Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Clin Cancer Res ; 30(11): 2309-2316, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38324398

ABSTRACT

In June 2022, the FDA extended the indication for lisocabtagene maraleucel (liso-cel) to include adults with large B-cell lymphoma (LBCL) who have refractory disease or relapse within 12 months of first-line chemoimmunotherapy (CIT), as well as transplant-ineligible adults with refractory disease or relapse after first-line CIT. Two clinical trials evaluating a single infusion of liso-cel preceded by lymphodepleting chemotherapy supported the second-line indications. TRANSFORM is a randomized, phase 3, open-label trial comparing liso-cel with standard second-line therapy, including planned autologous hematopoietic stem cell transplantation (HSCT), in 184 transplant-eligible patients. On interim analysis, event-free survival (EFS) by independent review committee (IRC) assessment was statistically significantly improved for the liso-cel arm, with a stratified hazard ratio of 0.34 [95% confidence interval (CI), 0.22-0.51; P < 0.0001]; the estimated median EFS was 10.1 months in the liso-cel arm versus 2.3 months in the control arm. PILOT is a single-arm phase 2 trial of second-line liso-cel in patients who were transplant-ineligible due to age or comorbidities but had adequate organ function for chimeric antigen receptor (CAR) T-cell therapy. Among 61 patients who received liso-cel (median age, 74 years), the IRC-assessed complete response rate was 54% (95% CI, 41-67). Among patients achieving complete response, the estimated 1-year rate of continued response was 68% (95% CI, 45-83). Of the 268 patients combined who received liso-cel as second-line therapy for LBCL, cytokine release syndrome occurred in 45% (Grade 3, 1.3%) and CAR T-cell-associated neurologic toxicities occurred in 27% (Grade 3, 7%), warranting a continued risk evaluation and mitigation strategy.


Subject(s)
Drug Approval , Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , United States Food and Drug Administration , Humans , United States , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/therapy , Aged , Male , Female , Middle Aged , Adult , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Biological Products/therapeutic use
2.
J Clin Pharmacol ; 63 Suppl 2: S110-S116, 2023 11.
Article in English | MEDLINE | ID: mdl-37942903

ABSTRACT

The current dosing strategy of immune globulin products for the treatment of primary immunodeficiency diseases (PIDDs) in the USA is based on total body weight (BW). The aim of our study was to assess the relationship between dose and trough level, and to determine whether an alternative dosing strategy should be considered for patients who are overweight or obese. We analyzed data in a total of 533 patients from 11 studies. We modeled the relationship between trough level and dose per week using a linear mixed model. We used an over-dispersed Poisson model to model the relationship between infection and trough level. In these analyses, we then combined the study-specific treatment effects using a random-effect or fixed-effect model. The mean administered dose per week was 9.77, 14.00, or 18.17 g in patients who were normal weight, overweight, or obese, respectively. Compared with a patient of normal weight, a 1 g increase in dose per week in a patient who was overweight was associated with a smaller increase in the trough level, 0.08 g/L less (95%CI -0.14 to -0.03 g/L), and a 1 g increase in dose per week in a patient who was obese was associated with a much smaller increase in trough level, 0.01 g/L less (95% CI -0.07 to 0.06 g/L). Last, for a 1 unit (g/L) increase in trough level, the expected number of infections remained the same, with a multiplicative factor of 1.01 (95%CI 0.98-1.04). Overall, we found no compelling evidence to justify a reconsideration of the current dosing strategy based on total BW for patients with PIDDs who are overweight or obese.


