Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.207
Filter
1.
Arch Dermatol Res ; 316(8): 589, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214920

ABSTRACT

Bermekimab is a human-derived recombinant monoclonal antibody that exhibits immunoregulatory activity by specifically blocking interleukin-1α activity. Four phase 2 studies evaluated efficacy and safety of bermekimab in patients with moderate-to-severe atopic dermatitis (AD). In addition, a novel human skin explant model was developed to assess bermekimab pharmacokinetics/pharmacodynamics and proteomic/transcriptomic effects. Study 1 (NCT03496974, N = 38) was an open-label, dose escalation study of subcutaneous bermekimab (200 mg or 400 mg). Study 2 (NCT04021862, N = 87) was a double-blind, placebo-controlled, randomized (1:1:1) study of subcutaneous bermekimab (400 mg every week (qw) or every 2 weeks) or placebo. GENESIS (NCT04791319, N = 198) was a double-blind, placebo- and active-comparator-controlled, randomized (1:1:2:2) study of placebo, subcutaneous bermekimab (350 mg or 700 mg qw), or dupilumab. LUNA (NCT04990440, N = 6) was a double-blind, placebo-controlled, randomized (4:1) study of intravenous bermekimab 800 mg qw or placebo. A novel human ex vivo skin pharmacodynamic assay supported phase 0 (NCT03953196) and phase 1 (NCT04544813) studies. In Study 1, 400 mg subcutaneous bermekimab showed improvement in efficacy assessments (e.g., ≥ 75% improvement of EASI over baseline, IGA 0/1, and worst itch); however, efficacy was not confirmed in Study 2 or GENESIS. Consequently, GENESIS and LUNA were terminated early. The novel human ex vivo skin pharmacodynamic assay demonstrated that bermekimab reduced downstream skin injury responses. Although bermekimab showed potential as an AD treatment in preclinical and early open-label trials, larger controlled studies (Study 2 and GENESIS) did not confirm those initial results.


Subject(s)
Dermatitis, Atopic , Interleukin-1alpha , Humans , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/metabolism , Male , Female , Adult , Double-Blind Method , Middle Aged , Injections, Subcutaneous , Treatment Outcome , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Young Adult , Skin/drug effects , Skin/pathology , Skin/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Adolescent , Severity of Illness Index , Aged
2.
Mol Pharm ; 21(9): 4465-4475, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39110837

ABSTRACT

Transdermal microneedles have demonstrated promising potential as an alternative to typical drug administration routes for the treatment of various diseases. As microneedles offer lower administration burden with enhanced patient adherence and reduced ecological footprint, there is a need for further exploitation of microneedle devices. One of the main objectives of this work was to initially develop an innovative biobased photocurable resin with high biobased carbon content comprising isobornyl acrylate (IBA) and pentaerythritol tetraacrylate blends (50:50 wt/wt). The optimization of the printing and curing process resulted in µNe3dle arrays with durable mechanical properties and piercing capacity. Another objective of the work was to employ the 3D printed hollow µNe3dles for the treatment of osteoporosis in vivo. The 3D printed µNe3dle arrays were used to administer denosumab (Dmab), a monoclonal antibody, to osteoporotic mice, and the serum concentrations of critical bone minerals were monitored for six months to assess recovery. It was found that the Dmab administered by the 3D printed µNe3dles showed fast in vitro rates and induced an enhanced therapeutic effect in restoring bone-related minerals compared to subcutaneous injections. The findings of this study introduce a novel green approach with a low ecological footprint for 3D printing of biobased µNe3dles, which can be tailored to improve clinical outcomes and patient compliance for chronic diseases.


Subject(s)
Osteoporosis , Printing, Three-Dimensional , Animals , Mice , Osteoporosis/drug therapy , Female , Denosumab/administration & dosage , Drug Delivery Systems/methods , Needles , Administration, Cutaneous , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics
3.
Antimicrob Agents Chemother ; 68(8): e0065524, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39012102

