1
|
Peyvandi F, Seidizadeh O, Mohsenian S, Garagiola I. Exploring nonreplacement therapies' impact on hemophilia and other rare bleeding disorders. Res Pract Thromb Haemost 2024; 8:102434. [PMID: 38873363 PMCID: PMC11169453 DOI: 10.1016/j.rpth.2024.102434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 06/15/2024] Open
Abstract
The management of hemophilia, von Willebrand disease (VWD), and rare coagulation disorders traditionally relied on replacement therapies, such as factor concentrates, to address clotting factor deficiencies. However, in recent years, the emergence of nonreplacement therapies has shown promise as an adjunctive approach, especially in hemophilia, and also for patients with VWD and rare bleeding disorders. This review article offers an overview of nonreplacement therapies, such as FVIII-mimicking agents and drugs aimed at rebalancing hemostasis by inhibiting natural anticoagulants, particularly in the management of hemophilia. The utilization of nonreplacement therapies in VWD and rare bleeding disorders has recently attracted attention, as evidenced by presentations at the International Society on Thrombosis and Haemostasis 2023 Congress. Nonreplacement therapies provide alternative methods for preventing bleeding episodes and enhancing patients' quality of life, as many of them are administered subcutaneously and allow longer infusion intervals, resulting in improved quality of life and comfort for patients.
Collapse
Affiliation(s)
- Flora Peyvandi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Angelo Bianchi Bonomi Hemophilia and Thrombosis Center and Fondazione Luigi Villa, Milan, Italy
- Università degli Studi di Milano, Department of Pathophysiology and Transplantation, Milan, Italy
| | - Omid Seidizadeh
- Università degli Studi di Milano, Department of Pathophysiology and Transplantation, Milan, Italy
| | - Samin Mohsenian
- Università degli Studi di Milano, Department of Pathophysiology and Transplantation, Milan, Italy
| | - Isabella Garagiola
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Angelo Bianchi Bonomi Hemophilia and Thrombosis Center and Fondazione Luigi Villa, Milan, Italy
| |
Collapse
|
2
|
Miesbach W, von Drygalski A, Smith C, Sivamurthy K, Pinachyan K, Bensen-Kennedy D, Drelich D, Kulkarni R. The current challenges faced by people with hemophilia B. Eur J Haematol 2024; 112:339-349. [PMID: 38082533 DOI: 10.1111/ejh.14135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 02/17/2024]
Abstract
Hemophilia B (HB) is a rare, hereditary disease caused by a defect in the gene encoding factor IX (FIX) and leads to varying degrees of coagulation deficiency. The prevailing treatment for people with HB (PWHB) is FIX replacement product. The advent of recombinant coagulation products ushered in a new era of safety, efficacy, and improved availability compared with plasma-derived products. For people with severe HB, lifelong prophylaxis with a FIX replacement product is standard of care. Development of extended half-life FIX replacement products has allowed for advancements in the care of these PWHB. Nonetheless, lifelong need for periodic dosing and complex surveillance protocols pose substantive challenges in terms of access, adherence, and healthcare resource utilization. Further, some PWHB on prophylactic regimens continue to experience breakthrough bleeds and joint damage, and subpopulations of PWHB, including women, those with mild-to-moderate HB, and those with inhibitors to FIX, experience additional unique difficulties. This review summarizes the current challenges faced by PWHB, including the unique subpopulations; identifying the need for improved awareness, personalized care strategies, and new therapeutic options for severe HB, which may provide future solutions for some of the remaining unmet needs of PWHB.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Roshni Kulkarni
- Michigan State University Center for Bleeding and Clotting Disorders, Lansing, Michigan, USA
| |
Collapse
|
3
|
Guzzardo GM, Sidonio R, Callaghan MU, Regling K. Early stage clinical trials for the treatment of hemophilia A. Expert Opin Investig Drugs 2022; 31:1169-1186. [PMID: 36265129 DOI: 10.1080/13543784.2022.2138742] [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] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Hemophilia A is a severe bleeding disorder affecting about 1 in 5,000 males. The gold standard for prophylaxis and treatment of acute bleeding has been factor (F) VIII concentrate. A multitude of treatment modalities are now available and under clinical investigation. AREAS COVERED This review discusses ongoing/recently completed early-phase clinical trials registered on ClinicalTrials.gov in patients with hemophilia A through April 2022. These new pipeline therapies are focused on addressing the safety and efficacy of new factor-related products, non-factor related products, and gene therapy options for hemophilia. EXPERT OPINION Current standard of care effectively prevents and treats acute bleeding and has significantly improved the quality of life in hemophilia. The biggest challenges in the improvement of care are treatment-related burden and the burden of cost in developing countries. New drugs under development are likely to enter practice by the end of this decade and address many of the unmet needs particularly of those with severe disease. Data is limited in unique populations (e.g. congenital/inherited FVIII inhibitors, non-severe hemophilia A, women/girls with hemophilia and children) which are important areas for future research; additional clinical trials and long-term outcome data are necessary prior to incorporating these new therapies in our treatment arsenal.
