mAbs), have been made in the past decade. Despite technological advancements in antibody design and manufacture, multiple challenges face antibody-mediated cancer therapy, such as instability in vivo, poor tumor penetration, limited response rate, and undesirable off-target cytotoxicity. In recent years, an increasing number of biomaterials-based delivery systems have been reported to enhance the antitumor efficacy of antibody drugs. This review summarizes the advances and breakthroughs in integrating biomaterials with therapeutic antibodies for enhanced cancer therapy. A brief introduction to the principal mechanism of antibody-based cancer therapy is first established, and then various antibody immobilization strategies are provided. Finally, the current state-of-the-art in biomaterials-based antibody delivery systems and their applications in cancer treatment are summarized, highlighting how the delivery systems augment the therapeutic efficacy of antibody drugs. The outlook and perspective on biomaterials-based delivery of antitumor antibodies are also discussed.
Collapse
Affiliation(s)
- Qian‐Ni Ye
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
| | - Yue Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Shenzhen Bay Laboratory Shenzhen 518132 P. R. China
| | - Song Shen
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Cong‐Fei Xu
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction South China University of Technology Guangzhou 510006 P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction South China University of Technology Guangzhou 510006 P. R. China
| |
Collapse
|
80
|
Dimelow R, Ji B, Struemper H. Pharmacokinetics of Belimumab in Children With Systemic Lupus Erythematosus. Clin Pharmacol Drug Dev 2021; 10:622-633. [PMID: 33245847 PMCID: PMC8246766 DOI: 10.1002/cpdd.889] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
The phase 2 placebo-controlled, double-blind PLUTO trial characterized the pharmacokinetics of belimumab plus standard systemic lupus erythematosus (SLE) therapy in patients with childhood-onset SLE (cSLE) and demonstrated similar efficacy and safety to that in adult SLE. Patients with active cSLE aged 5-17 years were randomized to intravenous belimumab 10 mg/kg every 4 weeks (n = 53). A linear 2-compartment population pharmacokinetics (popPK) model with first-order elimination was developed, and an exploratory exposure-response analysis assessed the impact of between-patient exposure variability on clinical response (SLE Responder Index 4 [SRI4]) in week 52, and occurrence of serious adverse events during the study. The popPK model estimated clearance of 158 mL/day, steady-state volume of distribution of 3.5 L, terminal half-life of 16.3 days, and distribution half-life of 0.8 days in the overall population. Fat-free mass (FFM) better characterized the pharmacokinetics than total body weight and was more consistent with allometric scaling theory; belimumab pharmacokinetics were largely determined by FFM. Age, sex, disease activity, and concomitant medication had no impact on pediatric belimumab exposure after accounting for body size. Individual and median steady-state pediatric pharmacokinetic profiles were similar to known adult profiles and pediatric exposure estimates for belimumab 10 mg/kg intravenously were consistent with adult exposures. Exposures were similar between SRI4 responders and nonresponders, and patients who did or did not experience a serious adverse event. There was no clinically relevant correlation between exposure and efficacy or safety, confirming belimumab 10 mg/kg intravenous dose every 4 weeks as appropriate for pediatric patients with cSLE.
Collapse
MESH Headings
- Administration, Intravenous
- Adolescent
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Child
- Child, Preschool
- Double-Blind Method
- Female
- Half-Life
- Humans
- Immunosuppressive Agents/administration & dosage
- Immunosuppressive Agents/adverse effects
- Immunosuppressive Agents/pharmacokinetics
- Linear Models
- Lupus Erythematosus, Systemic/drug therapy
- Male
- Models, Biological
- Tissue Distribution
- Treatment Outcome
Collapse
Affiliation(s)
- Richard Dimelow
- GSKClinical Pharmacology Modeling & SimulationStevenageHertfordshireUK
| | - Beulah Ji
- GSKClinical Pharmacology Modeling & SimulationStevenageHertfordshireUK
| | - Herbert Struemper
- GSKClinical Pharmacology Modeling & SimulationResearch Triangle ParkNorth CarolinaUSA
| |
Collapse
|
81
|
Davda J, Reynolds K, Davis JD, Smith PF. Blueprint for pandemic response: Focus on translational medicine, clinical pharmacology and pharmacometrics. Br J Clin Pharmacol 2021; 87:3398-3407. [PMID: 33855747 DOI: 10.1111/bcp.14859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/19/2021] [Accepted: 04/04/2021] [Indexed: 12/14/2022] Open
Abstract
Perhaps the most important lesson learned from the COVID-19 pandemic is that of preparedness. Enhanced surveillance systems for early threat detection will be crucial to maximizing response time for implementation of public health measures and mobilization of resources in containing an emerging pandemic. Recent outbreaks have been dominated by viral pathogens, with RNA respiratory viruses being the most likely to have pandemic potential. These should therefore be a preparedness priority. Tools in the areas of virology, drug discovery, clinical pharmacology, translational medicine and pharmacometrics should be considered key components in the rapid identification and development of existing and novel interventions for a pandemic response. Prioritization of therapeutics should be based on in vitro activity, likelihood of achieving effective drug concentrations at the site of action, and safety profile at the doses that will be required for clinical efficacy. Deployment strategies must be tailored to the epidemiology of the disease, and the adequacy of the response should be re-evaluated in view of evolving epidemiological factors. An interdisciplinary framework integrating drug pharmacology, viral kinetics, epidemiology and health economics could help optimize the deployment strategy by improving decision-making around who to treat, when to treat, and with what type of intervention for optimal outcomes. Lastly, while an effective vaccine will ultimately end a pandemic, antiviral drug intervention guided by clinical pharmacology principles will continue to play a critical role in any pandemic response.