ABSTRACT

We report the results of a first-in-human phase 1 clinical study to evaluate TRL1068, a native human monoclonal antibody that disrupts bacterial biofilms with broad-spectrum activity against both Gram-positive and Gram-negative species. The study population consisted of patients with chronic periprosthetic joint infections (PJIs) of the knee or hip, including both monomicrobial and polymicrobial infections, that are highly resistant to antibiotics due to biofilm formation. TRL1068 was administered via a single pre-surgical intravenous infusion in three sequentially ascending dose groups (6, 15, and 30 mg/kg). Concomitant perioperative antibiotics were pathogen-targeted as prescribed by the treating physician. In this double-blinded study, 4 patients were randomized to receive placebo and 11 patients to receive TRL1068 on day 1, as well as targeted antibiotics for 7 days prior to the scheduled removal of the infected implant and placement of an antibiotic-eluting spacer as the first stage of the standard of care two-stage exchange arthroplasty. No adverse events attributable to TRL1068 were reported. TRL1068 serum half-life was 15-18 days. At day 8, the concentration in synovial fluid was approximately 60% of the blood level and thus at least 15-fold above the threshold for biofilm-disrupting activity in vitro. Explanted prostheses were sonicated to release adherent bacteria for culture, with elimination of the implant bacteria observed in 3 of the 11 patients who received TRL1068, which compares favorably to prior PJI treatments. None of the patients who received TRL1068 had a relapse of the original infection by the end of the study (day 169). CLINICAL TRIALS: This study is registered with ClinicalTrials.gov as NCT04763759.


Subject(s)
Anti-Bacterial Agents , Antibodies, Monoclonal , Biofilms , Prosthesis-Related Infections , Humans , Biofilms/drug effects , Prosthesis-Related Infections/drug therapy , Prosthesis-Related Infections/microbiology , Female , Male , Middle Aged , Aged , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Double-Blind Method , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology
4.
J Pharm Biomed Anal ; 249: 116364, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39047461

ABSTRACT

In preclinical protein therapeutic development studies, the emergence of anti-drug antibodies (ADA) can potentially impact drug pharmacokinetics and safety. While immunogenicity assessment is not mandatory in preclinical studies, banking samples can be valuable for interpreting unexpected pharmacological responses. Immunoassays that use generic reagents across different drug molecules can simplify ADA assessment and expedite sample evaluations. This work showcases the ability of the Gyrolab automated immunoassay platform to detect and quantify both drug-free and drug-bound (total) ADAs to monoclonal antibody (mAb) therapeutics in cynomolgus monkey preclinical studies. Compared to the previously reported total ADA ELISA, the Gyrolab assay exhibited a wider signal dynamic range and increased drug tolerance. Similar sensitivity, dynamic range and cut point factors were observed for four therapeutic mAbs of different isotypes using the Gyrolab assay. Here we present a comparison of ADA assays using bridging ELISA, total ADA ELISA and total ADA Gyrolab formats in a cynomolgus monkey study where the subjects were treated with a single dose of a mAb therapeutic. We demonstrate that the total ADA assays detected host ADA responses at earlier time points compared to the bridging ELISA. The Gyrolab assay has the best correlation between signal-to-noise (S/N) and titer over a wide ADA concentration range, highlighting the utility of Gyrolab in S/N reporting of ADA response to eliminate the need for secondary titer assays. Collectively, our results demonstrate that the generic ADA Gyrolab assay minimizes the necessity for extensive assay development and optimization for therapeutic mAbs, streamlining preclinical immunogenicity assessment to enable interpretation of pharmacological data.


Subject(s)
Antibodies, Monoclonal , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Macaca fascicularis , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Enzyme-Linked Immunosorbent Assay/methods , Drug Evaluation, Preclinical/methods , Immunoassay/methods
5.
MAbs ; 16(1): 2384104, 2024.
Article in English | MEDLINE | ID: mdl-39083118

ABSTRACT

In vitro assessments for the prediction of pharmacokinetic (PK) behavior of biotherapeutics can help identify corresponding liabilities significantly earlier in the discovery timeline. This can minimize the need for extensive early in vivo PK characterization, thereby reducing animal usage and optimizing resources. In this study, we recommend bolstering classical developability workflows with in vitro measures correlated with PK. In agreement with current literature, in vitro measures assessing nonspecific interactions, self-interaction, and FcRn interaction are demonstrated to have the highest correlations to clearance in hFcRn Tg32 mice. Crucially, the dataset used in this study has broad sequence diversity and a range of physicochemical properties, adding robustness to our recommendations. Finally, we demonstrate a computational approach that combines multiple in vitro measurements with a multivariate regression model to improve the correlation to PK compared to any individual assessment. Our work demonstrates that a judicious choice of high throughput in vitro measurements and computational predictions enables the prioritization of candidate molecules with desired PK properties.