Subject(s)
Overweight , Primary Immunodeficiency Diseases , Humans , Immunoglobulin G/therapeutic use , Obesity/complications , Obesity/drug therapy , Overweight/complications , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/drug therapy , Clinical Trials as Topic
4.
Int Immunopharmacol ; 119: 109915, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36842918

ABSTRACT

In Part 1, we provided a general description of macromolecules, pharmacokinetics (PK) characteristics in non-pregnant subjects, and the physiological changes during pregnancy. Here we further elaborate on the impact of pregnancy on the PK of antibodies through illustrative case studies (immunoglobulins, infliximab, adalimumab and eculizumab). Using published data from nonclinical and clinical studies, we present measured or calculated PK parameters from pregnant subjects comparing with data from non-pregnant subjects, if available. Due to the paucity of PK data evaluating PK of antibodies during pregnancy, we also provide examples of PK studies for small molecules. Finally, we draw conclusions on the nature and direction of PK changes for both antibodies and small molecules as well as provide recommendations for areas that would benefit from further studies.


Subject(s)
Models, Biological , Pharmacokinetics , Pregnancy , Female , Humans , Adalimumab , Infliximab
5.
Int Immunopharmacol ; 117: 109914, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36841154

ABSTRACT

Pharmacokinetics (PK) studies are important to determine a safe and effective dose of both small and large molecule drugs. Intrinsic factors such as pregnancy can substantially alter the PK of a drug. Several PK studies have been published for small molecules administered during pregnancy, but such investigations are scarce for macromolecules including monoclonal and polyclonal antibodies. In this part 1 of 2 reviews, we first provide a general description of macromolecule drugs, the PK differences with small molecules, and current knowledge on their absorption, distribution, metabolism and elimination in non-pregnant subjects. We then review in detail the physiological changes during pregnancy. While some of the physiologic adaptions of pregnancy, for example increased plasma volume and cardiac output, are expected to impact PK of antibody therapeutics, the effects of others, such as increased GFR and altered immune responses are not fully understood. We conclude that further investigations are needed to fully elucidate how pregnancy can impact PK properties of macromolecules.


Subject(s)
Antibodies, Monoclonal , Antibodies , Pharmacokinetics , Female , Humans , Pregnancy , Antibodies/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology
7.
Pharm Res ; 39(8): 1669-1680, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35552984

ABSTRACT

Model-informed drug development (MIDD) is a powerful approach to support drug development and regulatory review. There is a rich history of MIDD applications at the U.S. Food and Drug Administration (FDA). MIDD applications span across the life cycle of the development of new drugs, generics, and biologic products. In new drug development, MIDD approaches are often applied to inform clinical trial design including dose selection/optimization, aid in the evaluation of critical regulatory review questions such as evidence of effectiveness, and development of policy. In the biopharmaceutics space, we see a trend for increasing role of computational modeling to inform formulation development and help strategize future in vivo studies or lifecycle plans in the post approval setting. As more information and knowledge becomes available pre-approval, quantitative mathematical models are becoming indispensable in supporting generic drug development and approval including complex generic drug products and are expected to help reduce overall time and cost. While the application of MIDD to inform the development of cell and gene therapy products is at an early stage, the potential for future application of MIDD include understanding and quantitative evaluation of information related to biological activity/pharmacodynamics, cell expansion/persistence, transgene expression, immune response, safety, and efficacy. With exciting innovations on the horizon, broader adoption of MIDD is poised to revolutionize drug development for greater patient and societal benefit.


Subject(s)
Drug Development , Drugs, Generic , Animals , Drug Approval , Humans , Life Cycle Stages , Pharmaceutical Preparations , United States , United States Food and Drug Administration
8.
Clin Infect Dis ; 75(1): e459-e465, 2022 08 24.
Article in English | MEDLINE | ID: mdl-34554235