Collapse
Affiliation(s)
| | - Kellie Reynolds
- Division of Infectious Disease Pharmacology (DIDP), Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS), Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - John D Davis
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | | |
Collapse
|
82
|
Gairy K, Knight C, Anthony P, Hoskin B. Burden of illness among subgroups of patients with primary Sjögren's syndrome and systemic involvement. Rheumatology (Oxford) 2021; 60:1871-1881. [PMID: 33147609 PMCID: PMC8023993 DOI: 10.1093/rheumatology/keaa508] [Citation(s) in RCA: 10] [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: 04/21/2020] [Revised: 07/19/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES To describe how patients with primary SS (pSS) and systemic organ involvement are classified and clustered in routine practice. METHODS This multinational, cross-sectional survey of real-world quantitative data was conducted across Europe and the US. Rheumatologists who treated seven or more adult patients per month with pSS and current/past systemic manifestations undertook a survey before completing a patient record form capturing demographic, clinical and treatment information for their next six eligible patients. Patients with a completed patient record form were invited to complete a patient self-completion questionnaire capturing insights into their disease and treatment. Subgroups were defined by physicians' assessment of disease severity; clusters were derived based on key clinical characteristics using latent class analysis. RESULTS Rheumatologists completed 316 physician surveys and 1879 patient record forms; 888 patients completed the patient self-completion questionnaire. pSS severity reflected organ involvement and symptomatology. Latent class analysis produced five clusters distinguished by the organ systems involved and the presence of pain and fatigue symptoms at the time of the survey. A minority of patients [n = 67 (4%)] were categorized with multiple organ involvement and the highest frequency of pain and fatigue. A total of 324 patients (17%) were categorized as 'low burden'. The remaining three clusters exhibited high frequencies of articular involvement but were distinguished by the extent of other organ system involvement. CONCLUSION Cluster analysis using a real-world cohort of patients with pSS and systemic organ involvement highlights the heterogeneous presentation of patients with pSS and confirms the importance of pain and fatigue as well as organ involvement when determining disease burden.
Collapse
Affiliation(s)
- Kerry Gairy
- Value Evidence & Outcomes, GlaxoSmithKline, Brentford, Middlesex, UK
| | - Claudia Knight
- Adelphi Real World, Adelphi Group, Macclesfield, Cheshire, UK
| | - Papa Anthony
- Adelphi Real World, Adelphi Group, Macclesfield, Cheshire, UK
| | - Ben Hoskin
- Adelphi Real World, Adelphi Group, Macclesfield, Cheshire, UK
| |
Collapse
|
83
|
Modeling Pharmacokinetics and Pharmacodynamics of Therapeutic Antibodies: Progress, Challenges, and Future Directions. Pharmaceutics 2021; 13:pharmaceutics13030422. [PMID: 33800976 PMCID: PMC8003994 DOI: 10.3390/pharmaceutics13030422] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022] Open
Abstract
With more than 90 approved drugs by 2020, therapeutic antibodies have played a central role in shifting the treatment landscape of many diseases, including autoimmune disorders and cancers. While showing many therapeutic advantages such as long half-life and highly selective actions, therapeutic antibodies still face many outstanding issues associated with their pharmacokinetics (PK) and pharmacodynamics (PD), including high variabilities, low tissue distributions, poorly-defined PK/PD characteristics for novel antibody formats, and high rates of treatment resistance. We have witnessed many successful cases applying PK/PD modeling to answer critical questions in therapeutic antibodies’ development and regulations. These models have yielded substantial insights into antibody PK/PD properties. This review summarized the progress, challenges, and future directions in modeling antibody PK/PD and highlighted the potential of applying mechanistic models addressing the development questions.
