Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Pharmacol Exp Ther ; 379(2): 134-146, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34413198

RESUMEN

Conjugation of small interfering RNA (siRNA) to tris N-acetylgalactosamine [(GalNAc)3] can enable highly selective, potent, and durable knockdown of targeted proteins in the liver. However, potential knowledge gaps between in vitro experiments, preclinical species, and clinical scenarios remain. A minimal physiologically based pharmacokinetic-pharmacodynamic model for GalNAc-conjugated siRNA (GalNAc-siRNA) was developed using published data for fitusiran (ALN-AT3), an investigational compound targeting liver antithrombin (AT), to delineate putative determinants governing the whole-body-to-cellular pharmacokinetic (PK) and pharmacodynamic (PD) properties of GalNAc-siRNA and facilitate preclinical-to-clinical translation. The model mathematically linked relevant mechanisms: 1) hepatic biodistribution, 2) tris-GalNAc binding to asialoglycoprotein receptors (ASGPRs) on hepatocytes, 3) ASGPR endocytosis and recycling, 4) endosomal transport and escape of siRNA, 5) cytoplasmic RNA-induced silencing complex (RISC) loading, 6) degradation of target mRNA by bound RISC, and 7) knockdown of protein. Physiologic values for 36 out of 48 model parameters were obtained from the literature. Kinetic parameters governing (GalNAc)3-ASGPR binding and internalization were derived from published studies of uptake in hepatocytes. The proposed model well characterized reported pharmacokinetics, RISC dynamics, and knockdown of AT mRNA and protein by ALN-AT3 in mice. The model bridged multiple PK-PD data sets in preclinical species (mice, rat, monkey) and successfully captured reported plasma pharmacokinetics and AT knockdown in a phase I ascending-dose study. Estimates of in vivo potency were similar (∼2-fold) across species. Subcutaneous absorption and serum AT degradation rate constants scaled across species by body weight with allometric exponents of -0.29 and -0.22. The proposed mechanistic modeling framework characterizes the unique PK-PD properties of GalNAc-siRNA. SIGNIFICANCE STATEMENT: Tris N-acetylgalactosamine (GalNAc)3-conjugated small interfering RNA (siRNA) therapeutics enable liver-targeted gene therapy and precision medicine. Using a translational and systems-based minimal physiologically based pharmacokinetic-pharmacodynamic (mPBPK-PD) modeling approach, putative determinants influencing GalNAc-conjugated siRNA (GalNAc-siRNA) functionality in three preclinical species and humans were investigated. The developed model successfully integrated and characterized relevant published in vitro-derived biomeasures, mechanistic PK-PD profiles in animals, and observed clinical PK-PD responses for an investigational GalNAc-siRNA (fitusiran). This modeling effort delineates the disposition and liver-targeted pharmacodynamics of GalNAc-siRNA.


Asunto(s)
Acetilgalactosamina/farmacocinética , Silenciador del Gen/fisiología , Modelos Biológicos , ARN Interferente Pequeño/farmacocinética , Acetilgalactosamina/genética , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Haplorrinos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ratones , ARN Interferente Pequeño/genética , Ratas , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
2.
Diabetes Obes Metab ; 22(12): 2437-2450, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33463043

RESUMEN

AIM: The aim of this study was to evaluate amino acids as glucagon receptor (GCGR)-specific biomarkers in rodents and cynomolgus monkeys in the presence of agonism of both glucagon-like peptide-1 receptor (GLP1R) and GCGR with a variety of dual agonist compounds. MATERIALS AND METHODS: Primary hepatocytes, rodents (normal, diet-induced obese and GLP1R knockout) and cynomolgus monkeys were treated with insulin (hepatocytes only), glucagon (hepatocytes and cynomolgus monkeys), the GLP1R agonist, dulaglutide, or a variety of dual agonists with varying GCGR potencies. RESULTS: A long-acting dual agonist, Compound 2, significantly decreased amino acids in both wild-type and GLP1R knockout mice in the absence of changes in food intake, body weight, glucose or insulin, and increased expression of hepatic amino acid transporters. Dulaglutide, or a variant of Compound 2 lacking GCGR agonism, had no effect on amino acids. A third variant with ~31-fold less GCGR potency than Compound 2 significantly decreased amino acids, albeit to a significantly lesser extent than Compound 2. Dulaglutide (with saline infusion) had no effect on amino acids, but an infusion of glucagon dose-dependently decreased amino acids on the background of GLP1R engagement (dulaglutide) in cynomolgus monkeys, as did Compound 2. CONCLUSIONS: These results show that amino acids are sensitive and translatable GCGR-specific biomarkers.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón , Receptores de Glucagón , Aminoácidos , Animales , Biomarcadores , Glucagón , Ratones , Ratones Endogámicos C57BL , Receptores de Glucagón/genética
3.
Br J Clin Pharmacol ; 81(1): 137-47, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26302359