Collapse
Affiliation(s)
- Gianna M Guzzardo
- Pediatric Hematology Oncology, Children's Hospital of Michigan, Detroit, MI, USA
| | - Robert Sidonio
- Pediatric Hematology Oncology, Emory University and Aflac Cancer and Blood Disorders, Atlanta, GA, USA
| | - Michael U Callaghan
- Agios Pharmaceuticals, Cambridge, MA, USA.,Department of Pediatrics, Central Michigan University School of Medicine, Mount Pleasant, MI, USA
| | - Katherine Regling
- Pediatric Hematology Oncology, Children's Hospital of Michigan, Detroit, MI, USA.,Department of Pediatrics, Central Michigan University School of Medicine, Mount Pleasant, MI, USA
| |
Collapse
|
4
|
Pasquiers B, Benamara S, Felices M, Nguyen L, Declèves X. Review of the Existing Translational Pharmacokinetics Modeling Approaches Specific to Monoclonal Antibodies (mAbs) to Support the First-In-Human (FIH) Dose Selection. Int J Mol Sci 2022; 23:12754. [PMID: 36361546 PMCID: PMC9657028 DOI: 10.3390/ijms232112754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 08/27/2023] Open
Abstract
The interest in therapeutic monoclonal antibodies (mAbs) has continuously growing in several diseases. However, their pharmacokinetics (PK) is complex due to their target-mediated drug disposition (TMDD) profiles which can induce a non-linear PK. This point is particularly challenging during the pre-clinical and translational development of a new mAb. This article reviews and describes the existing PK modeling approaches used to translate the mAbs PK from animal to human for intravenous (IV) and subcutaneous (SC) administration routes. Several approaches are presented, from the most empirical models to full physiologically based pharmacokinetic (PBPK) models, with a focus on the population PK methods (compartmental and minimal PBPK models). They include the translational approaches for the linear part of the PK and the TMDD mechanism of mAbs. The objective of this article is to provide an up-to-date overview and future perspectives of the translational PK approaches for mAbs during a model-informed drug development (MIDD), since the field of PK modeling has gained recently significant interest for guiding mAbs drug development.
Collapse
Affiliation(s)
- Blaise Pasquiers
- PhinC Development, 91300 Massy, France
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| | | | | | | | - Xavier Declèves
- Université Paris Cité, Inserm UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France
| |
Collapse
|
5
|
Konishi K, Nakamura K, Hanada Y, Kitanaga Y, Kubo S, Kinugasa F, Yamajuku D, Maeda M, Yamamoto N, Minematsu T, Ohbuchi M, Kondo Y, Sumida T. Preclinical Characterization of ASP2713, a Novel Igβ and FcγRIIB Cross-Linking Antibody, for Prediction of Human Pharmacokinetics and Clinically Effective Dose. J Pharm Sci 2022; 111:2630-2638. [PMID: 35700799 DOI: 10.1016/j.xphs.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/24/2022]
Abstract
Previously, we reported the fundamental pharmacological characteristics of a novel Igβ and Fc gamma receptor IIB cross-linking antibody, ASP2713, as a new treatment option for systemic lupus erythematosus. The aims of the present study were to investigate ASP2713's characteristics with regard to pharmacological effect, pharmacokinetics (PK), and receptor occupancy, and to predict its human PK and clinically effective dose. The relationship between the concentration and receptor occupancy of ASP2713 for Igβ of B cell receptors was examined using whole blood B cells. Calculated EC50 values in cynomolgus monkeys and healthy volunteers were 0.35 and 0.058 μg/mL, respectively. Dose-dependent inhibition of anti-tetanus toxoid (TTx) antibody production, PK, and receptor occupancy of ASP2713 in TTx-sensitized cynomolgus monkeys suggested a minimally effective dose of 1 mg/kg by single intravenous (IV) administration. Scaling-up of monkey PK parameters to humans by allometric scaling predicted a clinically effective dose of 0.4 mg/kg IV administration at 4-week intervals to maintain a trough concentration in humans which achieved the same receptor occupancy expected at the effective dose in monkeys. This study aids in understanding the characteristics of ASP2713 and can be used as a basis for clinical dose setting.
Collapse
Affiliation(s)
- Kentaro Konishi
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan.
| | - Koji Nakamura
- Discovery Accelerator, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Yuichi Hanada
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Yukihiro Kitanaga
- Discovery Accelerator, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Satoshi Kubo
- Discovery Accelerator, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Fumitaka Kinugasa
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Daisuke Yamajuku
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Masashi Maeda
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Nobuchika Yamamoto
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | | | - Masato Ohbuchi
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Yuya Kondo
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
6
|
Haraya K, Tsutsui H, Komori Y, Tachibana T. Recent Advances in Translational Pharmacokinetics and Pharmacodynamics Prediction of Therapeutic Antibodies Using Modeling and Simulation. Pharmaceuticals (Basel) 2022; 15:ph15050508. [PMID: 35631335 PMCID: PMC9145563 DOI: 10.3390/ph15050508] [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: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) have been a promising therapeutic approach for several diseases and a wide variety of mAbs are being evaluated in clinical trials. To accelerate clinical development and improve the probability of success, pharmacokinetics and pharmacodynamics (PKPD) in humans must be predicted before clinical trials can begin. Traditionally, empirical-approach-based PKPD prediction has been applied for a long time. Recently, modeling and simulation (M&S) methods have also become valuable for quantitatively predicting PKPD in humans. Although several models (e.g., the compartment model, Michaelis–Menten model, target-mediated drug disposition model, and physiologically based pharmacokinetic model) have been established and used to predict the PKPD of mAbs in humans, more complex mechanistic models, such as the quantitative systemics pharmacology model, have been recently developed. This review summarizes the recent advances and future direction of M&S-based approaches to the quantitative prediction of human PKPD for mAbs.
Collapse
Affiliation(s)
- Kenta Haraya
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
- Correspondence:
| | - Haruka Tsutsui
- Discovery Biologics Department, Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan;
| | - Yasunori Komori
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| | - Tatsuhiko Tachibana
- Pharmaceutical Science Department, Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba 412-8513, Japan; (Y.K.); (T.T.)