ABSTRACT

BACKGROUND: After the failure of antibody therapies in treating hospitalized patients with coronavirus disease 2019 (COVID-19), we investigated the impact of viral replication on the pharmacokinetics and efficacy of a hyperimmune severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin (CoVIG) product in treating SARS-CoV-2 infection using an adult Syrian hamster model. METHODS: The CoVIG was manufactured from plasma donors who had recovered from COVID-19. The dose used (400 mg/kg) was based on the dose given in clinical trials to hospitalized patients with COVID-19. Hamsters were given a single dose of CoVIG 2 days after challenge with the SARS-CoV-2 virus (isolate NY/PV08410/2020), followed by sampling of blood, nasal, tracheal, and lung tissues at different time points. The blood samples were assayed for anti-SARS-CoV-2 spike binding and used to calculate pharmacokinetic (PK) parameters. Nasal wash, tracheal, and lung tissue samples were assayed for viral replication by polymerase chain reaction (subgenomic messenger RNA). RESULTS: CoVIG-treated hamsters showed a reduction in viral replication in the lower respiratory tract, but minimal reduction in the upper respiratory tract, after challenge with SARS-CoV-2. Challenge resulted in altered PK parameters proportionate to viral replication, resulting in decreased area under the curve, accelerated clearance, and shorter half-life of CoVIG. CONCLUSIONS: These data indicate that in the presence of actively replicating SARS-CoV-2 virus, PK parameters are altered and should trigger an adjustment in CoVIG dosing.


Subject(s)
COVID-19 Drug Treatment , Adult , Animals , Cricetinae , Humans , Immunoglobulins, Intravenous/therapeutic use , Lung , Mesocricetus , SARS-CoV-2
10.
J Clin Pharmacol ; 61 Suppl 1: S108-S116, 2021 06.
Article in English | MEDLINE | ID: mdl-34185903

ABSTRACT

There is a growing interest in the use of physiologically based pharmacokinetic (PBPK) models as clinical pharmacology drug development tools. In PBPK modeling, not every organ or physiological parameter is required, leading to the development of a minimal PBPK (mPBPK) model, which is simple and efficient. The objective of this study was to streamline mPBPK modeling approaches and enable straightforward prediction of clearance of protein-based products in children. Four mPBPK models for scaling clearance from adult to children were developed and evaluated on Excel spreadsheets using (1) liver and kidneys; (2) liver, kidneys, and skin; (3) liver, kidneys, skin, and lymph; and (4) interstitial, lymph, and plasma volume. There were 35 therapeutic proteins with a total of 113 observations across different age groups (premature neonates to adolescents). For monoclonal and polyclonal antibodies, more than 90% of observations were within a 0.5- to 2-fold prediction error for all 4 methods. For nonantibodies, 79% to 100% of observations were within the 0.5- to 2-fold prediction error for the 4 different methods. Methods 1 and 4 provided the best results, >90% of the total observations were within the 0.5- to 2-fold prediction error for all 3 classes of protein-based products across a wide age range. The precision of clearance prediction was comparatively lower in children ≤2 years of age vs older children (>2 years of age) with methods 1 and 4 predicting 80% to 100% and 75% to 90% of observations within the 0.5- to 2-fold prediction error, respectively. The results of the study indicated that mPBPK models can be developed on spreadsheets, with acceptable performance for prediction of clearance.


Subject(s)
Biological Products/pharmacokinetics , Drug Elimination Routes/physiology , Metabolic Clearance Rate/physiology , Models, Biological , Pediatrics/methods , Proteins/pharmacokinetics , Adolescent , Age Factors , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Biological Products/administration & dosage , Child , Child, Preschool , Humans , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/pharmacokinetics , Infant , Infant, Newborn , Proteins/administration & dosage
11.
CPT Pharmacometrics Syst Pharmacol ; 10(4): 286-290, 2021 04.
Article in English | MEDLINE | ID: mdl-33608998

ABSTRACT

As part of the US Food and Drug Administration (FDA)'s Prescription Drug User Fee Act (PDUFA) VI commitments, the Center for Biologics Evaluation and Research (CBER) and Center for Drug Evaluation and Research (CDER) are conducting a model-informed drug development (MIDD) pilot program. Sponsor(s) who apply and are selected will be granted meetings that aim to facilitate the application of MIDD approaches throughout the product development lifecycle and the regulatory process. Due to their complex mechanisms of action and limited clinical experience, cell and gene therapies have the potential to benefit from the application of MIDD methods, which may facilitate their safety and efficacy evaluations. Leveraging data that are generated from all stages of drug development into appropriate modeling and simulation techniques that inform decisions remains challenging. Additional discussions regarding the application of quantitative modeling approaches to drug development decisions, such as through the MIDD pilot program, may be crucial for both the sponsor(s) and regulatory review teams. Here, we share some perspectives on the opportunities and challenges for utilizing MIDD approaches for product review, which we hope will encourage investigators to publish their experiences and application of MIDD in gene therapy product development.