Collapse
|
84
|
Qi T, Cao Y. In Translation: FcRn across the Therapeutic Spectrum. Int J Mol Sci 2021; 22:3048. [PMID: 33802650 PMCID: PMC8002405 DOI: 10.3390/ijms22063048] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
As an essential modulator of IgG disposition, the neonatal Fc receptor (FcRn) governs the pharmacokinetics and functions many therapeutic modalities. In this review, we thoroughly reexamine the hitherto elucidated biological and thermodynamic properties of FcRn to provide context for our assessment of more recent advances, which covers antigen-binding fragment (Fab) determinants of FcRn affinity, transgenic preclinical models, and FcRn targeting as an immune-complex (IC)-clearing strategy. We further comment on therapeutic antibodies authorized for treating SARS-CoV-2 (bamlanivimab, casirivimab, and imdevimab) and evaluate their potential to saturate FcRn-mediated recycling. Finally, we discuss modeling and simulation studies that probe the quantitative relationship between in vivo IgG persistence and in vitro FcRn binding, emphasizing the importance of endosomal transit parameters.
Collapse
Affiliation(s)
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA;
| |
Collapse
|
85
|
Li TR, Chatterjee M, Lala M, Abraham AK, Freshwater T, Jain L, Sinha V, de Alwis DP, Mayawala K. Pivotal Dose of Pembrolizumab: A Dose-Finding Strategy for Immuno-Oncology. Clin Pharmacol Ther 2021; 110:200-209. [PMID: 33462831 DOI: 10.1002/cpt.2170] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022]
Abstract
Despite numerous publications emphasizing the value of dose finding, drug development in oncology is dominated by the mindset that higher dose provides higher efficacy. Examples of dose finding implemented by biopharmaceutical firms can change this mindset. The purpose of this article is to outline a pragmatic dose selection strategy for immuno-oncology (IO) and other targeted monoclonal antibodies (mAbs). The approach was implemented for pembrolizumab. Selecting a recommended phase II dose (RP2D) with a novel mechanism of action is often challenging due to uncertain relationships between pharmacodynamics measurements and clinical end points. Additionally, phase I efficacy and safety data are generally inadequate for RP2D selection for IO mAbs. Here, the RP2D was estimated based on phase I (clinical study KN001 A and A2) pharmacokinetics data as the dose required for target saturation, which represents a surrogate for maximal pharmacological effect for antagonist mAbs. Due to limitations associated with collecting and analyzing tumor biopsies, characterizing intratumoral target engagement (TE) is challenging. To overcome this gap, a physiologically-based pharmacokinetic model was implemented to predict intratumoral TE. As tumors are spatially heterogeneous, TE was predicted in well-vascularized and poorly vascularized tumor regions. Additionally, impact of differences in target expression, for example, due to interindividual variability and cancer type, was simulated. Simulations showed that 200 mg every 3 weeks can achieve ≥ 90% TE in clinically relevant scenarios, resulting in the recommendation of 200 mg every 3 weeks as the RP2D. Randomized dose comparison studies (KN001 B2 and D) showing similar efficacy over a fivefold dose/exposure range confirmed the RP2D as the pivotal dose.
Collapse
Affiliation(s)
- Tommy R Li
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Manash Chatterjee
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Mallika Lala
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Anson K Abraham
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Tomoko Freshwater
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Lokesh Jain
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Vikram Sinha
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Dinesh P de Alwis
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| | - Kapil Mayawala
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc, Kenilworth, New Jersey, USA
| |
Collapse
|
86
|
Atypical Ebola Virus Disease in a Nonhuman Primate following Monoclonal Antibody Treatment Is Associated with Glycoprotein Mutations within the Fusion Loop. mBio 2021; 12:mBio.01438-20. [PMID: 33436428 PMCID: PMC7844533 DOI: 10.1128/mbio.01438-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ebola virus remains a global threat to public health and biosecurity, yet we still know relatively little about its pathogenesis and the complications that arise following recovery. With nearly 20,000 survivors from the 2013–2016 West African outbreak, as well as over 1,000 survivors of the recent outbreak in the DRC, we must consider the consequences of virus persistence and recrudescent disease, even if they are rare. Ebola virus (EBOV) is responsible for numerous devastating outbreaks throughout Africa, including the 2013–2016 West African outbreak as well as the two recent outbreaks in the Democratic Republic of the Congo (DRC), one of which is ongoing. Although EBOV disease (EVD) has typically been considered a highly lethal acute infection, increasing evidence suggests that the virus can persist in certain immune-privileged sites and occasionally lead to EVD recrudescence. Little is understood about the processes that contribute to EBOV persistence and recrudescence, in part because of the rarity of these phenomena but also because of the absence of an animal model that recapitulates them. Here, we describe a case of EBOV persistence associated with atypical EVD in a nonhuman primate (NHP) following inoculation with EBOV and treatment with an experimental monoclonal antibody cocktail. Although this animal exhibited only mild signs of acute EVD, it developed severe disease 2 weeks later and succumbed shortly thereafter. Viremia was undetectable at the time of death, despite abundant levels of viral RNA in most tissues, each of which appeared to harbor a distinct viral quasispecies. Remarkably, sequence analysis identified a single mutation in glycoprotein (GP) that not only resisted antibody-mediated neutralization but also increased viral growth kinetics and virulence. Overall, this report represents the most thoroughly characterized case of atypical EVD in an NHP described thus far, and it provides valuable insight into factors that may contribute to EBOV persistence and recrudescent disease.