Subject(s)
Workflow , Animals , Mice , Humans , Antibodies, Monoclonal/pharmacokinetics , Receptors, Fc/metabolism , Mice, Transgenic , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism
6.
Mol Pharm ; 21(8): 3838-3847, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38949095

ABSTRACT

Claudin18.2 (CLDN18.2), due to its high expression in various gastric cancer tissues, is considered an optimal target for antitumor drug molecules. In this study, we obtained the labeled compounds of [125I]I-zolbetuximab using the Iodogen method. Under the optimum labeling conditions, the molar activity of [125I]I-zolbetuximab was 1.75 × 102 GBq/µmol, and the labeling efficiency was more than 99%. The labeled compounds exhibited excellent in vitro stability in both phosphate buffer saline (PBS, pH = 7.4) and fetal bovine serum systems (FBS) (radiochemical purity >90% at 72 h). The uptake percentage of [125I]I-zolbetuximab in MKN45-CLDN18.2 cells is 24.69 ± 0.84% after 6 h. The saturation binding assay and specificity assay further demonstrated the high specificity of [125I]I-zolbetuximab for CLDN18.2. The long retention at the tumor site and rapid metabolic clearance at other organ sites of [125I]I-zolbetuximab were observed in small-animal SPECT-CT imaging. The same trend was also observed in the biodistribution study. Due to the excellent targeting ability of zolbetuximab for CLDN18.2, [125I]I-zolbetuximab exhibits strong specific binding and retention with cells and tumors highly expressing CLDN18.2. However, the balance between mAb's longer cycle time in vivo and targeting binding and retention ability should be intensively considered for using this kind of radiopharmaceutical in the diagnosis and treatment of CLDN18.2-positive gastric cancer.


Subject(s)
Claudins , Animals , Humans , Mice , Tissue Distribution , Cell Line, Tumor , Claudins/metabolism , Iodine Radioisotopes , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/chemistry , Mice, Nude , Xenograft Model Antitumor Assays , Mice, Inbred BALB C , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/chemistry , Female , Male , Rats
7.
Clin Drug Investig ; 44(8): 587-599, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39073504

ABSTRACT

BACKGROUND AND OBJECTIVES: Nipocalimab is a high-affinity, fully human, effectorless immunoglobulin G1 monoclonal antibody targeting the neonatal Fc receptor and is currently under evaluation for the treatment of rare and prevalent immunoglobulin G autoantibody-mediated and alloantibody-mediated diseases. This phase I, randomized, double-blind, placebo-controlled, single-dose escalation study in healthy Japanese volunteers (N = 24) assessed the safety, pharmacokinetics, and effect on the serum immunoglobulin G level of single doses of nipocalimab. METHODS: Volunteers were grouped into three cohorts and received intravenous nipocalimab at 10, 30, or 60 mg/kg or placebo. To complement the study, genetic variation in the Fcγ receptor and transporter subunit of the neonatal Fc receptor was analyzed in Japanese and diverse populations. RESULTS: Nipocalimab was generally safe and well tolerated at all dose levels, with three (12.5% [3/24]) volunteers experiencing treatment-emergent adverse events across all nipocalimab doses. Mean serum immunoglobulin G levels decreased in a dose-dependent manner from baseline with nipocalimab treatment compared with placebo. Maximum serum nipocalimab concentrations demonstrated proportional increases with dose, while the area under the concentration-time curve was dose dependent and demonstrated non-linear increases with dose. Mean observed half-life was longer as the dose increased. Analysis of genetic variation in Fcγ receptor and transporter identified no unique Japanese variants or variants that alter amino acid sequences in or near the neonatal Fc receptor immunoglobulin G binding site targeted by nipocalimab. CONCLUSIONS: The comparable pharmacokinetic/pharmacodynamic profiles and highly conserved neonatal Fc receptor structure among diverse populations further support the clinical development of nipocalimab for the treatment of various immunoglobulin G autoantibody-mediated and alloantibody-mediated diseases across global sites and populations, including the Japanese population.


Subject(s)
Healthy Volunteers , Immunoglobulin G , Receptors, Fc , Humans , Double-Blind Method , Adult , Male , Female , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Asian People , Young Adult , Japan , Dose-Response Relationship, Drug , Middle Aged , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Histocompatibility Antigens Class I/genetics , East Asian People
8.
Int J Pharm ; 661: 124419, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38972522

ABSTRACT

This paper studies the transport of monoclonal antibodies through skin tissue and initial lymphatics, which impacts the pharmacokinetics of monoclonal antibodies. Our model integrates a macroscale representation of the entire skin tissue with a mesoscale model that focuses on the papillary dermis layer. Our results indicate that it takes hours for the drugs to disperse from the injection site to the papillary dermis before entering the initial lymphatics. Additionally, we observe an inhomogeneous drug distribution in the interstitial space of the papillary dermis, with higher drug concentrations near initial lymphatics and lower concentrations near blood capillaries. To validate our model, we compare our numerical simulation results with experimental data, finding a good alignment. Our parametric studies on the drug molecule properties and injection parameters suggest that a higher diffusion coefficient increases the transport and uptake rate while binding slows down these processes. Furthermore, shallower injection depths lead to faster lymphatic uptake, whereas the size of the injection plume has a minor effect on the uptake rate. These findings advance our understanding of drug transport and lymphatic absorption after subcutaneous injection, offering valuable insights for optimizing drug delivery strategies and the design of biotherapeutics.


