Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Pharm Biomed Anal ; 245: 116138, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38636191

RESUMEN

Q-1802 is a humanized bispecific antibody targeting programmed death-ligand 1 (PD-L1) and Claudin 18.2 (CLDN18.2). It can bind to CLDN18.2 and mediate antibody-dependent cell-mediated cytotoxicity against tumor cells. The Fc segment of the antibody recognizing PD-L1 blocks PD-1 signaling and activates innate immunity and adaptive immunity. In this study, we report the development, validation, and application of sensitive and high-throughput enzyme-linked immunosorbent assays (ELISA) to measure the concentrations of Q-1802 in ICR mouse serum. The assay is sensitive, with a lower limit of quantification of 50 ng/mL, has a broad dynamic range of 50-3200 ng/mL, and exhibits excellent precision and accuracy. These assays were 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 Q-1802 in ICR mouse serum. The development and validation steps of assays met the required criteria for validation, which suggested that these can be applied to quantify Q-1802, as well as in PK studies.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática , Ratones Endogámicos ICR , Animales , Ensayo de Inmunoadsorción Enzimática/métodos , Ratones , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Biespecíficos/sangre , Reproducibilidad de los Resultados , Masculino , Humanos , Antígeno B7-H1
2.
Thromb Haemost ; 121(10): 1289-1298, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33641138

RESUMEN

INTRODUCTION: Emicizumab prophylaxis improves coagulation function in congenital hemophilia A, regardless of inhibitor presence. We recently reported that emicizumab enhanced the coagulant potentials, ex vivo, in plasmas from patients with acquired hemophilia A (PwAHAs) at diagnosis. However, coagulant effects of emicizumab in PwAHAs during the clinical course remain unclear. AIM: To assess ex vivo coagulant effects of emicizumab in PwAHAs during the clinical course. METHODS/RESULTS: Blood samples were obtained from 14 PwAHAs on (median) days 0 and 6 during a severe-bleeding phase, and days 27 and 59 during a reduced-bleeding phase with elevated endogenous factor VIII (FVIII) and decreased inhibitor titers. If administered a single dose of 3 or 6 mg/kg, or two doses at 6 mg/kg followed by 3 mg/kg, estimated plasma emicizumab concentrations (10/5/2.5, 20/10/5, and 30/15/7.5 µg/mL on days 0-7/30/60, respectively) could be used to represent potential changes, based on the half-life (T 1/2: ∼30 days). Emicizumab concentrations that covered maximum plasma concentrations of each dosage were used for spiking on day 0. Ex vivo addition of estimated emicizumab to PwAHA's plasma containing endogenous FVIII and/or inhibitor, without and with recombinant (r)FVIIa administration during immunosuppressive therapy, increased the calculated Ad|min1| values, assessed by clot waveform analysis, and their coagulant potentials were below normal levels. Rotational thromboelastometry revealed that ex vivo emicizumab addition resulted in the further improvement of coagulant potentials in whole bloods when combined with rFVIIa administration. CONCLUSION: Based on ex vivo and in vitro data, emicizumab has the potential to be effective in clinical situations for PwAHAs.


Asunto(s)
Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Monoclonales Humanizados/administración & dosificación , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/administración & dosificación , Hemofilia A/tratamiento farmacológico , Hemorragia/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Biespecíficos/sangre , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacocinética , Autoanticuerpos/sangre , Niño , Coagulantes/sangre , Coagulantes/farmacocinética , Monitoreo de Drogas , Factor VIII/inmunología , Femenino , Hemofilia A/sangre , Hemofilia A/inmunología , Hemorragia/sangre , Hemorragia/inmunología , Humanos , Masculino , Persona de Mediana Edad , Tromboelastografía , Adulto Joven
3.
Blood ; 137(3): 410-419, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-32766881