RESUMEN

AIMS: Evidence suggests that the rate of oral drug absorption changes during early childhood. Yet, respective clinical implications are currently unclear, particularly for preterm neonates. The objective of this study was to evaluate changes in oral drug absorption after birth for different Biopharmaceutics Classification System (BCS) class I and II compounds to better understand respective implications for paediatric pharmacotherapy. METHODS: Two paradigm compounds were selected for BCS class I (paracetamol (acetaminophen) and theophylline) and II (indomethacin and ibuprofen), respectively, based on the availability of clinical literature data following intravenous and oral dosing. A comparative population pharmacokinetic analysis was performed in a step-wise manner in NONMEM® 7.2 to characterize and predict changes in oral drug absorption after birth for paracetamol, theophylline and indomethacin. RESULTS: A one compartment model with an age-dependent maturation function for oral drug absorption was found appropriate to characterize the pharmacokinetics of paracetamol. Our findings indicate that the rate at which a drug is absorbed from the GI tract reaches adult levels within about 1 week after birth. The maturation function for paracetamol was found applicable to theophylline and indomethacin once solubility limitations were overcome via drug formulation. The influence of excipients on solubility and, hence, oral bioavailability was confirmed for ibuprofen, a second BCS class II compound. CONCLUSIONS: The findings of our study suggest that the processes underlying changes in oral drug absorption after birth are drug-independent and that the maturation function identified for paracetamol may be generally applicable to other BCS class I and II compounds for characterizing drug absorption in preterm as well as term neonates.


Asunto(s)
Recién Nacido/metabolismo , Absorción Intestinal , Acetaminofén/farmacocinética , Administración Oral , Biofarmacia/métodos , Humanos , Ibuprofeno/farmacocinética , Indometacina/farmacocinética , Recien Nacido Prematuro , Teofilina/farmacocinética
4.
Antimicrob Agents Chemother ; 58(11): 6675-84, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25155592

RESUMEN

Recent clinical trials indicate that the use of azithromycin is associated with the emergence of macrolide resistance. The objective of our study was to simultaneously characterize free target site concentrations and correlate them with the MIC90s of clinically relevant pathogens. Azithromycin (500 mg once daily [QD]) was administered orally to 6 healthy male volunteers for 3 days. The free concentrations in the interstitial space fluid (ISF) of muscle and subcutaneous fat tissue as well as the total concentrations in plasma and polymorphonuclear leukocytes (PMLs) were determined on days 1, 3, 5, and 10. All concentrations were modeled simultaneously in NONMEM 7.2 using a tissue distribution model that accounts for nonlinear protein binding and ionization state at physiological pH. The model performance and parameter estimates were evaluated via goodness-of-fit plots and nonparametric bootstrap analysis. The model we developed described the concentrations at all sampling sites reasonably well and showed that the overall pharmacokinetics of azithromycin is driven by the release of the drug from acidic cell/tissue compartments. The model-predicted unionized azithromycin (AZM) concentrations in the cytosol of PMLs (6.0 ± 1.2 ng/ml) were comparable to the measured ISF concentrations in the muscle (8.7 ± 2.9 ng/ml) and subcutis (4.1 ± 2.4 ng/ml) on day 10, whereas the total PML concentrations were >1,000-fold higher (14,217 ± 2,810 ng/ml). The total plasma and free ISF concentrations were insufficient to exceed the MIC90s of the skin pathogens at all times. Our results indicate that the slow release of azithromycin from low pH tissue/cell compartments is responsible for the long terminal half-life of the drug and thus the extended period of time during which free concentrations reside at subinhibitory concentrations.