Subject(s)
Antibodies, Monoclonal , Models, Biological , Injections, Subcutaneous , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Skin Absorption , Skin/metabolism , Lymphatic System/metabolism , Biological Transport , Humans , Computer Simulation , Animals
9.
Int J Pharm ; 661: 124446, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38996825

ABSTRACT

The pharmaceutical industry has experienced a remarkable increase in the use of subcutaneous injection of monoclonal antibodies (mAbs), attributed mainly to its advantages in reducing healthcare-related costs and enhancing patient compliance. Despite this growth, there is a limited understanding of how tissue mechanics, physiological parameters, and different injection devices and techniques influence the transport and absorption of the drug. In this work, we propose a high-fidelity computational model to study drug transport and absorption during and after subcutaneous injection of mAbs. Our numerical model includes large-deformation mechanics, fluid flow, drug transport, and blood and lymphatic uptake. Through this computational framework, we analyze the tissue material responses, plume dynamics, and drug absorption. We analyze different devices, injection techniques, and physiological parameters such as BMI, flow rate, and injection depth. Finally, we compare our numerical results against the experimental data from the literature.


Subject(s)
Antibodies, Monoclonal , Models, Biological , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Injections, Subcutaneous , Humans , Biological Transport , Computer Simulation , Skin Absorption
10.
Mol Pharm ; 21(9): 4416-4429, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39058284

ABSTRACT

Monoclonal antibodies (mAbs) have high binding specificity and affinity, making them attractive for treating brain diseases. However, their effectiveness is limited by poor blood-brain barrier (BBB) penetration and rapid central nervous system (CNS) clearance. Our group identified blood-brain barrier modulator (BBBM) peptides that improved mAb penetration across the BBB into the brain. In this study, we investigated the pharmacokinetics of a mAb delivered to the brain using BBBMs after intravenous (IV) administration and explored the impact of antibody format (size, neonatal Fc receptor (FcRn) binding, hyaluronic acid binding) on brain clearance following direct injection into the central nervous system (CNS) via intracerebroventricular (ICV) injection. IRDye800CW-labeled antibodies were administered into C57BL/6 mice via ICV or IV injection, and organ concentrations were measured after various time points. When a mAb was coadministered with a BBBM peptide, the permeation of mAb across the BBB was increased compared to mAb alone at early time points; however, the mAb was cleared within 2 h from the brain. ICV experiments revealed that an antibody Fab fragment had a higher brain exposure than a mAb, and that a Fab fused to a hyaluronic acid binding domain (Fab-VG1) showed remarkable improvement in brain exposure. These findings suggest that BBBMs and antibody format optimization may be promising strategies for enhancing brain retention of therapeutic antibodies.


Subject(s)
Antibodies, Monoclonal , Blood-Brain Barrier , Brain , Mice, Inbred C57BL , Receptors, Fc , Animals , Blood-Brain Barrier/metabolism , Mice , Brain/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Receptors, Fc/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Hyaluronic Acid/chemistry , Male , Immunoglobulin Fab Fragments , Peptides/chemistry , Peptides/pharmacokinetics , Tissue Distribution
11.
Anal Biochem ; 694: 115623, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39059567

ABSTRACT

CM310 is a recombinant humanized monoclonal antibody targeting Interleukin (IL)-4 receptor alpha (IL-4Rα). IL-4Rα blockade prevents IL-4 and IL-13 from binding to their receptor, thereby inhibiting downstream signaling pathways that drive Type 2 helper T-cell (Th2) inflammation. CM310 holds potential for treating Th2-related inflammatory diseases, such as asthma, atopic dermatitis and chronic sinusitis with nasal polyposis. In this study, a direct enzyme-linked immunosorbent assay (ELISA) was developed to measure the concentrations of CM310 in rat serum. Seven calibration standards (ranging from 25 to 1600 ng/mL) and three quality controls (70, 500 and 1250 ng/mL) were defined. The limit of detection (LOD), lower limit of quantification (LLOQ) and upper limit of quantification (ULOQ) were 13, 25 and 1600 ng/mL, respectively. The method exhibited excellent precision and accuracy and successfully applied to in vitro serum stability and pharmacokinetic (PK) studies. In conclusion, we have developed and validated a highly sensitive and selective method for measuring CM310 in Sprague-Dawley rats. The development and validation ELISA method met the acceptable criteria, which suggested that these can be applied to quantify CM310, as well as in PK studies.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Rats, Sprague-Dawley , Animals , Enzyme-Linked Immunosorbent Assay/methods , Rats , Recombinant Proteins , Antibodies, Monoclonal, Humanized/pharmacokinetics , Male , Limit of Detection , Humans , Interleukin-4 Receptor alpha Subunit/antagonists & inhibitors , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/blood
12.
BMC Infect Dis ; 24(1): 712, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033300