Subject(s)
Drug Development/legislation & jurisprudence , Genetic Therapy/methods , Immunotherapy, Adoptive/adverse effects , Computer Simulation , Dependovirus/chemistry , Dependovirus/metabolism , Humans , Immunotherapy, Adoptive/methods , Models, Biological , Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods , Pharmacokinetics , Research Design , Safety , Technology Assessment, Biomedical/statistics & numerical data , Treatment Outcome
12.
Drugs R D ; 21(1): 1-8, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33259037

ABSTRACT

At present, no cure is available for COVID-19 but vaccines, antiviral drugs, immunoglobulins, or the combination of immunoglobulins with antiviral drugs have been suggested and are in clinical trials. The purpose of this paper is to discuss the role of a pharmacokinetic and viral load analysis as a basis for adjusting immunoglobulin dosing to treat COVID-19. We reviewed the pre-clinical and clinical literature that describes the impact of a high antigen load on pharmacokinetic data following antibody treatment. Representative examples are provided to illustrate the effect of high viral and tumor loads on antibody clearance. We then highlight the implications of these factors for facilitating the development and dosing of hyperimmune anti-SARS CoV2 immunoglobulin. Both nonclinical and clinical examples indicate that high antigen loads, whether they be viral, bacterial, or tumoral in origin, result in increased clearance and decreased area under the curve and half-life of antibodies. A dosing strategy that matches the antigen load can be achieved by giving initially high doses and adjusting the frequency of dosing intervals based on pharmacokinetic parameters. We suggest that study design and dose selection for immunoglobulin products for the treatment of COVID-19 require special considerations such as viral load, antibody-virus interaction, and dosing adjustment based on the pharmacokinetics of the antibody.


Subject(s)
Antibodies, Viral/blood , Antiviral Agents/administration & dosage , COVID-19 Drug Treatment , COVID-19/blood , Immunoglobulins/administration & dosage , Viral Load/drug effects , Antigens, Viral/blood , Antiviral Agents/pharmacokinetics , Dose-Response Relationship, Drug , Humans , Immunoglobulins/blood , Viral Load/physiology
13.
Haemophilia ; 26(5): 817-825, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32842165

ABSTRACT

INTRODUCTION: Emerging, systematic approaches for capturing patient input, such as preference elicitation, can provide valuable information for the benefit-risk assessment of medical products for treating bleeding disorders, such as haemophilia. AIM: This study aims to identify existing and develop new methods to capture, rank and summarize preference scores for clotting factor therapies. METHODS: Haemophilia patient preference data were compiled from studies identified through literature review and publicly available US FDA patient-focused drug development meeting documents. Text mining was performed to identify major themes across studies. A standardized preference score was estimated and aggregated. RESULTS: Ten preference studies that employed qualitative (n = 3), and quantitative methods (n = 7) met the inclusion criteria. Text mining of qualitative and quantitative studies revealed similar themes as the standardized preference attribute importance. We found that seven quantitative studies employed discrete choice experiments (DCE)/conjoint analysis (CA) and examined a range of 5-12 attributes. For DCE/CA studies published prior to 2014 (n = 4), safety attributes (inhibitor and viral safety) were among the most important attributes, accounting for ~46% of the total utility measured. DCE/CA studies published after 2014 (n = 3) focused on frequency of infusion and reduction of bleeding risk, accounting for ~67% of the total utility. Interestingly, two studies that used different preference elicitation approaches (DCE and a monadic conjoint approach) both ranked infusion frequency as the most important attribute. CONCLUSIONS: Although there are few published patient preference studies for haemophilia, the results of this study can be viewed in the larger context of enhancing scientific methods of incorporating patient input in medical product development.