RESUMEN

Acquired hemophilia A (AHA) is a severe bleeding disorder caused by inhibiting autoantibodies to coagulation factor VIII (FVIII). For hemostatic treatment, bypassing agents and human or porcine FVIII are currently standard of care. Emicizumab is a bispecific, FVIII-mimetic therapeutic antibody that reduced the annualized bleeding rates in congenital hemophiliacs. Here, we report on 6 male and 6 female patients with AHA treated with emicizumab (all data medians and interquartile range), age 74 (64-80) years, initial FVIII <1%; inhibitor titer 22.3 Bethesda units (BU)/mL (range, 3-2000). Eight patients had severe bleeding. Emicizumab was started, 3 mg/kg subcutaneously, weekly for 2 to 3 doses, followed by 1.5 mg/kg every 3 weeks to keep the lowest effective FVIII levels. For FVIII monitoring, chromogenic assays with human and bovine reagents were used. All patients received immunosuppression with steroids and/or rituximab. After the first dose of emicizumab, activated partial thromboplastin time normalized in 1 to 3 days, FVIII (human reagents) exceeded 10% after 11 (7.5-12) days. Hemostatic efficacy was obtained and bypassing therapy stopped after 1.5 (1-4) days. FVIII (bovine reagents) exceeded 50%, indicating complete remission after 115 (67-185) days, and emicizumab was stopped after 31 (15-79) days. A median of 5 injections (range, 3-9) were given. No patient died of bleeding or thromboembolism, and no breakthrough bleeding was observed after the first dose of emicizumab. In conclusion, emicizumab seems to be an effective hemostatic therapy for AHA, with the advantages of subcutaneous therapy, good hemostatic efficacy, early discharge, and reduction of immunosuppression and adverse events.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Hemofilia A/tratamiento farmacológico , Anciano , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/sangre , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/sangre , Relación Dosis-Respuesta a Droga , Factor VIII/metabolismo , Femenino , Hemofilia A/sangre , Hemostáticos/uso terapéutico , Humanos , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Inducción de Remisión , Resultado del Tratamiento
4.
Clin Pharmacol Drug Dev ; 10(1): 30-38, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32433829

RESUMEN

This phase 1, open-label, single-center study evaluated the pharmacokinetics (PK), pharmacodynamics, safety, and tolerability of single-dose emicizumab in healthy Chinese males. Overall, 16 subjects received a single subcutaneous dose of 1-mg/kg emicizumab. Blood samples were obtained before dosing on day 1 and at regular intervals over 16 weeks after dosing for PK evaluation. A single 1-mg/kg subcutaneous dose of emicizumab was safe and well tolerated in healthy Chinese male subjects in the study. Mean (± standard deviation) area under the concentration-time curve from time 0 to infinity and maximum concentration were 287 ± 74.2 µg⋅d/mL and 7.11 ± 1.77 µg/mL, respectively, with a terminal half-life of 26.7 (±4.3) days. Emicizumab administration did not show significant impact on pharmacodynamic markers tested, which mostly remained stable throughout the study. One subject tested positive for antidrug antibody, with no impact on his PK or safety profile. Compared with results from healthy Japanese and Caucasian subjects receiving the same dose in previous clinical trials, the current results further indicated the absence of difference of emicizumab PK profile across Chinese, Japanese, and Caucasian subjects, validating the use of similar therapeutic doses in Asian and non-Asian populations.


Asunto(s)
Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales Humanizados/farmacocinética , Adulto , Anticuerpos/sangre , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/sangre , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacología , Pueblo Asiatico , Factor VIII/análisis , Voluntarios Sanos , Humanos , Inyecciones Subcutáneas , Masculino , Población Blanca , Adulto Joven
5.
Assay Drug Dev Technol ; 19(1): 17-26, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33232610

RESUMEN

B cell maturation antigen (BCMA) is a membrane-bound receptor that is overexpressed on multiple myeloma cells and can be targeted with biotherapeutics. Soluble shed forms of membrane-associated receptors in circulation can act as a drug sink, especially when it is present in high molar ratio compared to drug concentration, potentially derailing the intended pharmacological mechanism and impacting pharmacokinetic (PK) measurements and efficacious dose predictions. In this study, we present a bioanalytical strategy for assessing dynamic levels of total soluble BCMA before and during treatment with a bispecific antibody targeting BCMA and CD3. Implementation of a ligand binding assay was not successful due to extensive bispecific antibody interference. Instead, we explored two types of immunoaffinity (IA) liquid chromatography-tandem mass spectrometry (LC-MS/MS) assays, one at the protein level and one at the surrogate peptide level. Ultimately, the protein-level IA-LC-MS/MS method was optimized for use in a cynomolgus monkey PK/pharmacodynamic study. In addition, we demonstrated that the method was easily adapted for use with human samples in preparation for translation to the clinic. This work demonstrates the benefit of flexibility and agility in bioanalytical method development in early drug development. Multiplatform suitability assessments enable rapid, resource-sparing identification and qualification of clinically translatable assays. We recommend early adoption of this strategy to provide enough time for critical reagent development and assay validation for analysis of shed targets.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antígeno de Maduración de Linfocitos B/sangre , Animales , Anticuerpos Biespecíficos/sangre , Antígeno de Maduración de Linfocitos B/inmunología , Cromatografía Liquida , Humanos , Macaca fascicularis , Espectrometría de Masas en Tándem
6.
Clin Chem Lab Med ; 59(2): 365-371, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32892172