Collapse
|
87
|
Chang HY, Wu S, Li Y, Zhang W, Burrell M, Webster CI, Shah DK. Brain pharmacokinetics of anti-transferrin receptor antibody affinity variants in rats determined using microdialysis. MAbs 2021; 13:1874121. [PMID: 33499723 PMCID: PMC7849817 DOI: 10.1080/19420862.2021.1874121] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 01/10/2023] Open
Abstract
Receptor-mediated transcytosis (RMT) is used to enhance the delivery of monoclonal antibodies (mAb) into the central nervous system (CNS). While the binding to endogenous receptors on the brain capillary endothelial cells (BCECs) may facilitate the uptake of mAbs in the brain, a strong affinity for the receptor may hinder the efficiency of transcytosis. To quantitatively investigate the effect of binding affinity on the pharmacokinetics (PK) of anti-transferrin receptor (TfR) mAbs in different regions of the rat brain, we conducted a microdialysis study to directly measure the concentration of free mAbs at different sites of interest. Our results confirmed that bivalent anti-TfR mAb with an optimal dissociation constant (KD) value (76 nM) among four affinity variants can have up to 10-fold higher transcytosed free mAb exposure in the brain interstitial fluid (bISF) compared to lower and higher affinity mAbs (5 and 174 nM). This bell-shaped relationship between KD values and the increased brain exposure of mAbs was also visible when using whole-brain PK data. However, we found that mAb concentrations in postvascular brain supernatant (obtained after capillary depletion) were almost always higher than the concentrations measured in bISF using microdialysis. We also observed that the increase in mAb area under the concentration curve in CSF compartments was less significant, which highlights the challenge in using CSF measurement as a surrogate for estimating the efficiency of RMT delivery. Our results also suggest that the determination of mAb concentrations in the brain using microdialysis may be necessary to accurately measure the PK of CNS-targeted antibodies at the site-of-actions in the brain.
Collapse
Affiliation(s)
- Hsueh-Yuan Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Shengjia Wu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Yingyi Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Wanying Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Matthew Burrell
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Carl I. Webster
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
88
|
The NF-κB/leukemia inhibitory factor/STAT3 signaling pathway in antibody-mediated suppression of Sindbis virus replication in neurons. Proc Natl Acad Sci U S A 2020; 117:29035-29045. [PMID: 33144502 DOI: 10.1073/pnas.2016691117] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Alphaviruses are positive-sense, enveloped RNA viruses that are important causes of viral encephalomyelitis. Sindbis virus (SINV) is the prototype alphavirus and preferentially infects neurons in rodents to induce an encephalomyelitis similar to the human disease. Using a mouse model of SINV infection of the nervous system, many of the immune processes involved in recovery from viral encephalomyelitis have been identified. Antibody specific to the SINV E2 glycoprotein plays an important role in recovery and is sufficient for noncytolytic suppression of virus replication in vivo and in vitro. To investigate the mechanism of anti-E2 antibody-mediated viral suppression, a reverse-phase protein array was used to broadly survey cellular signaling pathway activation following antibody treatment of SINV-infected differentiated AP-7 neuronal cells. Anti-E2 antibody induced rapid transient NF-κB and later sustained Y705 STAT3 phosphorylation, outlining an intracellular signaling cascade activated by antiviral antibody. Because NF-κB target genes include the STAT3-activating IL-6 family cytokines, expression of these messenger RNAS (mRNAs) was assessed. Expression of leukemia inhibitory factor (LIF) cytokine mRNA, but not other IL-6 family member mRNAs, was up-regulated by anti-E2 antibody. LIF induced STAT3 Y705 phosphorylation in infected differentiated AP-7 cells but did not inhibit virus replication. However, anti-E2 antibody localized the LIF receptor to areas of E2 expression on the infected cell surface, and LIF enhanced the antiviral effects of antibody. These findings identify activation of the NF-κB/LIF/STAT3 signaling cascade as involved in inducing antibody-mediated viral suppression and highlight the importance of nonneutralizing antibody functions in viral clearance from neurons.
Collapse
|
89
|
Nicolas A, Dejoux A, Poirier C, Aubrey N, Péan JM, Velge-Roussel F. Contribution of Intrinsic Fluorescence to the Design of a New 3D-Printed Implant for Releasing SDABS. Pharmaceutics 2020; 12:pharmaceutics12100921. [PMID: 32993086 PMCID: PMC7601711 DOI: 10.3390/pharmaceutics12100921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 11/23/2022] Open
Abstract
Single-domain antibodies (sdAbs) offer great features such as increased stability but are hampered by a limited serum half-life. Many strategies have been developed to improve the sdAb half-life, such as protein engineering and controlled release systems (CRS). In our study, we designed a new product that combined a hydrogel with a 3D-printed implant. The results demonstrate the implant’s ability to sustain sdAb release up to 13 days through a reduced initial burst release followed by a continuous release. Furthermore, formulation screening helped to identify the best sdAb formulation conditions and improved our understanding of our CRS. Through the screening step, we gained knowledge about the influence of the choice of polymer and about potential interactions between the sdAb and the polymer. To conclude, this feasibility study confirmed the ability of our CRS to extend sdAb release and established the fundamental role of formulation screening for maximizing knowledge about our CRS.