Asunto(s)
Antibacterianos/farmacocinética , Azitromicina/farmacocinética , Adulto , Antibacterianos/sangre , Antibacterianos/uso terapéutico , Azitromicina/sangre , Azitromicina/uso terapéutico , Farmacorresistencia Bacteriana , Semivida , Voluntarios Sanos , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/microbiología
5.
Br J Clin Pharmacol ; 76(6): 988-96, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23528073

RESUMEN

AIM(S): The current investigation aims to provide new insights into fetal exposure to tacrolimus in utero by evaluating maternal and umbilical cord blood (venous and arterial), plasma and unbound concentrations at delivery. This study also presents a case report of tacrolimus excretion via breast milk. METHODS: Maternal and umbilical cord (venous and arterial) samples were obtained at delivery from eight solid organ allograft recipients to measure tacrolimus and metabolite bound and unbound concentrations in blood and plasma. Tacrolimus pharmacokinetics in breast milk were assessed in one subject. RESULTS: Mean (±SD) tacrolimus concentrations at the time of delivery in umbilical cord venous blood (6.6 ± 1.8 ng ml(-1)) were 71 ± 18% (range 45-99%) of maternal concentrations (9.0 ± 3.4 ng ml(-1)). The mean umbilical cord venous plasma (0.09 ± 0.04 ng ml(-1)) and unbound drug concentrations (0.003 ± 0.001 ng ml(-1)) were approximately one fifth of the respective maternal concentrations. Arterial umbilical cord blood concentrations of tacrolimus were 100 ± 12% of umbilical venous concentrations. In addition, infant exposure to tacrolimus through the breast milk was less than 0.3% of the mother's weight-adjusted dose. CONCLUSIONS: Differences between maternal and umbilical cord tacrolimus concentrations may be explained in part by placental P-gp function, greater red blood cell partitioning and higher haematocrit levels in venous cord blood. The neonatal drug exposure to tacrolimus via breast milk is very low and likely does not represent a health risk to the breastfeeding infant.


Asunto(s)
Parto Obstétrico , Inmunosupresores/farmacocinética , Leche Humana/química , Trasplante de Órganos , Placenta/metabolismo , Tacrolimus/farmacocinética , Adulto , Lactancia Materna , Femenino , Sangre Fetal/química , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/sangre , Inmunosupresores/uso terapéutico , Recién Nacido , Circulación Placentaria , Embarazo , Tacrolimus/administración & dosificación , Tacrolimus/sangre , Tacrolimus/uso terapéutico
6.
Clin Pharmacol Ther ; 113(3): 483-485, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36710622

RESUMEN

Prioritization of diversity, equity, and inclusion in all facets of our work is long overdue for the clinical pharmacology community. Increasing diversity in clinical research will deepen our understanding of nuanced patient populations and help improve all patient outcomes. Fostering an inclusive and diverse workforce will lead to broader perspectives that can better inform critical decisions and create work environments where everyone can thrive. In this call to action, we invite you to join us.


Asunto(s)
Farmacología Clínica , Humanos , Diversidad Cultural , Recursos Humanos
7.
CPT Pharmacometrics Syst Pharmacol ; 12(5): 639-655, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36752286

RESUMEN

The main objective of this tutorial is to provide the readers with a roadmap of how to establish increasingly complex target-mediated drug disposition (TMDD) models for monoclonal antibodies. To this end, we built mathematical models, each with a detailed visualization, starting from the basic TMDD model by Mager and Jusko to the well-established, physiologically based model by Li et al. in a step-wise fashion to highlight the relative importance of key physiological processes that impact mAb kinetics and system dynamics. As the models become more complex, the question of structural and parameter identifiability arises. To address this question, we work through a trastuzumab case example to guide the modeler's choice for model and parameter optimization in light of the context of use. We leave the readers of this tutorial with a brief summary of the advantages and limitations of each model expansion, as well as the model source codes for further self-guided exploration and hands-on analysis.