Subject(s)
Blood Coagulation Factors/therapeutic use , Hemophilia A/blood , Blood Coagulation Factors/pharmacology , Female , Humans , Male
14.
J Clin Pharmacol ; 60(11): 1453-1460, 2020 11.
Article in English | MEDLINE | ID: mdl-32501555

ABSTRACT

Dosing of coagulation factor products is mainly determined based on a patient's body weight; however, several studies have reported high interindividual variability in their pharmacokinetics (PK). The objective of this study was to develop and evaluate 2 sparse sampling methods for the estimation of AUC of recombinant factor IX (BeneFIX) as proof of concept for dose individualization. A population pharmacokinetic model was used to generate the plasma factor IX activity-versus-time data. The linear limited sampling model (LLSM) was developed based on the correlation of factor IX activity versus AUC0-72 hours following screening of several blood sampling times in adolescent and adult subjects (n = 90 subjects). Factor IX trough concentrations were predicted from a relationship established from AUC versus factor IX activity measured 72 hours postdosing. Using the best selected sampling time, the LLSM and Bayesian model were validated in separate data sets (n = 75 subjects). Using the LLSM and Bayesian analysis, a blood sample at 24 hours predicted AUC with bias and root mean square error < 5% and < 15%, respectively. The predicted trough concentrations were ≥1 IU/dL in 99% and 100% of subjects by the LLSM and Bayesian model, respectively. The average factor IX dose for a target AUC of 800 IU·h/dL was 61, 60, and 63 IU/kg using the extensive (reference), LLSM and Bayesian model, respectively. Overall, the AUC, trough concentrations and individualized dosing of recombinant factor IX could be reasonably predicted using the LLSM and Bayesian model.


Subject(s)
Drug Monitoring/methods , Factor IX/metabolism , Factor IX/pharmacokinetics , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Adolescent , Adult , Area Under Curve , Bayes Theorem , Computer Simulation , Drug Dosage Calculations , Factor IX/administration & dosage , Humans , Linear Models , Monte Carlo Method , Recombinant Proteins/administration & dosage
15.
Antibodies (Basel) ; 9(2)2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32575458

ABSTRACT

Immunoglobulins (IGs) are widely used for the treatment of immunodeficiency syndromes and several autoimmune diseases. In neonates, IGs have been used for the treatment of alloimmune thrombocytopenia, in neonatal infections and in the rare cases of neonatal Kawasaki disease. This review aims to examine the various dosing regimens of IGs following intravenous (IV) and subcutaneous (SC) administration, pharmacokinetics (PK) of IGs, and the importance of trough values for the prevention of infections in patients with primary immune deficiency (PID). The review also focuses on the mechanism of catabolism of IGs and the impact on the half-life of IGs. Data and reviews were obtained from the literature and the FDA package inserts. The authors suggest that for dosing, the PK of IGs should be evaluated on the baseline-corrected concentrations since this approach provides an accurate estimate of half-life and clearance of IGs. We also suggest employing clearance as a primary PK parameter for dosing determination of IGs. We suggest that IV dosing would be more effective if given more frequently to adjust for the increased clearance at high doses and because the baseline-corrected half-life is much shorter than the baseline-uncorrected half-life. Regarding SC administration, the dose should be adjusted based on the absolute bioavailability (determined against IV dosing) of the product. Finally, we highlight clinical and PK data gaps for optimum and individualized dosing of IGs.