| |
Collapse
|
7
|
Pittman DD, Rakhe S, Bowley SR, Jasuja R, Barakat A, Murphy JE. Hemostatic efficacy of marstacimab alone or in combination with bypassing agents in hemophilia plasmas and a mouse bleeding model. Res Pract Thromb Haemost 2022; 6:e12679. [PMID: 35316941 PMCID: PMC8925002 DOI: 10.1002/rth2.12679] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/07/2022] [Indexed: 11/06/2022] Open
Abstract
Background Patients with hemophilia have deficiencies in intrinsic coagulation factors and can develop inhibitors that limit the effectiveness of replacement coagulation factors. Marstacimab, a human monoclonal antibody, binds and inhibits the human tissue factor pathway inhibitor. Marstacimab is currently under development as a potential prophylactic treatment to prevent bleeding episodes in patients with hemophilia A and B. Objective To assess the effects of marstacimab alone or in combination with the bypassing agent recombinant factor FVIIa (rFVIIa) or activated prothrombin complex concentrate (aPCC) on thrombin generation and bleeding. Methods Marstacimab and/or rFVIIa or aPCC were added to hemophilic A or B plasma or nonhemophilic plasma in vitro. Hemostatic activity was measured using the thrombin generation assay. In vivo effects were assessed using a mouse acute bleeding model. Male hemophilia A mice were dosed with marstacimab plus aPCC before tail clip; blood loss was quantified by measuring hemoglobin. Results Marstacimab plus rFVIIa or aPCC slightly increased peak thrombin levels compared with either agent alone. This increase was within the reported range for nonhemophilic plasma and did not exceed levels observed in nonhemophilic plasma treated with marstacimab alone. Hemophilia A mice that received 200 U/kg aPCC had significantly reduced bleeding (62%) compared with vehicle-treated mice (p < 0.05), and marstacimab plus aPCC reduced bleeding by 83.3% compared with vehicle (p= 0.0009). Conclusions Marstacimab alone or with bypassing agents increased hemostasis in hemophilia plasma without generating excessive thrombin. The hemostatic activity of marstacimab plus aPCC was confirmed in hemophilia A mice.
Collapse
Affiliation(s)
| | - Swapnil Rakhe
- Rare Disease Research UnitPfizer Inc.CambridgeMassachusettsUSA
| | | | - Reema Jasuja
- Rare Disease Research UnitPfizer Inc.CambridgeMassachusettsUSA
| | - Amey Barakat
- Rare Disease Research UnitPfizer Inc.CambridgeMassachusettsUSA
| | - John E. Murphy
- Rare Disease Research UnitPfizer Inc.CambridgeMassachusettsUSA
| |
Collapse
|
8
|
Kwak EY, Kim MJ, Park JH, Jung HW, Jung ME. Target-mediated drug disposition modeling of an anti-TFPI antibody (MG1113) in cynomolgus monkeys to predict human pharmacokinetics and pharmacodynamics. J Thromb Haemost 2021; 19:1425-1435. [PMID: 33448093 DOI: 10.1111/jth.15244] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND MG1113 is a human monoclonal antibody of tissue factor pathway inhibitor (TFPI) under development for prophylaxis for hemophilia patients with or without inhibitors against factor VIII products, which have been used for the treatment of hemophilia. Because TFPI is a negative regulator in the extrinsic coagulation pathway, neutralization of TFPI function by MG1113 can potentially increase coagulation activity by bypassing the intrinsic coagulation pathway, which factor VIII activates. OBJECTIVES This study aims to determine the correlation between pharmacokinetics (PK) and pharmacodynamics (PD) after administering MG1113 to monkeys and to predict the PK and PD of MG1113 in humans by the Target-Mediated Drug Disposition (TMDD) model using the results from monkeys. METHODS The PK profile of MG1113 and the PD effect on the free TFPI level were evaluated after intravenous (IV) and subcutaneous (SC) administrations of MG1113 (2.5, 5, and 10 mg/kg) to male cynomolgus monkeys. After setting up the PK/PD model on monkeys, PK parameters on humans were calculated using allometric scaling, and then clinically effective doses were predicted applying the TMDD model. RESULTS AND CONCLUSIONS MG1113 showed nonlinear PK after both IV and SC administrations at the dosing range from 2.5 to10 mg/kg. The concentrations of MG1113 versus TFPI could be characterized a dose-response relationship using a TMDD model. The TMDD modeling and simulation built in this study were used to simulate various dosage regimens of MG1113 to apply to the first-in-human study design, and moreover expected to be referred to establish the dose for further clinical trials.
Collapse
Affiliation(s)
- Eun-Young Kwak
- Department of GC Pharma R&D Center, GC Pharma, Gyeonggi-do, Korea
| | - Min Ju Kim
- Department of GC Pharma R&D Center, GC Pharma, Gyeonggi-do, Korea
| | - Jin Hyun Park
- Department of GC Pharma R&D Center, GC Pharma, Gyeonggi-do, Korea
| | - Ha Wook Jung
- Department of GC Pharma R&D Center, GC Pharma, Gyeonggi-do, Korea
| | | |
Collapse
|
9
|
Mahlangu JN. Progress in the Development of Anti-tissue Factor Pathway Inhibitors for Haemophilia Management. Front Med (Lausanne) 2021; 8:670526. [PMID: 34026796 PMCID: PMC8131856 DOI: 10.3389/fmed.2021.670526] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/24/2021] [Indexed: 12/28/2022] Open
Abstract
The unprecedented progress in addressing unmet needs in haemophilia care to date includes developing several novel therapies that rebalance haemostasis by restoring thrombin generation in patients with haemophilia A or B with and without inhibitors. These novel therapies are FVIII mimetics, antithrombin interference RNA therapy and several monoclonal antibodies directed against the tissue factor pathway inhibitor (anti-TFPI). In this review, we provide an update on the progress made in developing anti-TFPI therapie. Phase 1 data from the three anti-TFPI studies showed acceptable safety profiles, and currently, available phase 2 data are encouraging. While these data support these molecules' further development progression, there is uncertainty on several aspects of their evolution. Two of the three anti-TFPIs have shown drug-related thrombosis, with one study consequently terminated. None of the thrombotic events is predictable with current monitoring tools, and none correlate with known coagulation parameters. All three anti-TFPIs undergo target mediated drug disposition, which impacts the formulation of dosing regimen fo these therapies. They would require more frequent dosing than some of the extended half-life clotting factor products and antithrombin RNAi therapy. There is no assay to measure the TFPI as the physiological levels are very low, which makes monitoring the impact of the anti-TFPI a challenge. The anti-TFPIs have several advantages, including their bioavailability when administered subcutaneously, their stable pharmacokinetics and their ability to prevent bleeds in haemophilia A or B patients with and without inhibitors. Whether these advantages can be realized will depend on the outcome of the currently ongoing studies.