RESUMEN

Objectives: Emicizumab, a monoclonal antibody mimicking the function of factor (F) VIII in the activation of FX by FIXa, is widely used for prophylaxis in hemophilia patients with or without inhibitors to FVIII. Although it is administered at fixed dose, its measurement could be occasionally required. In principle, the emicizumab procoagulant effect could be assessed by the one-stage assay (OSA) currently used to measure FVIII. However, the OSA for FVIII presents with limitations. Furthermore, owing to its potent FVIII-like activity, emicizumab interferes with the measurement of the inhibitor to FVIII, which is often needed in patients on emicizumab. Methods: We prepared test samples by spiking a FVIII-deficient plasma with graded amounts of emicizumab. We modified the OSA for FVIII and tested plasma samples for emicizumab concentrations. Furthermore the chromogenic assay (CA) for FVIII with bovine reagents was used to assess for the FVIII inhibitor in patients on emicizumab. Results: Slight modification of the OSA for FVIII (i.e., higher test plasma dilution and longer coagulometer acquisition time) made the regular OSA as a reliable laboratory tool to measure emicizumab concentration as shown by the identity of the regression (observed vs. expected) lines. Furthermore, the inhibitors to FVIII in patients on emicizumab, which were negative when measured by the regular Bethesda assay, were reliably measured by the CA assay employing bovine reagents. Conclusions: The methods currently used to measure FVIII can be easily modified to make the general clinical laboratory able to assist clinicians when dealing with patients on emicizumab.


Asunto(s)
Anticuerpos Biespecíficos/sangre , Anticuerpos Monoclonales Humanizados/sangre , Coagulantes/sangre , Factor VIII/metabolismo , Hemofilia A/diagnóstico , Anciano , Animales , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales Humanizados/metabolismo , Unión Competitiva , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Recolección de Muestras de Sangre , Bovinos , Niño , Coagulantes/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Persona de Mediana Edad , Tiempo de Tromboplastina Parcial , Estándares de Referencia , Reproducibilidad de los Resultados
7.
Bioanalysis ; 12(8): 509-517, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32351119

RESUMEN

Aim: The work was aimed at developing a bioanalytical approach to identify immunogenic parts of a bispecific F(ab) fragment and to characterize the immune response seen in a preclinical study. Experimental: The bioanalytical method consists of a set of domain detection assays that use germlined variants of the drug. Results: The method demonstrated that anti-drug antibodies (ADAs) were predominantly directed against both antigen-binding sites of the drug. Conclusion: The method was capable to discriminate between ADAs directed against one of the antigen-binding sites, both sites or the constant domain, allowing for an estimation of the relative binding prevalence for these subunits. The developed approach provides a practical and robust solution for exploratory characterization of ADAs against multidomain biotherapeutics.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/inmunología , Bioensayo , Preparaciones Farmacéuticas/química , Animales , Anticuerpos Biespecíficos/sangre , Anticuerpos Monoclonales/sangre , Reacciones Antígeno-Anticuerpo , Macaca fascicularis , Preparaciones Farmacéuticas/sangre
8.
Proc Natl Acad Sci U S A ; 117(18): 9851-9856, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32327606

RESUMEN

Toward the goal of increasing the throughput of high-resolution mass characterization of intact antibodies, we developed a RapidFire-mass spectrometry (MS) assay using electrospray ionization. We achieved unprecedented screening throughput as fast as 15 s/sample, which is an order of magnitude improvement over conventional liquid chromatography (LC)-MS approaches. The screening enabled intact mass determination as accurate as 7 ppm with baseline resolution at the glycoform level for intact antibodies. We utilized this assay to characterize and perform relative quantitation of antibody species from 248 samples of 62 different cell line clones at four time points in 2 h using RapidFire-time-of-flight MS screening. The screening enabled selection of clones with the highest purity of bispecific antibody production and the results significantly correlated with conventional LC-MS results. In addition, analyzing antibodies from a complex plasma sample using affinity-RapidFire-MS was also demonstrated and qualified. In summary, the platform affords high-throughput analyses of antibodies, including bispecific antibodies and potential mispaired side products, in cell culture media, or other complex matrices.


Asunto(s)
Anticuerpos Biespecíficos/sangre , Anticuerpos/sangre , Ensayos Analíticos de Alto Rendimiento/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos , Anticuerpos/aislamiento & purificación , Anticuerpos Biespecíficos/aislamiento & purificación , Línea Celular , Cromatografía Liquida/métodos , Humanos
9.
Cancer Sci ; 111(4): 1314-1323, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31971321