16.
Int Immunopharmacol ; 82: 106358, 2020 Mar 02.
Article in English | MEDLINE | ID: mdl-32135491

ABSTRACT

BACKGROUND: Immunoglobulins are widely used across multiple therapeutic areas such as immunodeficiency syndromes, infection and autoimmune diseases. The pharmacokinetics (PK) of immunoglobulins are well characterized in adults, but very little is known about the PK of immunoglobulins in neonates and infants. OBJECTIVE: The objective of the present study was to characterize the PK of Gammagard, an immunoglobulin, in very low birth weight preterm neonates. METHOD: Gammagard concentration-time data from very low birth weight neonates (bodyweight range 0.78-1.38 kg, n = 20) following intravenous administration of 500 mg/kg and 750 mg/kg were obtained from the literature. The data were analyzed with and without baseline correction using extensive blood samples (8 blood samples). Model-independent (non-compartmental) analysis was used to characterize the PK of Gammagard. RESULTS: Based on uncorrected baseline concentration-time data, the clearance and half-life of Gammagard were 3.1 ± 0.7 mL/day and 22 ± 6 days, respectively. Based on corrected baseline concentration-time data, the clearance and half-life of Gammagard were 20.2 ± 7.4 mL/day and 5.3 ± 2.2 days, respectively. CONCLUSION: The dose of immunoglobulins should be adjusted based on the PK of baseline corrected rather than baseline uncorrected profiles because baseline corrected PK parameters especially half-life reconciles with PK principles.

17.
Int Immunopharmacol ; 80: 106192, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31931361

ABSTRACT

BACKGROUND: Immunoglobulin products are widely used across multiple therapeutic areas such as immunodeficiency syndromes, infection and autoimmune diseases. The pharmacokinetics (PK) of immunoglobulins are well characterized in adults, but very little is known about the PK of immunoglobulins in neonates and infants (<2 years of age). OBJECTIVE: The objectives of the present study were: (1) characterize the PK of immunoglobulin intravenous preparation using model-independent (non-compartmental analysis), and (2) develop and evaluate a population PK model with extensive blood samples (8 blood samples) and sparse blood samples (2-3 blood samples). METHOD: Immunoglobulin G (IgG) concentration versus time data from very low birth weight neonates (n = 20) following intravenous administration were analyzed using nonlinear mixed effect modeling and non-compartmental approaches. Population pharmacokinetic models were developed from extensive and sparse sampling schemes. Models were evaluated based on the difference in objective function, goodness-of-fit plots and simulation based visual predictive check analysis. RESULTS: A non-compartmental analysis of IgG from neonates (bodyweight range 0.78-1.38 kg) indicated an average clearance of 3.0 ± 2.1 mL/day and volume of distribution at steady state 68 ± 25 mL. The population pharmacokinetic model from extensive sampling adequately described concentration- time data with mean clearance (2.7 mL/day), volume of central compartment (8.7 mL) and peripheral compartment (60 mL). The clearance and volume of distribution estimates using sparse sampling model (1 pre-and 2 post-dose blood samples) were comparable with extensive sampling. CONCLUSION: Our study provides important bridging data in scaling PK and dosing of immunoglobulins across a wide age range.


Subject(s)
Immunoglobulins, Intravenous/pharmacokinetics , Immunologic Factors/pharmacokinetics , Infant, Very Low Birth Weight/metabolism , Models, Biological , Humans , Immunoglobulins, Intravenous/blood , Immunologic Factors/blood , Infant, Newborn
18.
CPT Pharmacometrics Syst Pharmacol ; 9(1): 21-28, 2020 01.
Article in English | MEDLINE | ID: mdl-31652029

ABSTRACT

The use of computational models in drug development has grown during the past decade. These model-informed drug development (MIDD) approaches can inform a variety of drug development and regulatory decisions. When used for regulatory decision making, it is important to establish that the model is credible for its intended use. Currently, there is no consensus on how to establish and assess model credibility, including the selection of appropriate verification and validation activities. In this article, we apply a risk-informed credibility assessment framework to physiologically-based pharmacokinetic modeling and simulation and hypothesize this evidentiary framework may also be useful for evaluating other MIDD approaches. We seek to stimulate a scientific discussion around this framework as a potential starting point for uniform assessment of model credibility across MIDD. Ultimately, an overarching framework may help to standardize regulatory evaluation across therapeutic products (i.e., drugs and medical devices).