Collapse
Affiliation(s)
- Johnny N. Mahlangu
- Haemophilia Comprehensive Care Centre, Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand and National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
10
|
Zhao Y, Weyand AC, Shavit JA. Novel treatments for hemophilia through rebalancing of the coagulation cascade. Pediatr Blood Cancer 2021; 68:e28934. [PMID: 33577709 PMCID: PMC8751759 DOI: 10.1002/pbc.28934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/19/2020] [Accepted: 01/12/2021] [Indexed: 11/11/2022]
Abstract
Hemophilia A and B are inherited hemorrhagic disorders that result from alterations in the coagulation cascade. Aside from spontaneous bleeding, the main complication of hemophilia is hemarthrosis. Progress over the last three decades, specifically prophylaxis using recombinant factor, has prevented hemarthrosis and lengthened patient life expectancies. However, many treatments require frequent dosing up to three times a week, and alloantibodies (inhibitors) against replacement factor continues to be an issue. These problems call for novel treatments for patients with hemophilia. Although there has been progress in extended half-life factors and mimetics of factor VIII, an alternative treatment methodology is to rebalance the activities of pro- and anticoagulant factors through inhibition of the natural anticoagulants: antithrombin, tissue factor pathway inhibitor, protein C, and protein S. This review will explore the efficacy of targeting these inhibitory pathways from preclinical development through clinical trials, and delve into concerns of thrombotic risk.
Collapse
Affiliation(s)
- Yakun Zhao
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Angela C. Weyand
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Jordan A. Shavit
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| |
Collapse
|
11
|
Martin EJ, Nolte ME, Kuhn J, Schmidt N, Pfaff N, Brophy DF. An in vitro pharmacodynamic spiking study of befovacimab, a tissue factor pathway inhibitor monoclonal antibody, in blood samples from patients with severe FVIII deficiency. Haemophilia 2021; 27:690-698. [PMID: 33915599 DOI: 10.1111/hae.14314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Tissue factor pathway inhibitor (TFPI) is an endogenous protein that inhibits the extrinsic (tissue factor) pathway and negatively regulates thrombin production during coagulation. Inhibiting TFPI may become a useful target for haemophilia drug development to allow greater thrombin generation without use of the intrinsic (contact) pathway. AIMS The in vitro effects of befovacimab, a humanized TFPI neutralizing antibody, were studied in whole blood and plasma samples from patients with severe FVIII deficiency. METHODS Blood and plasma obtained from participants was supplemented in vitro with befovacimab (0.5, 1, 5, 10 and 100 nM) or recombinant factor VIII (rFVIII) 5-, 10- and 40% and analysed using rotational thromboelastometry (ROTEM), thrombin generation assay (TGA) and the dilute prothrombin time (dPT) assay. The in vitro coagulation effects of befovacimab were compared to samples supplemented with rFVIII. RESULTS Befovacimab induced consistent pro-coagulant responses in ROTEM parameters including reduction in clotting times and increases in α-angle; induced reductions in dPT clotting time; and improvements in TGA parameters (reduced lag time and increased thrombin generation parameters). There was a modest concentration-dependent response generally from 0.5- to 10 nM, after which, the pharmacodynamic effect plateaued through the 100 nM concentration. Befovacimab concentrations of 5 to 10 nM showed pro-coagulant activity comparable to blood samples supplemented with rFVIII 10-40%. CONCLUSIONS Befovacimab has modest dose-response effects from 0.5 to 10 nM with minimal improvement with higher concentrations. In vitro befovacimab blood concentrations of 5 to 10 nM had pro-coagulant effects similar to blood supplemented with rFVIII 10- to 40%.
Collapse
Affiliation(s)
- Erika J Martin
- Coagulation Advancement Laboratory, Department of Pharmacotherapy & Outcomes Sciences, Virginia Commonwealth University (VCU) School of Pharmacy, Richmond, VA, USA
| | - Melinda E Nolte
- Department of Internal Medicine, Division of Hematology/Oncology, VCU School of Medicine, Richmond, VA, USA
| | - Janice Kuhn
- Department of Internal Medicine, Division of Hematology/Oncology, VCU School of Medicine, Richmond, VA, USA
| | - Nicole Schmidt
- Research & Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Nils Pfaff
- Research & Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | - Donald F Brophy
- Coagulation Advancement Laboratory, Department of Pharmacotherapy & Outcomes Sciences, Virginia Commonwealth University (VCU) School of Pharmacy, Richmond, VA, USA
| |
Collapse
|
12
|
Bahmid NA, Heising J, Fogliano V, Dekker M. Packaging Design Using Mustard Seeds as a Natural Antimicrobial: A Study on Inhibition of Pseudomonas fragi in Liquid Medium. Foods 2020; 9:E789. [PMID: 32560102 PMCID: PMC7353601 DOI: 10.3390/foods9060789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/03/2020] [Accepted: 06/10/2020] [Indexed: 11/16/2022] Open
Abstract
Pseudomonas fragi is the dominant spoilage organism in various foods, especially in spoiled milk, fish, and meats. Its growth can be inhibited by releasing allyl isothiocyanate (AITC) from ground mustard seeds in food packages. This paper aims to investigate the antimicrobial potential of ground mustard seeds against P. fragi growth and the effectiveness of released AITC concentration from mustard seeds on microbial inhibition of the spoilage bacteria growing in the liquid medium. The AITC concentration in the headspace and the liquid medium was measured and the growth of P. fragi in the liquid medium was monitored. Depending on the concentration of AITC, not only growth was inhibited but a reduction of the total count of P. fragi was even observed. The inactivation rate (k) of P. fragi was estimated using first-order inactivation kinetics and the minimum gaseous-released AITC to inactivate P. fragi was determined. Higher AITC concentration in the headspace and liquid medium was observed when using a higher amount of ground mustard seeds and a lower food to headspace ratio. Increasing the amount of ground mustard seeds (>100 mg per 10 mL liquid medium) led to full inactivation of P. fragi in 48 hours. By using an inhibition sigmoid Emax model, the minimum gaseous-released AITC for inactivation of P. fragi in 48 hours was observed around 15 µg/L headspace. These results indicate that inhibition of the spoilage bacteria and extending the shelf life using ground mustard seeds is only possible by applying a careful design of the packaging system.