ABSTRACT

BACKGROUND: The ambitious goal to eliminate new pediatric HIV infections by 2030 requires accelerated prevention strategies in high-risk settings such as South Africa. One approach could be pre-exposure prophylaxis (PrEP) with broadly neutralizing anti-HIV-1 monoclonal antibodies (bNAbs). The aim of our study is to define the optimal dose(s), the ideal combination(s) of bNAbs in terms of potency and breadth, and timing of subcutaneous (SC) administration(s) to prevent breast milk transmission of HIV. METHODS: Two bNAbs, CAP256V2LS and VRC07-523LS, will be assessed in a sequential and randomized phase I, single-site, single-blind, dose-finding trial. We aim to investigate the 28-day safety and pharmacokinetics (PK) profile of incrementally higher doses of these bNAbs in breastfeeding HIV-1 exposed born without HIV neonates alongside standard of care antiretroviral (ARV) medication to prevent (infants) or treat (mothers) HIV infection. The trial design includes 3 steps and 7 arms (1, 2, 3, 4, 5, 6 and 6b) with 8 infants in each arm. The first step will evaluate the safety and PK profile of the bNAbs when given alone as a single subcutaneous (SC) administration at increasing mg/kg body weight doses within 96 h of birth: arms 1, 2 and 3 at doses of 5, 10, and 20 mg/kg of CAP256V2LS, respectively; arms 4 and 5 at doses of 20 and 30 mg/kg of VRC07-523LS, respectively. Step two will evaluate the safety and PK profile of a combination of the two bNAbs administered SC at fixed doses within 96 h of birth. Step three will evaluate the safety and PK profile of the two bNAbs administered SC in combination at fixed doses, after 3 months. Arms 1 and 6 will follow sequential recruitment, whereas randomization will occur sequentially between arms (a) 2 & 4 and (b) 3 & 5. Before each randomization, a safety pause will allow review of safety data of the preceding arms. DISCUSSION: The results of this trial will guide further studies on bNAbs to prevent breast milk transmission of HIV. PROTOCOL VERSION: Version 4.0 dated 15 March 2024. TRIAL REGISTRATION: Pan African Clinical Trial Registry (PACTR): PACTR202205715278722, 21 April 2022; South African National Clinical Trial Registry (SANCTR): DOH-27-062022-6058.


Subject(s)
HIV Antibodies , HIV Infections , HIV-1 , Female , Humans , Infant , Infant, Newborn , Male , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Breast Feeding , Broadly Neutralizing Antibodies/immunology , Broadly Neutralizing Antibodies/administration & dosage , Clinical Trials, Phase I as Topic , HIV Antibodies/administration & dosage , HIV Infections/prevention & control , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/transmission , HIV-1/immunology , Infectious Disease Transmission, Vertical/prevention & control , Injections, Subcutaneous , Pre-Exposure Prophylaxis/methods , Single-Blind Method , South Africa
13.
J Pharm Sci ; 113(9): 2915-2921, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986869

ABSTRACT

Antibodies blocking programmed death-1 (PD-1) and its natural ligand programmed death-ligand 1 (PD-L1) have been proved to be promising strategies in recent years. Hundreds of PD-1/PD-L1 antibodies are under development worldwide. Prediction of human pharmacokinetics (PK) in the preclinical stage is critical for designing dosing regimens in first-in-human studies. This study aims to predict the PK of PD-1/PD-L1 antibodies in human by scaling of monkey data. A systematic literature search of published articles on the PK of PD-1/PD-L1 antibodies in cynomolgus monkey and in human was conducted. Allometric scaling (AS), the species time-invariant (STIV) method, as well as physiologically based pharmacokinetic (PBPK) modeling were investigated. Six antibodies (avelumab, atezolizumab, nivolumab, pembrolizumab, cemiplimab, and zimberelimab) were included for investigation. The exponents used in this study were 0.85 and 1 for clearance (CL) and distribution volume (V), respectively, both for AS and STIV methods. The generic PBPK model for macromolecules in PK-Sim was used without further modifications. The dissociation constant of the antibody for binding to FcRn (KD) in endosome space for human was assumed to be two-fold of that for monkey. Predicted human CLs for the majority of drugs were within the observed range, while Vs were not well predicted using the AS method. The STIV method and the generic PBPK model can be employed to translate concentration-time curves of PD-1/PD-L1 antibodies from cynomolgus monkey to human with comparable efficacy. The results of this study provide reference for the early development of PD-1/PD-L1 antibodies.