Collapse
Affiliation(s)
- Alexandre Nicolas
- GICC EA 7501, Faculty of Medicine, University of Tours, 37032 Tours, France;
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Alice Dejoux
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Cécile Poirier
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Nicolas Aubrey
- ISP UMR 1282, INRA, Team BioMAP, University of Tours, 37200 Tours, France;
| | - Jean-Manuel Péan
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Florence Velge-Roussel
- GICC EA 7501, Faculty of Medicine, University of Tours, 37032 Tours, France;
- Correspondence: ; Tel.: +33-(0)2-4736-6058
| |
Collapse
|
90
|
Attwood MM, Jonsson J, Rask-Andersen M, Schiöth HB. Soluble ligands as drug targets. Nat Rev Drug Discov 2020; 19:695-710. [PMID: 32873970 DOI: 10.1038/s41573-020-0078-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
Abstract
Historically, the main classes of drug targets have been receptors, enzymes, ion channels and transporters. However, owing largely to the rise of antibody-based therapies in the past two decades, soluble protein ligands such as inflammatory cytokines have become an increasingly important class of drug targets. In this Review, we analyse drugs targeting ligands that have reached clinical development at some point since 1992. We identify 291 drugs that target 99 unique ligands, and we discuss trends in the characteristics of the ligands, drugs and indications for which they have been tested. In the last 5 years, the number of ligand-targeting drugs approved by the FDA has doubled to 34, while the number of clinically validated ligand targets has doubled to 22. Cytokines and growth factors are the predominant types of targeted ligands (70%), and inflammation and autoimmune disorders, cancer and ophthalmological diseases are the top therapeutic areas for both approved agents and agents in clinical studies, reflecting the central role of cytokine and/or growth factor pathways in such diseases.
Collapse
Affiliation(s)
- Misty M Attwood
- Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Jörgen Jonsson
- Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Mathias Rask-Andersen
- Medical Genetics and Genomics, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Functional Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden. .,Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
91
|
Tran VL, Novell A, Tournier N, Gerstenmayer M, Schweitzer-Chaput A, Mateos C, Jego B, Bouleau A, Nozach H, Winkeler A, Kuhnast B, Larrat B, Truillet C. Impact of blood-brain barrier permeabilization induced by ultrasound associated to microbubbles on the brain delivery and kinetics of cetuximab: An immunoPET study using 89Zr-cetuximab. J Control Release 2020; 328:304-312. [PMID: 32860928 DOI: 10.1016/j.jconrel.2020.08.047] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/22/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022]
Abstract
Epidermal growth factor receptor (EGFR), involved in cell proliferation and migration, is overexpressed in ~50% of glioblastomas. Anti-EGFR based strategies using monoclonal antibodies (mAb) such as cetuximab (CTX) have been proposed for central nervous system (CNS) cancer therapy. However, the blood-brain barrier (BBB) drastically restricts their brain penetration which limits their efficacy for the treatment of glioblastomas. Herein, a longitudinal PET imaging study was performed to assess the relevance and the impact of focused ultrasound (FUS)-mediated BBB permeabilization on the brain exposure to the anti-EGFR mAb CTX over time. For this purpose, FUS permeabilization process with microbubbles was applied on intact BBB mouse brain before the injection of 89Zr-labeled CTX for longitudinal imaging monitoring. FUS induced a dramatic increase in mAb penetration to the brain, 2 times higher compared to the intact BBB. The transfer of 89Zr-CTX from blood to the brain was rendered significant by FUS (kuptake = 1.3 ± 0.23 min-1 with FUS versus kuptake = 0 ± 0.006 min-1 without FUS). FUS allowed significant and prolonged exposure to mAb in the brain parenchyma. This study confirms the potential of FUS as a target delivery method for mAb in CNS.