Collapse
Affiliation(s)
- Nur Alim Bahmid
- Food Quality and Design Group, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands; (N.A.B.); (J.H.); (V.F.)
- Agriculture and Forestry Faculty, Universitas Sulawesi Barat, Majene 91412, Indonesia
| | - Jenneke Heising
- Food Quality and Design Group, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands; (N.A.B.); (J.H.); (V.F.)
| | - Vincenzo Fogliano
- Food Quality and Design Group, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands; (N.A.B.); (J.H.); (V.F.)
| | - Matthijs Dekker
- Food Quality and Design Group, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands; (N.A.B.); (J.H.); (V.F.)
| |
Collapse
|
13
|
Butterfield JSS, Hege KM, Herzog RW, Kaczmarek R. A Molecular Revolution in the Treatment of Hemophilia. Mol Ther 2020; 28:997-1015. [PMID: 31843450 PMCID: PMC7132613 DOI: 10.1016/j.ymthe.2019.11.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022] Open
Abstract
For decades, the monogenetic bleeding disorders hemophilia A and B (coagulation factor VIII and IX deficiency) have been treated with systemic protein replacement therapy. Now, diverse molecular medicines, ranging from antibody to gene to RNA therapy, are transforming treatment. Traditional replacement therapy requires twice to thrice weekly intravenous infusions of factor. While extended half-life products may reduce the frequency of injections, patients continue to face a lifelong burden of the therapy, suboptimal protection from bleeding and joint damage, and potential development of neutralizing anti-drug antibodies (inhibitors) that require less efficacious bypassing agents and further reduce quality of life. Novel non-replacement and gene therapies aim to address these remaining issues. A recently approved factor VIII-mimetic antibody accomplishes hemostatic correction in patients both with and without inhibitors. Antibodies against tissue factor pathway inhibitor (TFPI) and antithrombin-specific small interfering RNA (siRNA) target natural anticoagulant pathways to rebalance hemostasis. Adeno-associated virus (AAV) gene therapy provides lasting clotting factor replacement and can also be used to induce immune tolerance. Multiple gene-editing techniques are under clinical or preclinical investigation. Here, we provide a comprehensive overview of these approaches, explain how they differ from standard therapies, and predict how the hemophilia treatment landscape will be reshaped.
Collapse
Affiliation(s)
| | - Kerry M Hege
- Department of Pediatrics, Indiana University School of Medicine, IUPUI-Wells Center for Pediatric Research, Indianapolis, IN, USA
| | - Roland W Herzog
- Department of Pediatrics, University of Florida, Gainesville, FL, USA; Department of Pediatrics, Indiana University School of Medicine, IUPUI-Wells Center for Pediatric Research, Indianapolis, IN, USA.
| | - Radoslaw Kaczmarek
- Department of Pediatrics, Indiana University School of Medicine, IUPUI-Wells Center for Pediatric Research, Indianapolis, IN, USA; Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, Poland.
| |
Collapse
|
14
|
Franchini M, Marano G, Pati I, Veropalumbo E, Vaglio S, Pupella S, Masiello F, Cruciani M, Mengoli C, Liumbruno GM. Investigational drugs to treat hemophilia. Expert Opin Investig Drugs 2020; 29:295-301. [DOI: 10.1080/13543784.2020.1722999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Massimo Franchini
- Italian National Blood Centre, National Institute of Health, Rome, Italy
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Giuseppe Marano
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | - Ilaria Pati
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | - Eva Veropalumbo
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | - Stefania Vaglio
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | - Simonetta Pupella
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | - Francesca Masiello
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | - Mario Cruciani
- Italian National Blood Centre, National Institute of Health, Rome, Italy
- Infection Control Committee and Antibiotic Stewardship Programme, AULSS9 Scaligera, Verona, Italy
| | - Carlo Mengoli
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | | |
Collapse
|
15
|
Neubert H, Shuford CM, Olah TV, Garofolo F, Schultz GA, Jones BR, Amaravadi L, Laterza OF, Xu K, Ackermann BL. Protein Biomarker Quantification by Immunoaffinity Liquid Chromatography–Tandem Mass Spectrometry: Current State and Future Vision. Clin Chem 2020; 66:282-301. [DOI: 10.1093/clinchem/hvz022] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Abstract
Immunoaffinity–mass spectrometry (IA-MS) is an emerging analytical genre with several advantages for profiling and determination of protein biomarkers. Because IA-MS combines affinity capture, analogous to ligand binding assays (LBAs), with mass spectrometry (MS) detection, this platform is often described using the term hybrid methods. The purpose of this report is to provide an overview of the principles of IA-MS and to demonstrate, through application, the unique power and potential of this technology. By combining target immunoaffinity enrichment with the use of stable isotope-labeled internal standards and MS detection, IA-MS achieves high sensitivity while providing unparalleled specificity for the quantification of protein biomarkers in fluids and tissues. In recent years, significant uptake of IA-MS has occurred in the pharmaceutical industry, particularly in the early stages of clinical development, enabling biomarker measurement previously considered unattainable. By comparison, IA-MS adoption by CLIA laboratories has occurred more slowly. Current barriers to IA-MS use and opportunities for expanded adoption are discussed. The path forward involves identifying applications for which IA-MS is the best option compared with LBA or MS technologies alone. IA-MS will continue to benefit from advances in reagent generation, more sensitive and higher throughput MS technologies, and continued growth in use by the broader analytical community. Collectively, the pursuit of these opportunities will secure expanded long-term use of IA-MS for clinical applications.