RESUMEN

Adult patients with relapsed/refractory (R/R) B-precursor acute lymphoblastic leukemia (ALL) have a poor prognosis. Blinatumomab is a bispecific T-cell engager (BiTE) immuno-oncology therapy with dual specificity for CD19 and CD3 that redirects patients' CD3-positive cytotoxic T cells to lyse malignant and normal B cells. We conducted an open-label, phase 1b/2 study to determine the safety, pharmacokinetics, efficacy and recommended dose of blinatumomab in Japanese adults with R/R B-precursor ALL. Patients received 9 µg/day blinatumomab during week 1 and 28 µg/day during weeks 2-4, with a 2-week treatment-free interval (6-week cycle); patients received 28 µg/day blinatumomab in subsequent cycles. Primary endpoints were the incidence of dose-limiting toxicities (DLT) in phase 1b and complete remission (CR)/CR with partial hematologic recovery (CRh) within the first two cycles in phase 2. A total of 26 patients enrolled and 25 (96%) reported grade ≥3 adverse events (mostly cytopenias). There were no DLT. CR/CRh within two cycles was achieved by 4 of 5 patients (80%) in phase 1b and 8 of 21 patients (38%) in phase 2. Among patients with evaluable minimal residual disease, 4 (100%) in phase 1b and 3 (38%) in phase 2 had a complete MRD response. Median RFS for 8 patients who achieved CR/CRh in phase 2 was 5 (95% CI: 3.5-6.4) months; median OS was not estimable. There were no significant associations between maximum cytokine levels or percentage of specific cell types during cycle 1 and response. Consistent with global studies, blinatumomab appeared to be safe and efficacious in Japanese adults with R/R ALL.


Asunto(s)
Anticuerpos Biespecíficos/administración & dosificación , Linfoma de Células B/tratamiento farmacológico , Neoplasia Residual/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/sangre , Anticuerpos Biespecíficos/farmacocinética , Antígenos CD19/genética , Antígenos CD19/inmunología , Linfocitos B/patología , Complejo CD3/genética , Complejo CD3/inmunología , Dexametasona/administración & dosificación , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Interferón gamma/sangre , Estimación de Kaplan-Meier , Linfoma de Células B/sangre , Linfoma de Células B/patología , Masculino , Persona de Mediana Edad , Neoplasia Residual/sangre , Neoplasia Residual/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangre , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Inducción de Remisión , Linfocitos T/inmunología
10.
Clin Pharmacokinet ; 59(4): 463-474, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31679130

RESUMEN

BACKGROUND AND OBJECTIVES: Blinatumomab (BLINCYTO®) is a novel bispecific T cell engager (BiTE®) approved in the USA for the treatment of relapsed or refractory B cell precursor acute lymphoblastic leukemia (ALL) in children and adults, as well as minimal residual disease ALL in adults. This analysis characterized the population pharmacokinetics of intravenous blinatumomab in pediatric and adult patients. METHODS: A total of 2417 serum concentrations of blinatumomab from 674 patients, including adult (n = 628) and pediatric patients (n = 46), from eight clinical studies were analyzed. The impact of covariates on pharmacokinetic parameters were explored, and significant covariates were further evaluated using a simulation approach. RESULTS: Blinatumomab pharmacokinetics were described by a one-compartment linear model with first-order elimination, a clearance (CL) of 2.22 L/h, and a central volume of 5.98 L. A statistically significant effect of body surface area (BSA) on CL was observed. The smallest BSA of 0.37 m2 in the pediatric population was associated with a 63% reduction in blinatumomab systemic CL, relative to an adult patient with the median BSA (1.88 m2), supporting the use of BSA-based dosing in patients of lower bodyweight. The BSA effect was minimal, with a ≤ 25% change in CL over the range of BSA in adults, supporting no need for BSA-based dosing. CONCLUSIONS: Blinatumomab pharmacokinetics were adequately described by a one-compartment linear model with first-order elimination. No covariates other than BSA on CL were identified as significant. BSA-based dosing should be considered for lightweight patients to minimize inter-subject variability in blinatumomab exposure.


Asunto(s)
Anticuerpos Biespecíficos/farmacocinética , Antineoplásicos/farmacocinética , Neoplasias Hematológicas/tratamiento farmacológico , Linfoma no Hodgkin/tratamiento farmacológico , Neoplasia Residual/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Administración Intravenosa , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/sangre , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Superficie Corporal , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Simulación de Paciente , Prevención Secundaria , Linfocitos T/efectos de los fármacos , Adulto Joven
11.
Immunogenetics ; 71(10): 605-615, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31776588

RESUMEN

Immunogenicity of biomolecules is one of the largest concerns in biological therapeutic drug development. Adverse immune responses as a result of immunogenicity to biotherapeutics range from mild hypersensitivity reactions to potentially life-threatening anaphylactic reactions and can negatively impact human health and drug efficacy. Numerous confounding patient-, product- or treatment-related factors can influence the development of an immune reaction against therapeutic proteins. The goal of this study was to investigate the relationship between pre-existing drug reactivity (PE-ADA), individual immunogenetics (MHC class II haplotypes), and development of treatment-induced antidrug antibodies (TE-ADA) in cynomolgus macaque. PE-ADA refers to the presence of antibodies immunoreactive against the biotherapeutic in treatment-naïve individuals. We observed that PE-ADA frequency against four different bispecific antibodies in naïve cynomolgus macaque is similar to that reported in humans. Additionally, we report a trend towards an increased incidence of TE-ADA development in macaques with high PE-ADA levels. In order to explore the relationship between MHC class II alleles and risk of ADA development, we obtained full-length MHC class II sequences from 60 cynomolgus macaques in our colony. We identified a total of 248 DR, DP, and DQ alleles and 236 unique haplotypes in our cohort indicating a genetically complex set of animals potentially reflective of the human population. Based on our observations, we propose the evaluation of the magnitude/frequency of pre-existing reactivity and consideration of MHC class II genetics as additional useful tools to understand the immunogenic potential of biotherapeutics.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Hipersensibilidad a las Drogas/inmunología , Genes MHC Clase II/genética , Genes MHC Clase II/inmunología , Inmunogenética , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/sangre , Hipersensibilidad a las Drogas/genética , Frecuencia de los Genes , Haplotipos , Macaca fascicularis , Masculino , Filogenia
12.
Methods Mol Biol ; 2024: 153-166, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31364048