Collapse
Affiliation(s)
- Vu Long Tran
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | - Nicolas Tournier
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | | | | | - Claudia Mateos
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | - Benoit Jego
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | - Alizée Bouleau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | - Hervé Nozach
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-Sur-Yvette, France
| | - Alexandra Winkeler
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | - Bertrand Kuhnast
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Gif sur Yvette 91191, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay 91401, France.
| |
Collapse
|
92
|
Kuchimanchi M, Monine M, Kandadi Muralidharan K, Woodward C, Penner N. Phase II Dose Selection for Alpha Synuclein-Targeting Antibody Cinpanemab (BIIB054) Based on Target Protein Binding Levels in the Brain. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:515-522. [PMID: 32613752 PMCID: PMC7499191 DOI: 10.1002/psp4.12538] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/20/2020] [Indexed: 12/27/2022]
Abstract
This modeling and simulation analysis was aimed at selecting doses of cinpanemab (BIIB054), a monoclonal antibody targeting aggregated α‐synuclein, for a phase II study in Parkinson’s disease (PD). Doses and regimens were proposed based on anticipated target concentration in brain interstitial fluid (ISF); in vitro/in vivo data on the affinity of monoclonal antibodies to the target protein; and safety, tolerability, and pharmacokinetic data (1–135 mg/kg intravenous administration) from a phase I single ascending dose (SAD) study. A population pharmacokinetic modeling approach was used to select intravenous doses of 250, 1,250, and 3,500 mg every 4 weeks, to maintain 50%, 90%, and > 90% of target binding in ISF of PD participants. A favorable safety profile from the SAD study—which showed that cinpanemab was generally well‐tolerated at doses up to 90 mg/kg, supported by modeling and simulations of the anticipated safety margins—allowed implementation of a fixed‐dose approach.
Collapse
Affiliation(s)
- Mita Kuchimanchi
- Clinical Pharmacology & Pharmacometrics, Biogen Inc., Cambridge, Massachusetts, USA
| | - Michael Monine
- Clinical Pharmacology & Pharmacometrics, Biogen Inc., Cambridge, Massachusetts, USA
| | | | - Caroline Woodward
- Clinical Pharmacology & Pharmacometrics, Biogen Inc., Cambridge, Massachusetts, USA
| | - Natalia Penner
- Clinical Pharmacology & Pharmacometrics, Biogen Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
93
|
Chua CYX, Ho J, Susnjar A, Lolli G, Di Trani N, Pesaresi F, Zhang M, Nance E, Grattoni A. Intratumoral Nanofluidic System for Enhancing Tumor Biodistribution of Agonist CD40 Antibody. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000055] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Jeremy Ho
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Weill Cornell Medical College New York NY 10065 USA
| | - Antonia Susnjar
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Graziano Lolli
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Department of Mechanical and Aerospace Engineering Polytechnic of Turin Turin 10129 Italy
| | - Nicola Di Trani
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- University of Chinese Academy of Science (UCAS) Shijingshan, 19 Yuquan Road Beijing 100049 China
| | - Federica Pesaresi
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Department of Electronics and Telecommunications Polytechnic of Turin Turin 10129 Italy
| | - Mengying Zhang
- Department of Chemical Engineering University of Washington Seattle WA 98195 USA
| | - Elizabeth Nance
- Department of Chemical Engineering University of Washington Seattle WA 98195 USA
| | - Alessandro Grattoni
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Department of Surgery Houston Methodist Hospital Houston TX 77030 USA
- Department of Radiation Oncology Houston Methodist Hospital Houston TX 77030 USA
| |
Collapse
|
94
|
Chang HP, Kim SJ, Shah DK. Whole-Body Pharmacokinetics of Antibody in Mice Determined using Enzyme-Linked Immunosorbent Assay and Derivation of Tissue Interstitial Concentrations. J Pharm Sci 2020; 110:446-457. [PMID: 32502472 DOI: 10.1016/j.xphs.2020.05.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/06/2020] [Accepted: 05/20/2020] [Indexed: 12/20/2022]
Abstract
Here we have reported whole-body disposition of wild-type IgG and FcRn non-binding IgG in mice, determined using Enzyme-Linked Immunosorbent Assay (ELISA). The disposition data generated using ELISA are compared with previously published biodistribution data generated using radiolabelled IgG. In addition, we introduce a novel concept of ABCIS values, which are defined as percentage ratios of tissue interstitial and plasma AUC values. These values can help in predicting tissue interstitial concentrations of monoclonal antibodies (mAbs) based on the plasma concentrations. Tissue interstitial concentrations derived from our study are also compared with previously reported values measured using microdialysis or centrifugation method. Lastly, the new set of biodistribution data generated using ELISA are used to refine the PBPK model for mAbs.
Collapse
Affiliation(s)
- Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Se Jin Kim
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
95
|
Shulgin B, Kosinsky Y, Omelchenko A, Chu L, Mugundu G, Aksenov S, Pimentel R, DeYulia G, Kim G, Peskov K, Helmlinger G. Dose dependence of treatment-related adverse events for immune checkpoint inhibitor therapies: a model-based meta-analysis. Oncoimmunology 2020; 9:1748982. [PMID: 32934874 PMCID: PMC7466858 DOI: 10.1080/2162402x.2020.1748982] [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: 08/29/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death-1 (PD-1) and/or cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint inhibitor (ICI) treatments are associated with adverse events (AEs), which may be dependent on ICI dose. Applying a model-based meta-analysis to evaluate safety data from published clinical trials from 2005 to 2018, we analyzed the dose/exposure dependence of ICI treatment-related AE (trAE) and immune-mediated AE (imAE) rates. Unlike with PD-1 inhibitor monotherapy, CTLA-4 inhibitor monotherapy exhibited a dose/exposure dependence on most AE types evaluated. Furthermore, combination therapy with PD-1 inhibitor significantly strengthened the dependence of trAE and imAE rates on CTLA-4 inhibitor dose/exposure.