Collapse
|
16
|
Shima M. Current progress and future direction in the treatment for hemophilia. Int J Hematol 2019; 111:16-19. [DOI: 10.1007/s12185-019-02786-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
|
17
|
Yuan D, Rode F, Cao Y. A systems pharmacokinetic/pharmacodynamic model for concizumab to explore the potential of anti-TFPI recycling antibodies. Eur J Pharm Sci 2019; 138:105032. [PMID: 31394258 PMCID: PMC6824202 DOI: 10.1016/j.ejps.2019.105032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 01/31/2023]
Abstract
Concizumab is a humanized monoclonal antibody in clinical investigation directed against membrane-bound and soluble tissue factor pathway inhibitor (mTFPI and sTFPI) for treatment of hemophilia. Concizumab displays a non-linear pharmacokinetic (PK) profile due to mTFPI-mediated endocytosis and necessitates a high dose and frequent dosing to suppress the abundant sTFPI, a negative regulator of coagulation. Recycling antibodies that can dissociate bound mTFPI/sTFPI in endosomes for degradation and rescue antibody from degradation have a potential in reducing the dose by extending antibody systemic persistence and sTFPI suppression. We developed a systems PK/pharmacodynamics (PD) model with nested endosome compartments to simulate the effect of decreased antibody binding to mTFPI/sTFPI in endosomes on antibody clearance and sTFPI suppression for exploring the potential of anti-TFPI recycling antibodies in reducing the dose. A dynamic model-building strategy was taken. A reduced PK/PD model without the endosome compartments was developed to optimize unknown target turnover parameters using concizumab PK data. The optimized parameters were then employed in the systems PK/PD model for simulations. The obtained systems PK/PD model adequately described the PK of concizumab in rabbits, monkeys, and humans and the PD in humans. The systems PK/PD model predicted that an anti-TFPI recycling antibody with a 100-fold higher mTFPI/sTFPI dissociation constant in endosomes than concizumab can extend sTFPI suppression from 12 days to 1 month. Thus, the systems PK/PD model provides a quantitative platform for guiding the engineering and translational development of anti-TFPI recycling antibodies.
Collapse
Affiliation(s)
- Dongfen Yuan
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frederik Rode
- Novo Nordisk, Translational DMPK, H. Lundbeck A/S, Denmark
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
18
|
Prophylactic treatment in hemophilic patients with inhibitors. Blood Coagul Fibrinolysis 2019; 30:S14-S18. [DOI: 10.1097/mbc.0000000000000823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Patel‐Hett S, Martin EJ, Mohammed BM, Rakhe S, Sun P, Barrett JC, Nolte ME, Kuhn J, Pittman DD, Murphy JE, Brophy DF. Marstacimab, a tissue factor pathway inhibitor neutralizing antibody, improves coagulation parameters of ex vivo dosed haemophilic blood and plasmas. Haemophilia 2019; 25:797-806. [DOI: 10.1111/hae.13820] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Sunita Patel‐Hett
- Rare Disease Research Unit Early Clinical Development, Pfizer Inc Cambridge MA USA
| | - Erika J. Martin
- Coagulation Advancement Laboratory, Department of Pharmacotherapy & Outcomes Science Virginia Commonwealth University (VCU) Richmond VA USA
| | - Bassem M. Mohammed
- Coagulation Advancement Laboratory, Department of Pharmacotherapy & Outcomes Science Virginia Commonwealth University (VCU) Richmond VA USA
- Department of Clinical Pharmacy, Faculty of Pharmacy Cairo University Cairo Egypt
- Department of Pathology, Microbiology, and Immunology Vanderbilt University Medical Center Nashville TN USA
| | - Swapnil Rakhe
- Rare Disease Research Unit Early Clinical Development, Pfizer Inc Cambridge MA USA
| | - Pengling Sun
- Rare Disease Research Unit Early Clinical Development, Pfizer Inc Cambridge MA USA
| | - John C. Barrett
- Division of Hematology/Oncology, Department of Internal Medicine VCU Richmond VA USA
| | - Melinda E. Nolte
- Division of Hematology/Oncology, Department of Internal Medicine VCU Richmond VA USA
| | - Janice Kuhn
- Division of Hematology/Oncology, Department of Internal Medicine VCU Richmond VA USA
| | - Debra D. Pittman
- Rare Disease Research Unit Early Clinical Development, Pfizer Inc Cambridge MA USA
| | - John E. Murphy
- Rare Disease Research Unit Early Clinical Development, Pfizer Inc Cambridge MA USA
| | - Donald F. Brophy
- Coagulation Advancement Laboratory, Department of Pharmacotherapy & Outcomes Science Virginia Commonwealth University (VCU) Richmond VA USA
| |
Collapse
|
20
|
Ahlberg J, Giragossian C, Li H, Myzithras M, Raymond E, Caviness G, Grimaldi C, Brown SE, Perez R, Yang D, Kroe-Barrett R, Joseph D, Pamulapati C, Coble K, Ruus P, Woska JR, Ganesan R, Hansel S, Mbow ML. Retrospective analysis of model-based predictivity of human pharmacokinetics for anti-IL-36R monoclonal antibody MAB92 using a rat anti-mouse IL-36R monoclonal antibody and RNA expression data (FANTOM5). MAbs 2019; 11:956-964. [PMID: 31068073 PMCID: PMC6601564 DOI: 10.1080/19420862.2019.1615345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Accurate prediction of the human pharmacokinetics (PK) of a candidate monoclonal antibody from nonclinical data is critical to maximize the success of clinical trials. However, for monoclonal antibodies exhibiting nonlinear clearance due to target-mediated drug disposition, PK predictions are particularly challenging. That challenge is further compounded for molecules lacking cross-reactivity in a nonhuman primate, in which case a surrogate antibody selective for the target in rodent may be required. For these cases, prediction of human PK must account for any interspecies differences in binding kinetics, target expression, target turnover, and potentially epitope. We present here a model-based method for predicting the human PK of MAB92 (also known as BI 655130), a humanized IgG1 κ monoclonal antibody directed against human IL-36R. Preclinical PK was generated in the mouse with a chimeric rat anti-mouse IgG2a surrogate antibody cross-reactive against mouse IL-36R. Target-specific parameters such as antibody binding affinity (KD), internalization rate of the drug target complex (kint), target degradation rate (kdeg), and target abundance (R0) were integrated into the model. Two different methods of assigning human R0 were evaluated: the first assumed comparable expression between human and mouse and the second used high-resolution mRNA transcriptome data (FANTOM5) as a surrogate for expression. Utilizing the mouse R0 to predict human PK, AUC0-∞ was substantially underpredicted for nonsaturating doses; however, after correcting for differences in RNA transcriptome between species, AUC0-∞ was predicted largely within 1.5-fold of observations in first-in-human studies, demonstrating the validity of the modeling approach. Our results suggest that semi-mechanistic models incorporating RNA transcriptome data and target-specific parameters may improve the predictivity of first-in-human PK.