RESUMEN

Antibody-based therapeutics have emerged as novel class of biopharmaceuticals over the last couple of decades with the advancements made in production and downstream processing technologies. The structural diversity of therapeutic antibodies has also evolved with the development of bispecific (and multispecific) antibodies and antibody-drug conjugates. With increased structural complexities and multi-modularity, there is a need to demonstrate that the entire structure is stable in vivo and arriving at its target site in an intact form. Proving that antibodies reach their target site unscathed is a challenging but essential step for showing effective delivery as well as showing whether failure in efficacy (if any) was related to its in vivo instability. This chapter describes a method for highly specific immuno-isolation followed by intact mass spectrometry of human Fc-containing antibody from serum of rats dosed with the antibody. The method provides an opportunity for evaluating antibody stability in the physiological environment by providing accurate validation of its molecular mass in vivo, as well as the potential to identify breakdown products.


Asunto(s)
Anticuerpos Biespecíficos/sangre , Fragmentos Fc de Inmunoglobulinas/sangre , Espectrometría de Masas/métodos , Animales , Cromatografía Liquida/métodos , Humanos , Ratas
13.
J Proteome Res ; 18(8): 3032-3041, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31267741

RESUMEN

Bispecific single chain antibody fragments (bi-scFv) represent an emerging class of biotherapeutics. We recently developed a fully human bi-scFv (EGFRvIII:CD3 bi-scFv) with the goal of redirecting CD3-expressing T cells to recognize and destroy malignant, EGFRvIII-expressing glioma. In mice, we showed that EGFRvIII:CD3 bi-scFv effectively treats orthotopic patient-derived malignant glioma and syngeneic glioblastoma. Here, we developed a targeted assay for pharmacokinetic (PK) analysis of EGFRvIII:CD3 bi-scFv, a necessary step in the drug development process. Using microflow liquid chromatography coupled to a high resolution parallel reaction monitoring mass spectrometry, and data analysis in Skyline, we developed a bottom-up proteomic assay for quantification of EGFRvIII:CD3 bi-scFv in both plasma and whole blood. Importantly, a protein calibrator, along with stable isotope-labeled EGFRvIII:CD3 bi-scFv protein, were used for absolute quantification. A PK analysis in a CD3 humanized mouse revealed that EGFRvIII:CD3 bi-scFv in plasma and whole blood has an initial half-life of ∼8 min and a terminal half-life of ∼2.5 h. Our results establish a sensitive, high-throughput assay for direct quantification of EGFRvIII:CD3 bi-scFv without the need for immunoaffinity enrichment. Moreover, these pharmacokinetic parameters will guide drug optimization and dosing regimens in future IND-enabling and phase I studies of EGFRvIII:CD3 bi-scFv.


Asunto(s)
Anticuerpos Biespecíficos/sangre , Complejo CD3/sangre , Receptores ErbB/sangre , Glioblastoma/sangre , Animales , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Biespecíficos/uso terapéutico , Complejo CD3/farmacocinética , Complejo CD3/uso terapéutico , Línea Celular Tumoral , Cromatografía Liquida , Receptores ErbB/farmacocinética , Receptores ErbB/uso terapéutico , Glioblastoma/inmunología , Glioblastoma/terapia , Humanos , Espectrometría de Masas , Ratones , Proteómica/métodos , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
AAPS J ; 21(4): 66, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31119428