Collapse
Affiliation(s)
| | | | | | - Lulu Chu
- PK Sciences Modeling & Simulation, Novartis Institutes of BioMedical Research, Cambridge, MA, USA
| | - Ganesh Mugundu
- Clinical Pharmacology & Quantitative Pharmacology, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, USA
| | - Sergey Aksenov
- Clinical Pharmacology & Quantitative Pharmacology, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, USA
| | | | | | | | - Kirill Peskov
- M&S Decisions LLC, Moscow, Russia
- I.M.Sechenov first Moscow State Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Gabriel Helmlinger
- Clinical Pharmacology & Toxicology, Obsidian Therapeutics, Cambridge, MA, USA
| |
Collapse
|
96
|
Conner KP, Devanaboyina SC, Thomas VA, Rock DA. The biodistribution of therapeutic proteins: Mechanism, implications for pharmacokinetics, and methods of evaluation. Pharmacol Ther 2020; 212:107574. [PMID: 32433985 DOI: 10.1016/j.pharmthera.2020.107574] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/30/2020] [Indexed: 02/08/2023]
Abstract
Therapeutic proteins (TPs) are a diverse drug class that include monoclonal antibodies (mAbs), recombinantly expressed enzymes, hormones and growth factors, cytokines (e.g. chemokines, interleukins, interferons), as well as a wide range of engineered fusion scaffolds containing IgG1 Fc domain for half-life extension. As the pharmaceutical industry advances more potent and selective protein-based medicines through discovery and into the clinical stages of development, it has become widely appreciated that a comprehensive understanding of the mechanisms of TP biodistribution can aid this endeavor. This review aims to highlight the literature that has advanced our understanding of the determinants of TP biodistribution. A particular emphasis is placed on the multi-faceted role of the neonatal Fc receptor (FcRn) in mAb and Fc-fusion protein disposition. In addition, characterization of the TP-target interaction at the cell-level is discussed as an essential strategy to establish pharmacokinetic-pharmacodynamic (PK/PD) relationships that may lead to more informed human dose projections during clinical development. Methods for incorporation of tissue and cell-level parameters defining these characteristics into higher-order mechanistic and semi-mechanistic PK models will also be presented.
Collapse
Affiliation(s)
- Kip P Conner
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| | - Siva Charan Devanaboyina
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| | - Veena A Thomas
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| | - Dan A Rock
- Dept. of Pharmacokinetics and Drug Metabolism, Amgen Inc, 1120 Veterans Blvd, South San Francisco, CA 94080, USA.
| |
Collapse
|
97
|
Castanha PMS, Erdos G, Watkins SC, Falo LD, Marques ETA, Barratt-Boyes SM. Reciprocal immune enhancement of dengue and Zika virus infection in human skin. JCI Insight 2020; 5:133653. [PMID: 31910161 DOI: 10.1172/jci.insight.133653] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) are closely related mosquito-borne flaviviruses that co-circulate in tropical regions and constitute major threats to global human health. Whether preexisting immunity to one virus affects disease caused by the other during primary or secondary infections is unknown but is critical in preparing for future outbreaks and predicting vaccine safety. Using a human skin explant model, we show that DENV-3 immune sera increased recruitment and infection of Langerhans cells, macrophages, and dermal dendritic cells following inoculation with DENV-2 or ZIKV. Similarly, ZIKV immune sera enhanced infection with DENV-2. Immune sera increased migration of infected Langerhans cells to the dermis and emigration of infected cells out of skin. Heterotypic immune sera increased viral RNA in the dermis almost 10-fold and reduced the amount of virus required to infect a majority of myeloid cells by 100- to 1000-fold. Enhancement was associated with cross-reactive IgG and induction of IL-10 expression and was mediated by both CD32 and CD64 Fcγ receptors. These findings reveal that preexisting heterotypic immunity greatly enhances DENV and ZIKV infection, replication, and spread in human skin. This relevant tissue model will be valuable in assessing the efficacy and risk of dengue and Zika vaccines in humans.