Collapse
Affiliation(s)
- Jennifer Ahlberg
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Craig Giragossian
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Hua Li
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Maria Myzithras
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Ernie Raymond
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Gary Caviness
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Christine Grimaldi
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Su-Ellen Brown
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Rocio Perez
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Danlin Yang
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Rachel Kroe-Barrett
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - David Joseph
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Chandrasena Pamulapati
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Kelly Coble
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Peter Ruus
- b Translational Medicine, Clinical Pharmacology , Boehringer Ingelheim Pharma GmbH & Co. KG , Ingelheim am Rein , Germany
| | - Joseph R Woska
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Rajkumar Ganesan
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - Steven Hansel
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| | - M Lamine Mbow
- a Biotherapeutics Disccovery Research , Boehringer Ingelheim Pharmaceuticals, Inc , Ridgefield , CT , USA
| |
Collapse
|
21
|
Xiang Y, Parng C, Olson K, Seletskaia E, Gorovits B, Jani D, Caiazzo T, Joyce A, Donley J. Neutralizing Antibody Assay Development with High Drug and Target Tolerance to Support Clinical Development of an Anti-TFPI Therapeutic Monoclonal Antibody. AAPS JOURNAL 2019; 21:46. [PMID: 30927117 DOI: 10.1208/s12248-019-0320-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/12/2019] [Indexed: 11/30/2022]
Abstract
Immunogenicity is a major challenge for protein therapeutics which can potentially reduce drug efficacy and safety and is often being monitored by anti-drug antibody (ADA) and neutralizing antibody (NAb) assays. Circulating targets and residual drugs in matrices can have significant impacts on accuracy of results from ADA and NAb assays, and sufficient drug and target tolerance for these assays are necessary. Here, we report the development of a competitive ligand binding (CLB) NAb assay for an anti-TFPI (tissue factor pathway inhibitor) monoclonal antibody (PF-06741086) with high drug and target tolerance to support ongoing clinical studies. A double acid affinity capture elution approach was used to mitigate drug interference, and a robust target removal strategy was employed to enhance target tolerance. The validated NAb assay has sensitivity of 313 ng/mL, drug tolerance of 50 μg/mL, and target tolerance of 1200 ng/mL. A step-by-step tutorial of assay development is described in this manuscript along with the rationale for using a high drug/target tolerant NAb assay. The NAb assay cut point factor obtained was 0.78. Other assay performance characteristics, e.g., precision and selectivity, are also discussed. This validated method demonstrated a superior drug and target tolerance to warrant specific and precise characterization of the NAb responses in support of ongoing clinical studies.
Collapse
|
22
|
Ljung R, Auerswald G, Benson G, Dolan G, Duffy A, Hermans C, Jiménez-Yuste V, Lambert T, Morfini M, Zupančić-Šalek S, Santagostino E. Inhibitors in haemophilia A and B: Management of bleeds, inhibitor eradication and strategies for difficult-to-treat patients. Eur J Haematol 2018; 102:111-122. [PMID: 30411401 PMCID: PMC6936224 DOI: 10.1111/ejh.13193] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/25/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
The standard therapy for patients with haemophilia is prophylactic treatment with replacement factor VIII (FVIII) or factor IX (FIX). Patients who develop inhibitors against FVIII/FIX face an increased risk of bleeding, and the likelihood of early development of progressive arthropathy, alongside higher treatment-related costs. Bypassing agents can be used to prevent and control bleeding, as well as the recently licensed prophylaxis, emicizumab, but their efficacy is less predictable than that of factor replacement therapy. Antibody eradication, by way of immune tolerance induction (ITI), is still the preferred management strategy for treating patients with inhibitors. This approach is successful in most patients, but some are difficult to tolerise and/or are unresponsive to ITI, and they represent the most complicated patients to treat. However, there are limited clinical data and guidelines available to help guide physicians in formulating the next treatment steps in these patients. This review summarises currently available treatment options for patients with inhibitors, focussing on ITI regimens and those ITI strategies that may be used in difficult-to-treat patients. Some alternative, non-ITI approaches for inhibitor management, are also proposed.