Subject(s)
Antibodies, Monoclonal , B7-H1 Antigen , Macaca fascicularis , Programmed Cell Death 1 Receptor , Animals , Humans , Antibodies, Monoclonal/pharmacokinetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Models, Biological , Species Specificity , Immune Checkpoint Inhibitors/pharmacokinetics
14.
J Pharm Biomed Anal ; 248: 116318, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38908237

ABSTRACT

We designed and developed 9MW2821, an anti-Nectin-4 antibody-drug conjugate (ADC) with an enzymatically cleavable valine-citrulline linker and monomethyl auristatin E (MMAE) as the payload. Four bioanalytical assays for total antibodies, conjugated antibodies, conjugated payload, and free payload were then developed and validated for the comprehensive evaluation of the multiple drug forms of 9MW2821. Specific sandwich enzyme-linked immunosorbent assays were used to quantify total antibodies and conjugated antibody, showing good drug-to-antibody ratio (DAR) tolerance. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to determine free MMAE, and conjugated MMAE was quantified using a combination of ligand-binding assay (LBA) and LC-MS/MS. Based on these four assays, we studied the serum stability and monkey pharmacokinetic profiles of 9MW2821, and the in vivo DAR of 9MW2821 was calculated and dynamically monitored. In conclusion, we developed and validated series of bioanalytical assays to quantify multiple forms of 9MW2821, a new ADC, and used the assays to evaluate the serum stability and monkey pharmacokinetic characteristics. The results indicate good linker stability and suggest that the developed assays can be further used in clinical settings.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Immunoconjugates , Oligopeptides , Tandem Mass Spectrometry , Immunoconjugates/pharmacokinetics , Immunoconjugates/chemistry , Immunoconjugates/blood , Animals , Tandem Mass Spectrometry/methods , Oligopeptides/pharmacokinetics , Oligopeptides/chemistry , Oligopeptides/blood , Enzyme-Linked Immunosorbent Assay/methods , Chromatography, Liquid/methods , Humans , Macaca fascicularis , Male , Reproducibility of Results , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics
15.
Jpn J Clin Oncol ; 54(8): 873-879, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38864246

ABSTRACT

BACKGROUND: PET/CT imaging with Zirconium-89 labeled [89Zr]Zr-DFO-girentuximab, which targets tumor antigen CAIX, may aid in the differentiation and characterization of clear cell renal cell carcinomas (RCC) and other renal and extrarenal lesions, and has been studied in European and American cohorts. We report results from a phase I study that evaluated the safety profile, biodistribution, and dosimetry of [89Zr]Zr-DFO-girentuximab in Japanese patients with suspected RCC. METHODS: Eligible adult patients received 37 MBq (± 10%; 10 mg mass dose) of intravenous [89Zr]Zr-DFO-girentuximab. Safety and tolerability profile was assessed based on adverse events, concomitant medications, physical examination, vital signs, hematology, serum chemistry, urinalysis, human anti-chimeric antibody measurement, and 12-lead electrocardiograms at predefined intervals. Biodistribution and normal organ and tumor dosimetry were evaluated with PET/CT images acquired at 0.5, 4, 24, 72 h and Day 5 ± 2 d after administration. RESULTS: [89Zr]Zr-DFO-girentuximab was administered in six patients as per protocol. No treatment-emergent adverse events were reported. Dosimetry analysis showed that radioactivity was widely distributed in the body, and that the absorbed dose in healthy organs was highest in the liver (mean ± standard deviation) (1.365 ± 0.245 mGy/MBq), kidney (1.126 ± 0.190 mGy/MBq), heart wall (1.096 ± 0.232 mGy/MBq), and spleen (1.072 ± 0.466 mGy/MBq). The mean effective dose, adjusted by the radioactive dose administered, was 0.470 mSv/MBq. The radiation dose was highly accumulated in the targeted tumor, while any abnormal accumulation in other organs was not reported. CONCLUSIONS: This study demonstrates that [89Zr]Zr-DFO-girentuximab administered to Japanese patients with suspected RCC has a favorable safety profile and is well tolerated and has a similar dosimetry profile to previously studied populations.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Positron Emission Tomography Computed Tomography , Radioisotopes , Zirconium , Humans , Carcinoma, Renal Cell/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Male , Kidney Neoplasms/diagnostic imaging , Female , Middle Aged , Aged , Zirconium/pharmacokinetics , Radioisotopes/administration & dosage , Radioisotopes/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Tissue Distribution , Adult , Japan , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/administration & dosage , East Asian People
16.
Clin Pharmacol Ther ; 116(3): 847-857, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38863178