Collapse
Affiliation(s)
- Priscila M S Castanha
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Biological Science Institute and Faculty of Medical Science, University of Pernambuco, Recife, Brazil.,Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | - Simon C Watkins
- Center for Biologic Imaging.,Department of Cell Biology, and.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Ernesto T A Marques
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Simon M Barratt-Boyes
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
98
|
Fogh JR, Jacobsen AM, Nguyen TTTN, Rand KD, Olsen LR. Investigating surrogate cerebrospinal fluid matrix compositions for use in quantitative LC-MS analysis of therapeutic antibodies in the cerebrospinal fluid. Anal Bioanal Chem 2020; 412:1653-1661. [PMID: 32008082 PMCID: PMC7026242 DOI: 10.1007/s00216-020-02403-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/17/2019] [Accepted: 01/10/2020] [Indexed: 11/29/2022]
Abstract
As quantitative analysis of biotherapeutics in cerebrospinal fluid (CSF) with LC-MS becomes increasingly widespread, there is a need for method developments towards higher sensitivity. By using artificial CSF (aCSF) in the development phase, the consumption of costly and sparsely available CSF can be limited. The aCSF compositions tested here were made from various dilutions of bovine serum albumin (BSA) or rat plasma to mimic the total protein concentration found in CSF. Focusing on monoclonal antibodies, the aCSF was spiked with human immunoglobulin (hIgG) and prepared with the bottom-up analysis technique using LC-MS. Assuming that the composition of the aCSF would affect the digest, the response from aCSF matrices was compared with CSF from rat, monkey, and dog in terms of estimated sample concentration and matrix effects. The samples were spiked with hIgG in the range of 10 to 1000 ng/mL and volumes of 10 μL were transferred to sample preparation. The results indicate that BSA dilutions from 300 to 2000 μg/mL and rat plasma dilutions of 0.5–2% provide the most accurate concentration estimates when compared with rat CSF. 1000 μg/mL BSA did not produce significantly different concentration estimates for 500 ng/mL samples when compared with CSF from rat, monkey, and dog, and can therefore be used as aCSF for several different species.
Collapse
Affiliation(s)
- Jens Rose Fogh
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2200, Copenhagen, Denmark.,Translational DMPK, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| | | | - Tam T T N Nguyen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2200, Copenhagen, Denmark
| | - Kasper D Rand
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2200, Copenhagen, Denmark
| | - Line Rørbæk Olsen
- Translational DMPK, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark.
| |
Collapse
|
99
|
Pharmacokinetic Characterization and Tissue Distribution of Fusion Protein Therapeutics by Orthogonal Bioanalytical Assays and Minimal PBPK Modeling. Molecules 2020; 25:molecules25030535. [PMID: 31991858 PMCID: PMC7037219 DOI: 10.3390/molecules25030535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/15/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Characterization of pharmacokinetic (PK) properties and target tissue distribution of therapeutic fusion proteins (TFPs) are critical in supporting in vivo efficacy. We evaluated the pharmacokinetic profile of an investigational TFP consisting of human immunoglobulin G4 fused to the modified interferon alpha by orthogonal bioanalytical assays and applied minimal physiologically based pharmacokinetic (PBPK) modeling to characterize the TFP pharmacokinetics in mouse. The conventional ligand binding assay (LBA), immunocapture-liquid chromatography/tandem mass spectrometry (IC-LC/MS) detecting the human IgG4 peptide or the interferon alpha peptide were developed to measure the TFP concentrations in mouse plasma and tumor. The minimal PBPK model incorporated a tumor compartment model was used for data fitting. The plasma clearance measured by LBA and IC-LC/MS was comparable in the range of 0.5–0.6 mL/h/kg. However, the tumor exposure measured by the generic human IgG4 IC-LC/MS was significantly underestimated compared with the interferon alpha specific IC-LC/MS and LBA. Furthermore, the minimal PBPK model simultaneously captured the relationship between plasma and tissue exposure. We proposed the streamlined practical strategy to characterize the plasma exposure and tumor distribution of a TFP by both LBA and IC-LC/MS. The minimal PBPK modeling was established for better understanding of pharmacokinetic profile of investigational TFPs in the biotherapeutic discovery.
Collapse
|
100
|
Antibody Conjugates-Recent Advances and Future Innovations. Antibodies (Basel) 2020; 9:antib9010002. [PMID: 31936270 PMCID: PMC7148502 DOI: 10.3390/antib9010002] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/18/2022] Open
Abstract
Monoclonal antibodies have evolved from research tools to powerful therapeutics in the past 30 years. Clinical success rates of antibodies have exceeded expectations, resulting in heavy investment in biologics discovery and development in addition to traditional small molecules across the industry. However, protein therapeutics cannot drug targets intracellularly and are limited to soluble and cell-surface antigens. Tremendous strides have been made in antibody discovery, protein engineering, formulation, and delivery devices. These advances continue to push the boundaries of biologics to enable antibody conjugates to take advantage of the target specificity and long half-life from an antibody, while delivering highly potent small molecule drugs. While the "magic bullet" concept produced the first wave of antibody conjugates, these entities were met with limited clinical success. This review summarizes the advances and challenges in the field to date with emphasis on antibody conjugation, linker-payload chemistry, novel payload classes, absorption, distribution, metabolism, and excretion (ADME), and product developability. We discuss lessons learned in the development of oncology antibody conjugates and look towards future innovations enabling other therapeutic indications.
Collapse
|