Collapse
Affiliation(s)
- Rolf Ljung
- Department of Clinical Sciences-Paediatrics, Lund University, Lund, Sweden.,Malmö Centre for Thrombosis and Haemostasis, Skåne University Hospital, Malmö, Sweden
| | - Guenter Auerswald
- Klinikum Bremen-Mitte, Professor Hess Children's Hospital, Bremen, Germany
| | - Gary Benson
- Haemophilia and Thrombosis Centre, Belfast City Hospital, Belfast, UK
| | - Gerry Dolan
- Centre for Haemostasis and Thrombosis, St Thomas' Hospital, London, UK
| | - Anne Duffy
- WFH Psychosocial Committee, Irish Haemophilia Society, Dublin, Ireland
| | - Cedric Hermans
- Haemostasis and Thrombosis Unit, Division of Haematology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | | | - Thierry Lambert
- Hemophilia Care Center, Faculté de Médecine Paris XI, Bicêtre AP-HP Hospital, Paris, France
| | | | - Silva Zupančić-Šalek
- University Hospital Centre Zagreb, School of Medicine, University of Osijek and Medical School University of Zagreb, Zagreb, Croatia
| | - Elena Santagostino
- Foundation IRCCS Cà Granda, Maggiore Hospital Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| |
Collapse
|
23
|
Anti-tissue factor pathway inhibitor (TFPI) therapy: a novel approach to the treatment of haemophilia. Int J Hematol 2018; 111:42-50. [PMID: 30302740 DOI: 10.1007/s12185-018-2548-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
Abstract
Novel approaches to the treatment of haemophilia are needed due to the limitations of the current standard of care, factor replacement therapy. Aspirations include lessening the treatment burden and effectively preventing joint damage. Treating haemophilia by restoring thrombin generation may be an effective approach. A promising target for restoring thrombin generation is tissue factor pathway inhibitor (TFPI), a multivalent Kunitz-type serine protease inhibitor that regulates tissue factor-induced coagulation via factor Xa-dependent feedback inhibition of the tissue factor-factor VIIa complex. Inhibition of TFPI reverts the coagulation process to a more primitive state evolutionarily, whilst regulation by other natural inhibitors is preserved. An aptamer and three monoclonal antibodies directed against TFPI have been investigated in clinical trials. As well as improving thrombin generation in the range associated with mild haemophilia, anti-TFPI therapies have the advantage of subcutaneous administration. However, the therapeutic window needs to be defined along with the potential for complications due to the novel mechanism of action. This review provides an overview of TFPI, its role in normal coagulation, the rationale for TFPI inhibition, and a summary of anti-TFPI therapies, previously or currently in development.
Collapse
|
24
|
Cardinal M, Kantaridis C, Zhu T, Sun P, Pittman DD, Murphy JE, Arkin S. A first-in-human study of the safety, tolerability, pharmacokinetics and pharmacodynamics of PF-06741086, an anti-tissue factor pathway inhibitor mAb, in healthy volunteers. J Thromb Haemost 2018; 16:1722-1731. [PMID: 29908043 DOI: 10.1111/jth.14207] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Indexed: 01/31/2023]
Abstract
Essentials Tissue factor pathway inhibitor (TFPI) is an antagonist of FXa and the TF-FVIIa complex. PF-06741086 is an IgG1 monoclonal antibody that targets the Kunitz-2 domain of TFPI. Single doses of PF-06741086 were evaluated in a phase 1 study in healthy volunteers. Data from this study support further investigation of PF-06741086 in individuals with hemophilia. SUMMARY Background Tissue factor pathway inhibitor (TFPI) is a protease inhibitor of the tissue factor-activated factor VII complex and activated FX. PF-06741086 is a mAb that targets TFPI to increase clotting activity. Objectives This study was a randomized, double-blind, sponsor-open, placebo-controlled, single intravenous or subcutaneous dose escalation study to evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of PF-06741086. Patients/Methods Volunteers who provided written informed consent were assigned to cohorts with escalating dose levels. Safety endpoints included treatment-emergent adverse events (TEAEs), infusion/injection site reactions, vital signs, electrocardiogram, and coagulation and hematology laboratory parameters. Pharmacokinetic (PK) and pharmacodynamic (PD) endpoints included exposures of PF-06741086 in plasma and measures of PF-06741086 pharmacology, respectively. Results Forty-one male volunteers were recruited overall. Thirty-two were dosed with PF-06741086 from 30 mg subcutaneously to 440 mg intravenously. All doses were safe and well tolerated. TEAEs were mild or moderate in severity, laboratory abnormalities were transient, there were no serious adverse events, there were no infusion/injection site reactions, and no dose escalation stopping criteria were met. Plasma exposures of PF-06741086 increased greater than proportionally with dose under the same dosing route. Coagulation pharmacology was demonstrated via total TFPI, dilute prothrombin time, D-dimer, prothrombin fragment 1 + 2 and thrombin generation assay parameters. Conclusions Single doses of PF-06741086 at multiple dose levels were safe and well tolerated in a healthy adult male population. The safety, PK and PD data from this study support progression to a multiple-dose study in hemophilic patients.
Collapse
MESH Headings
- Adolescent
- Adult
- Antibodies, Anti-Idiotypic/blood
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Neutralizing/blood
- Dose-Response Relationship, Drug
- Double-Blind Method
- Fatigue/chemically induced
- Half-Life
- Hemodynamics/drug effects
- Hemostatics/administration & dosage
- Hemostatics/adverse effects
- Hemostatics/blood
- Hemostatics/pharmacology
- Humans
- Injections, Intravenous
- Injections, Subcutaneous
- Lipoproteins/antagonists & inhibitors
- Lipoproteins/blood
- Lipoproteins/immunology
- Male
- Middle Aged
- Pain/chemically induced
- Young Adult
Collapse
Affiliation(s)
- M Cardinal
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - C Kantaridis
- Pfizer Clinical Research Unit, Brussels, Belgium
| | - T Zhu
- Early Clinical Development, Pfizer Inc., Cambridge, MA, USA
| | - P Sun
- Early Clinical Development, Pfizer Inc., Cambridge, MA, USA
| | - D D Pittman
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - J E Murphy
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - S Arkin
- Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, USA
| |
Collapse
|