ABSTRACT

Data from phase IIb/III and phase III studies were used to characterize the population pharmacokinetics of risankizumab and its exposure-response relationships for efficacy and safety in ulcerative colitis (UC) patients. A two-compartment model with first-order absorption and elimination accurately described risankizumab pharmacokinetics. Although certain covariates, namely, body weight, serum albumin, fecal calprotectin, sex, corticosteroid use, advanced therapy inadequate response, and pancolitis, were statistically correlated with risankizumab clearance, their impact on exposure was not clinically meaningful for efficacy or safety. Phase II exposure-response analyses demonstrated that the 1,200 mg intravenous (IV) induction dose at Weeks 0, 4, and 8 achieved near maximal response for all efficacy end points, with suboptimal efficacy from the 600 mg and little added benefit from the 1,800 mg regimens, justifying 1,200 mg IV as the induction dose in the phase III study. Phase III exposure-response analyses for efficacy during induction showed statistically significant exposure-response relationships at Week 12 following 1,200 mg IV at Weeks 0, 4, and 8, in line with phase IIb results. Exposure-response analyses for maintenance demonstrated modest improvement in Week 52 efficacy when increasing the subcutaneous dose from 180 mg to 360 mg with largely overlapping confidence intervals. Exposure-response analyses for safety indicated no apparent exposure-dependent safety events over the induction or maintenance treatment. Based on these results, the recommended dosing regimen for risankizumab in UC patients is 1,200 mg IV at Weeks 0, 4, and 8, followed by 180 mg or 360 mg subcutaneously at Week 12 and every 8 weeks thereafter.


Subject(s)
Antibodies, Monoclonal , Colitis, Ulcerative , Dose-Response Relationship, Drug , Models, Biological , Humans , Colitis, Ulcerative/drug therapy , Male , Female , Adult , Middle Aged , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Treatment Outcome , Young Adult , Aged , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/adverse effects , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Adolescent
17.
J Pharm Sci ; 113(9): 2879-2894, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38906252

ABSTRACT

In vivo clearance mechanisms of therapeutic monoclonal antibodies (mAbs) encompass both target-mediated and target-independent processes. Two distinct determinants of overall mAb clearance largely separate of target-mediated influences are non-specific cellular endocytosis and subsequent pH-dependent mAb recycling mediated by the neonatal Fc receptor (FcRn), where inter-mAb variability in the efficiency of both processes is observed. Here, we implemented a functional cell-based FcRn recycling assay via Madin-Darby canine kidney type II cells stably co-transfected with human FcRn and its light chain ß2-microglobulin. Next, a series of pH-dependent internalization studies using a model antibody demonstrated proper function of the human FcRn complex. We then applied our cellular assays to assess the contribution of both FcRn and non-specific interactions in the cellular turnover for a panel of 8 clinically relevant mAbs exhibiting variable human pharmacokinetic behavior. Our results demonstrate that the interplay of non-specific endocytosis rates, pH-dependent non-specific interactions, and engagement with FcRn all contribute to the overall recycling efficiency of therapeutic monoclonal antibodies. The predictive capacity of our assay approach was highlighted by successful identification of all mAbs within our panel possessing clearance in humans greater than 5 mL/day/kg. These results demonstrate that a combination of cell-based in vitro assays can properly resolve individual mechanisms underlying the overall in vivo recycling efficiency and non-target mediated clearance of therapeutic mAbs.


Subject(s)
Antibodies, Monoclonal , Endocytosis , Histocompatibility Antigens Class I , Receptors, Fc , Receptors, Fc/metabolism , Receptors, Fc/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/metabolism , Dogs , Animals , Humans , Endocytosis/physiology , Madin Darby Canine Kidney Cells , Hydrogen-Ion Concentration
18.
Anal Methods ; 16(26): 4240-4246, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38780038

ABSTRACT

Daratumumab, a pivotal treatment for multiple myeloma, exhibits considerable inter-patient variability in pharmacological clinical outcomes, likely attributed to serum concentration that may underscore the need for its therapeutic drug monitoring. This study aims to develop and validate a straightforward analytical method for quantifying daratumumab in serum, focusing on intact light chain determination, using liquid chromatography high-resolution mass spectrometry. The sample preparation involved immunoglobulin enrichment using Melon gel followed by a reduction step to dissociate the light from the heavy chains of immunoglobulins. The latter were then separated using a MabPac RP 2.1 × 50 mm chromatographic column and the intact light chains were detected and quantified using a Q Exactive Orbitrap mass spectrometer operating in ESI-positive ion mode at 17 500 resolution. The method demonstrated excellent linearity (R2 > 0.992) across a serum concentration range of 100 to 2000 µg mL-1 and good precision and accuracy: intra- and interday relative errors ranged from -5.1% to 6.5%, with a relative standard deviation of less than 5.8%. Clinical suitability was confirmed by analyzing 80 clinical samples from multiple myeloma patients treated with 1800 mg of daratumumab. 99% of the samples fell within the analytical range with a mean daratumumab concentration evaluated before the next administration (Ctrough) of 398 µg mL-1. These findings highlighted that intact light chain monoclonal antibody quantification could be a valid and robust alternative to either immunoassays or to LC-MS/MS targeting peptides for measuring daratumumab in clinical samples, positioning it as a suitable method for therapeutic drug monitoring applications.