Subject(s)
Computer Simulation , Drug Development/methods , Models, Biological , Decision Making , Humans , Pharmacokinetics , Risk Assessment
19.
AAPS J ; 21(5): 96, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31376048

ABSTRACT

Most immune responses to biotherapeutic proteins involve the development of anti-drug antibodies (ADAs). New drugs must undergo immunogenicity assessments to identify potential risks at early stages in the drug development process. This immune response is T cell-dependent. Ex vivo assays that monitor T cell proliferation often are used to assess immunogenicity risk. Such assays can be expensive and time-consuming to carry out. Furthermore, T cell proliferation requires presentation of the immunogenic epitope by major histocompatibility complex class II (MHCII) proteins on antigen-presenting cells. The MHC proteins are the most diverse in the human genome. Thus, obtaining cells from subjects that reflect the distribution of the different MHCII proteins in the human population can be challenging. The allelic frequencies of MHCII proteins differ among subpopulations, and understanding the potential immunogenicity risks would thus require generation of datasets for specific subpopulations involving complex subject recruitment. We developed TCPro, a computational tool that predicts the temporal dynamics of T cell counts in common ex vivo assays for drug immunogenicity. Using TCPro, we can test virtual pools of subjects based on MHCII frequencies and estimate immunogenicity risks for different populations. It also provides rapid and inexpensive initial screens for new biotherapeutics and can be used to determine the potential immunogenicity risk of new sequences introduced while bioengineering proteins. We validated TCPro using an experimental immunogenicity dataset, making predictions on the population-based immunogenicity risk of 15 protein-based biotherapeutics. Immunogenicity rankings generated using TCPro are consistent with the reported clinical experience with these therapeutics.


Subject(s)
Antibodies/immunology , Drug Development/methods , Proteins/immunology , Antigen-Presenting Cells/immunology , Cell Proliferation/physiology , Computer Simulation , Humans , Proteins/administration & dosage , Risk Assessment/methods , T-Lymphocytes/immunology
20.
Regul Toxicol Pharmacol ; 108: 104436, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31381939

ABSTRACT

BACKGROUND: In recent years, there has been great interest from academia, industry and government scientists for an increased understanding of the mode of action of vaccine adjuvants to characterize the safety and efficacy of vaccines. In this context, pharmacokinetic (PK) and biodistribution studies are useful for quantifying the concentration of vaccine adjuvants in mechanistically or toxicologically relevant target tissues. METHODS: In this study, we conducted a comparative analysis of the PK and biodistribution profile of radiolabeled squalene for up to 336 h (14 days) after intramuscular injection of mice with adjuvanted H5N1 influenza vaccines. The evaluated adjuvants included an experimental-grade squalene-in-water (SQ/W) emulsion (AddaVax®) and an adjuvant system (AS03®) that contained squalene and α-tocopherol in the oil phase of the emulsion. RESULTS: The half-life of the initial exponential decay from quadriceps muscle was 1.5 h for AS03 versus 12.9 h for AddaVax. At early time points (1-6 h), there was about a 10-fold higher concentration of labeled squalene in draining lymph nodes following AS03 injection compared to AddaVax. The area-under-concentration curve up to 336 h (AUC0-336hr) and peak concentration of squalene in spleen (immune organ) was about 1.7-fold higher following injection of AS03 than AddaVax. The peak systemic tissue concentration of squalene from the two adjuvants, with or without antigen, remained below 1% of injected dose for toxicologically relevant target tissues, such as spinal cord, brain, and kidney. The pharmacokinetics of AS03 was unaffected by the presence of H5N1 antigen. CONCLUSIONS: This study demonstrates a rapid decline of AS03 from the quadriceps muscles of mice as compared to conventional SQ/W emulsion adjuvant, with an increased transfer to mechanistically relevant tissues such as local lymph nodes. Systemic tissue exposure to potential toxicological target tissues was very low.


Subject(s)
Adjuvants, Immunologic/pharmacokinetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/pharmacokinetics , Polysorbates/pharmacokinetics , Squalene/pharmacokinetics , alpha-Tocopherol/pharmacokinetics , Animals , Antigens/immunology , Drug Combinations , Emulsions , Female , Injections, Intramuscular , Lymph Nodes/metabolism , Male , Mice, Inbred BALB C , Quadriceps Muscle/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...