RESUMEN

CD3 bispecific antibody constructs recruit cytolytic T cells to kill tumor cells, offering a potent approach to treat cancer. T cell activation is driven by the formation of a trimolecular complex (trimer) between drugs, T cells, and tumor cells, mimicking an immune synapse. A translational quantitative systems pharmacology (QSP) model is proposed for CD3 bispecific molecules capable of predicting trimer concentration and linking it to tumor cell killing. The model was used to quantify the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a CD3 bispecific targeting P-cadherin (PF-06671008). It describes the disposition of PF-06671008 in the central compartment and tumor in mouse xenograft models, including binding to target and T cells in the tumor to form the trimer. The model incorporates T cell distribution to the tumor, proliferation, and contraction. PK/PD parameters were estimated for PF-06671008 and a tumor stasis concentration (TSC) was calculated as an estimate of minimum efficacious trimer concentration. TSC values ranged from 0.0092 to 0.064 pM across mouse tumor models. The model was translated to the clinic and used to predict the disposition of PF-06671008 in patients, including the impact of binding to soluble P-cadherin. The predicted terminal half-life of PF-06671008 in the clinic was approximately 1 day, and P-cadherin expression and number of T cells in the tumor were shown to be sensitive parameters impacting clinical efficacy. A translational QSP model is presented for CD3 bispecific molecules, which integrates in silico, in vitro and in vivo data in a mechanistic framework, to quantify and predict efficacy across species.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Complejo CD3/inmunología , Cadherinas/metabolismo , Modelos Biológicos , Animales , Anticuerpos Biespecíficos/sangre , Anticuerpos Biespecíficos/farmacocinética , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Células HCT116 , Humanos , Inmunoterapia , Activación de Linfocitos , Macaca fascicularis , Ratones SCID , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Investigación Biomédica Traslacional , Ensayos Antitumor por Modelo de Xenoinjerto
15.
MAbs ; 11(5): 861-869, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31099718

RESUMEN

Despite recent advances in the development of tools to predict immunogenicity risk of biotherapeutic molecules, the ability of a protein to elicit the formation of anti-drug antibodies (ADA) remains one of the most common causes for termination of clinical development programs. In this study, we use ADA assays to detect and measure pre-existing reactivity or the ability of a molecule to produce an ADA-like response in serum from treatment-naïve, healthy donors. We report herein that the magnitude of pre-existing reactivity evaluated pre-clinically and expressed as the 90th percentile of Tier 2 inhibition correlates with the subsequent rate of ADA emergence in the clinic. Furthermore, a multi-domain biotherapeutic (IgG-scFv bispecific antibody) showed the highest pre-existing reactivity and incidence of treatment-emergent ADA (TE-ADA) (57% and 93%, respectively). Using the components of the multidomain molecule in the Tier 2 step of the ADA assay, we were able to identify the scFv as the target of the serum pre-existing reactivity. Most importantly, the domain specificity of pre-existing ADA was the same as that of the TE-ADA from patients treated with the molecule. Based on these data, we propose the evaluation of the magnitude and of the domain specificity of pre-existing reactivity as a powerful tool to understand the immunogenic potential of novel biotherapeutics.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos de Cadena Única/inmunología , Adulto , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/sangre , Formación de Anticuerpos , Terapia Biológica/efectos adversos , Epítopos/inmunología , Femenino , Humanos , Sueros Inmunes/inmunología , Masculino , Persona de Mediana Edad , Riesgo , Anticuerpos de Cadena Única/efectos adversos , Anticuerpos de Cadena Única/sangre , Adulto Joven
16.
Cancer Chemother Pharmacol ; 82(3): 407-418, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29926131

RESUMEN

PURPOSE: The phase I study characterized the safety, pharmacokinetics, anti-tumor activity, and recommended phase II dose/schedule of LY3164530 in patients with advanced or metastatic cancer. METHODS: Patients received LY3164530 on days 1 and 15 (Schedule 1: 300, 600, 1000, and 1250 mg) or Days 1, 8, 15, and 22 (Schedule 2: 500 and 600 mg) of each 28 days cycle. Dose escalation used a modified toxicity probability interval model. RESULTS: Dose escalation defined a maximum tolerated dose (MTD) of 1000 mg on Schedule 1 and 500 mg on Schedule 2. Treatment-emergent adverse events related to study treatment were consistent with epidermal growth factor receptor (EGFR) inhibition and included maculopapular rash/dermatitis acneiform (83%, Grade 3/4 17%), hypomagnesemia (55%, Grade 3/4 7%), paronychia (35%), fatigue (28%, Grade 3/4 3%), skin fissures (24%), and hypokalemia (21%, Grade 3/4 7%). Partial response was achieved in three patients on Schedule 2 with colorectal cancer (n = 2) or squamous cell cancer. Overall response rate (ORR) was 10.3%, disease control rate (ORR + stable disease [SD]) was 51.7 and 17.2% of patients had SD ≥ 4 months. The in vivo stability of the bispecific antibody was confirmed. Schedule 2 provided greater and more consistent inhibition of mesenchymal-epithelial transition (MET)/EGFR throughout the dosing interval than Schedule 1. CONCLUSIONS: Although this study defined the LY3164530 MTD and pharmacokinetics on both schedules, significant toxicities associated with EGFR inhibition and lack of a potential predictive biomarker limit future development. Nonetheless, the results provide insight into the development of bispecific antibody therapy.