Subject(s)
Antibodies, Monoclonal , Drug Monitoring , Immunoglobulin Light Chains , Multiple Myeloma , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacokinetics , Humans , Drug Monitoring/methods , Multiple Myeloma/drug therapy , Multiple Myeloma/blood , Immunoglobulin Light Chains/blood , Chromatography, Liquid/methods , Mass Spectrometry/methods , Reproducibility of Results , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Chromatography, High Pressure Liquid/methods
19.
Clin Pharmacol Ther ; 116(2): 435-447, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38797892

ABSTRACT

Mirikizumab is a humanized anti-interleukin-23p19 monoclonal antibody being developed for ulcerative colitis (UC) and Crohn's disease. We characterized the relationship of mirikizumab systemic exposure with efficacy and safety end points in patients with UC using phase II (NCT02589665) and III (NCT03518086, NCT03524092) trial data. Exposure-response models were developed for clinical remission, clinical response, endoscopic remission, and change in modified Mayo score following induction (50-1,000 mg i.v. every 4 weeks) and maintenance (200 mg s.c. every 4 or 12 weeks) treatment. These models evaluated observed and pharmacokinetic model-predicted mirikizumab exposures as the exposure measure. Key safety event rates were compared across mirikizumab exposure quartiles in the phase III trial. Mirikizumab efficacy in patients with UC showed an apparent positive association with systemic exposure following both induction and maintenance. However, further analysis found this relationship to be overstated by the presence of confounding factors that were not among the tested patient covariates. While prior biologic experience and baseline disease severity showed statistically significant influences on estimated placebo effect, no patient factors affected the mirikizumab effect parameters in any of the phase III exposure-response models. There was no apparent mirikizumab concentration relationship with any adverse event of special interest. When the phase II and III data and confounding are considered together, efficacy was unlikely to be strongly affected by variation in exposures across individual patients at the phase III dose. Together with the previously demonstrated mirikizumab exposure insensitivity to patient factors, these findings indicate that mirikizumab dose adjustment to patient characteristics is not required.


Subject(s)
Colitis, Ulcerative , Dose-Response Relationship, Drug , Severity of Illness Index , Humans , Colitis, Ulcerative/drug therapy , Male , Female , Adult , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/administration & dosage , Treatment Outcome , Middle Aged , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Agents/therapeutic use , Gastrointestinal Agents/administration & dosage , Interleukin-23 Subunit p19/antagonists & inhibitors , Remission Induction , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage
20.
J Pharmacokinet Pharmacodyn ; 51(4): 307-317, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38700803

ABSTRACT

Pharmacokinetic modeling of monoclonal antibodies (mAbs) with non-linear binding is based on equations of the target-mediated drug disposition (Mager and Jusko, J Pharmacokinet Pharmacodyn 28:507-532, 2001). These equations demonstrated their utility in countless examples and drug development programs. The model assumes that the mAb drug and the target have only one binding site each while, in reality, most antibodies have two binding sites. Thus, the currently used model does not correspond to the biological process that it aims to describe. The correct mechanistic model should take into account both binding sites. We investigated, using simulations, whether this discrepancy is important and when it is advisable to use a model with correct stoichiometric 2-to-1 ratio. We show that for soluble targets when elimination rate of the drug-target complex is comparable with the elimination rate of the drug or lower, and when measurements of both total drug and total target concentrations are available, the model with 1-to-1 (monovalent) binding cannot describe data simulated from the model with 2-to-1 (bivalent) binding. In these cases, models with correct stoichiometric assumptions may be necessary for an adequate description of the observed data. Also, a model with allosteric binding that encompasses both 2-to-1 and 1-to-1 binding models as particular cases was proposed and applied. It was shown to be identifiable given the detailed concentration data of total drug and total target.


Subject(s)
Antibodies, Monoclonal , Models, Biological , Antibodies, Monoclonal/pharmacokinetics , Humans , Computer Simulation , Binding Sites
SELECTION OF CITATIONS
SEARCH DETAIL