Asunto(s)
Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/efectos adversos , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Adulto , Anciano , Anticuerpos Biespecíficos/sangre , Biomarcadores de Tumor/análisis , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/sangre , Receptores ErbB/inmunología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias/sangre , Neoplasias/enzimología , Neoplasias/patología , Proteínas Proto-Oncogénicas c-met/sangre , Proteínas Proto-Oncogénicas c-met/inmunología
17.
J Thromb Haemost ; 16(7): 1383-1390, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29734520

RESUMEN

Essentials Emicizumab (Emi) affects the APTT-based assays of factor (F)VIII activity and inhibitor titer. A mixture of two anti-Emi monoclonal antibodies (mAb) effectively neutralized the Emi activity. Anti-Emi mAbs completely eliminated the influence of Emi on FVIII activity and inhibitor titer. The inclusion of anti-Emi mAbs in routine FVIII assays would be useful for Emi-treated patients. SUMMARY: Background Emicizumab is an anti-factor (F)IXa/X bispecific monoclonal antibody (mAb), mimicking the factor (F)VIIIa cofactor activity. Emicizumab does not require activation by thrombin and its shortening effect on the activated partial prothrombin time (APTT) is more pronounced than that of factor (F)VIII. APTT-based FVIII activity (FVIII:C) and FVIII inhibiter titer measurements are influenced by the presence of emicizumab. Aim To establish a reliable APTT-based assay to measure FVIII in the presence of emicizumab. Methods Plasmas from hemophilia A (HA) patients without or with inhibitors were studied using one-stage FVIII:C and Bethesda inhibitor assays. Two recombinant anti-idiotype mAbs to emicizumab (anti-emicizumab mAbs) were prepared, rcAQ8 to anti-FIXa-Fab and rcAJ540 to anti-FX-Fab. Results The combined anti-idiotype mAbs (2000 nm each) eliminated the effects of emicizumab on APTTs of HA plasmas without or with inhibitor by competitive inhibition of antibody binding to FIX(a)/FX(a). Measurements of FVIII coagulation activity in HA plasmas without inhibitor were overestimated in the presence of emicizumab (1 µm = ~150 µg mL-1 ) at all reference levels of FVIII. The addition of anti-emicizumab mAbs to the assay mixtures completely neutralized the emicizumab and facilitated accurate determination of FVIII:C. Anti-FVIII inhibitor titers were undetectable in the presence of emicizumab in HA plasmas with inhibitor or normal plasmas mixed with anti-FVIII neutralizing antibodies. These effects of emicizumab were completely counteracted by the addition of the anti-idiotype mAbs, allowing accurate assessment of inhibitor titers. Conclusion The in vitro inclusion of anti-emicizumab mAbs in the standard one-stage coagulation assays prevented interference by emicizumab and enabled accurate measurements of FVIII:C and inhibitor titers.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/sangre , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Factor VIII/análisis , Hemofilia A/sangre , Tiempo de Tromboplastina Parcial , Anticuerpos Biespecíficos/sangre , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/inmunología , Unión Competitiva , Coagulantes/sangre , Coagulantes/inmunología , Relación Dosis-Respuesta a Droga , Factor IXa/inmunología , Factor IXa/metabolismo , Factor VIII/inmunología , Factor Xa/inmunología , Factor Xa/metabolismo , Hemofilia A/diagnóstico , Hemofilia A/inmunología , Humanos , Valor Predictivo de las Pruebas , Unión Proteica , Reproducibilidad de los Resultados
18.
Thromb Haemost ; 117(7): 1348-1357, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28451690

RESUMEN

Emicizumab, a humanised bispecific antibody recognising factors (F) IX/IXa and X/Xa, can accelerate FIXa-catalysed FX activation by bridging FIXa and FX in a manner similar to FVIIIa. However, details of the emicizumab-antigen interactions have not been reported so far. In this study, we first showed by surface plasmon resonance analysis that emicizumab bound FIX, FIXa, FX, and FXa with moderate affinities (KD = 1.58, 1.52, 1.85, and 0.978 µM, respectively). We next showed by immunoblotting analysis that emicizumab recognised the antigens' epidermal growth factor (EGF)-like domains. We then performed KD-based simulation of equilibrium states in plasma for quantitatively predicting the ways that emicizumab would interact with the antigens. The simulation predicted that only a small part of plasma FIX, FX, and emicizumab would form antigen-bridging FIX-emicizumab-FX ternary complex, of which concentration would form a bell-shaped relationship with emicizumab concentration. The bell-shaped concentration dependency was reproduced by plasma thrombin generation assays, suggesting that the plasma concentration of the ternary complex would correlate with emicizumab's cofactor activity. The simulation also predicted that at 10.0-100 µg/ml of emicizumab-levels shown in a previous study to be clinically effective-the majority of plasma FIX, FX, and emicizumab would exist as monomers. In conclusion, emicizumab binds FIX/FIXa and FX/FXa with micromolar affinities at their EGF-like domains. The KD-based simulation predicted that the antigen-bridging ternary complex formed in circulating plasma would correlate with emicizumab's cofactor activity, and the majority of FIX and FX would be free and available for other coagulation reactions.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Factor VIIIa/inmunología , Anticuerpos Biespecíficos/sangre , Anticuerpos Monoclonales Humanizados/sangre , Especificidad de Anticuerpos , Reacciones Antígeno-Anticuerpo , Sitios de Unión , Materiales Biomiméticos/farmacología , Simulación por Computador , Factor IX/antagonistas & inhibidores , Factor IX/inmunología , Factor IXa/antagonistas & inhibidores , Factor IXa/inmunología , Factor X/antagonistas & inhibidores , Factor X/inmunología , Factor Xa/inmunología , Inhibidores del Factor Xa/sangre , Inhibidores del Factor Xa/inmunología , Inhibidores del Factor Xa/farmacología , Humanos , Modelos Inmunológicos
19.
MAbs ; 8(7): 1319-1335, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27532598

RESUMEN

An antibody format, termed Fab-dsFv, has been designed for clinical indications that require monovalent target binding in the absence of direct Fc receptor (FcR) binding while retaining substantial serum presence. The variable fragment (Fv) domain of a humanized albumin-binding antibody was fused to the C-termini of Fab constant domains, such that the VL and VH domains were individually connected to the Cκ and CH1 domains by peptide linkers, respectively. The anti-albumin Fv was selected for properties thought to be desirable to ensure a durable serum half-life mediated via FcRn. The Fv domain was further stabilized by an inter-domain disulfide bond. The bispecific format was shown to be thermodynamically and biophysically stable, and retained good affinity and efficacy to both antigens simultaneously. In in vivo studies, the serum half-life of Fab-dsFv, 2.6 d in mice and 7.9 d in cynomolgus monkeys, was equivalent to Fab'-PEG.


Asunto(s)
Anticuerpos Biespecíficos/sangre , Fragmentos Fab de Inmunoglobulinas , Región Variable de Inmunoglobulina , Albúmina Sérica/metabolismo , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/inmunología , Semivida , Humanos , Fragmentos Fab de Inmunoglobulinas/sangre , Fragmentos Fab de Inmunoglobulinas/química , Región Variable de Inmunoglobulina/sangre , Región Variable de Inmunoglobulina/química , Ratones , Albúmina Sérica/inmunología
20.
N Engl J Med ; 374(21): 2044-53, 2016 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-27223146

RESUMEN

BACKGROUND: In patients with severe hemophilia A, standard treatment is regular prophylactic and episodic intravenous infusions of factor VIII. However, these treatments are burdensome, especially for children, and may lead to the formation of anti-factor VIII alloantibodies (factor VIII inhibitors). Emicizumab (ACE910), a humanized bispecific antibody mimicking the cofactor function of factor VIII, was developed to abate these problems. METHODS: We enrolled 18 Japanese patients with severe hemophilia A (with or without factor VIII inhibitors) in an open-label, nonrandomized, interindividual dose-escalation study of emicizumab. The patients received subcutaneous emicizumab weekly for 12 weeks at a dose of 0.3, 1.0, or 3.0 mg per kilogram of body weight (cohorts 1, 2, and 3, respectively). The end points were safety and pharmacokinetic and pharmacodynamic profiles. An additional, exploratory end point was the annualized bleeding rate, calculated as 365.25 times the number of bleeding episodes, divided by the number of days in the treatment period as compared with the 6 months before enrollment. RESULTS: Emicizumab was associated with neither serious adverse events nor clinically relevant coagulation abnormalities. Plasma concentrations of emicizumab increased in a dose-dependent manner. Activated partial-thromboplastin times remained short throughout the study. The median annualized bleeding rates in cohorts 1, 2, and 3 decreased from 32.5 to 4.4, 18.3 to 0.0, and 15.2 to 0.0, respectively. There was no bleeding in 8 of 11 patients with factor VIII inhibitors (73%) and in 5 of 7 patients without factor VIII inhibitors (71%). Episodic use of clotting factors to control bleeding was reduced. Antibodies to emicizumab did not develop. CONCLUSIONS: Once-weekly subcutaneous administration of emicizumab markedly decreased the bleeding rate in patients who had hemophilia A with or without factor VIII inhibitors. (Funded by Chugai Pharmaceutical; JapicCTI number, 121934.).


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor VIII/antagonistas & inhibidores , Hemofilia A/tratamiento farmacológico , Hemorragia/prevención & control , Adolescente , Adulto , Anticuerpos Biespecíficos/sangre , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacología , Niño , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Factor IX/análisis , Factor VIII/uso terapéutico , Factor X/análisis , Hemofilia A/complicaciones , Hemofilia A/inmunología , Hemorragia/etiología , Humanos , Inyecciones Subcutáneas/efectos adversos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...