Asunto(s)
Anticuerpos Monoclonales , Farmacología Clínica , Humanos , Anticuerpos Monoclonales/farmacología , Simulación por Computador , Distribución Tisular , Modelos Biológicos
8.
Clin Transl Sci ; 16(8): 1431-1444, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37154518

RESUMEN

Growth Differentiation Factor-15 (GDF15) is a circulating polypeptide linked to cellular stress and metabolic adaptation. GDF15's half-life is ~3 h and activates the glial cell line-derived neurotrophic factor family receptor alpha-like (GFRAL) receptor expressed in the area postrema. To characterize sustained GFRAL agonism on food intake (FI) and body weight (BW), we tested a half-life extended analog of GDF15 (Compound H [CpdH]) suitable for reduced dosing frequency in obese cynomolgus monkeys. Animals were chronically treated once weekly (q.w.) with CpdH or long-acting GLP-1 analog dulaglutide. Mechanism-based longitudinal exposure-response modeling characterized effects of CpdH and dulaglutide on FI and BW. The novel model accounts for both acute, exposure-dependent effects reducing FI and compensatory changes in energy expenditure (EE) and FI occurring over time with weight loss. CpdH had linear, dose-proportional pharmacokinetics (terminal half-life ~8 days) and treatment caused exposure-dependent reductions in FI and BW. The 1.6 mg/kg CpdH reduced mean FI by 57.5% at 1 week and sustained FI reductions of 31.5% from weeks 9-12, resulting in peak reduction in BW of 16 ± 5%. Dulaglutide had more modest effects on FI and peak BW loss was 3.8 ± 4.0%. Longitudinal modeling of both the FI and BW profiles suggested reductions in BW observed with both CpdH and dulaglutide were fully explained by exposure-dependent reductions in FI without increase in EE. Upon verification of the pharmacokinetic/pharmacodynamic relationship established in monkeys and humans for dulaglutide, we predicted that CpdH could reach double digit BW loss in humans. In summary, a long-acting GDF15 analog led to sustained reductions in FI in overweight monkeys and holds potential for effective clinical obesity pharmacotherapy.


Asunto(s)
Ingestión de Alimentos , Obesidad , Humanos , Animales , Obesidad/metabolismo , Pérdida de Peso , Peso Corporal/fisiología , Primates , Factor 15 de Diferenciación de Crecimiento/farmacología , Factor 15 de Diferenciación de Crecimiento/uso terapéutico
9.
Cell Host Microbe ; 31(5): 751-765.e11, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37098341

RESUMEN

Treating and preventing infections by antimicrobial-resistant bacterial pathogens is a worldwide problem. Pathogens such as Staphylococcus aureus produce an array of virulence determinants, making it difficult to identify single targets for the development of vaccines or monoclonal therapies. We described a human-derived anti-S. aureus monoclonal antibody (mAb)-centyrin fusion protein ("mAbtyrin") that simultaneously targets multiple bacterial adhesins, resists proteolysis by bacterial protease GluV8, avoids Fc engagement by S. aureus IgG-binding proteins SpA and Sbi, and neutralizes pore-forming leukocidins via fusion with anti-toxin centyrins, while maintaining Fc- and complement-mediated functions. Compared with the parental mAb, mAbtyrin protected human phagocytes and boosted phagocyte-mediated killing. The mAbtyrin also reduced pathology, reduced bacterial burden, and protected from different types of infections in preclinical animal models. Finally, mAbtyrin synergized with vancomycin, enhancing pathogen clearance in an animal model of bacteremia. Altogether, these data establish the potential of multivalent mAbs for treating and preventing S. aureus diseases.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Animales , Humanos , Staphylococcus aureus , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/microbiología , Anticuerpos Monoclonales/uso terapéutico , Fagocitos/metabolismo , Leucocidinas/metabolismo , Leucocidinas/uso terapéutico
10.
Ther Drug Monit ; 34(6): 660-70, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23007747

RESUMEN

BACKGROUND: Information on the pharmacokinetics of tacrolimus during pregnancy is limited to case reports despite the increasing number of pregnant women being prescribed tacrolimus for immunosuppression. METHODS: Blood, plasma, and urine samples were collected over 1 steady-state dosing interval from women treated with oral tacrolimus during early to late pregnancy (n = 10) and postpartum (n = 5). Total and unbound tacrolimus as well as metabolite concentrations in blood and plasma were assayed by a validated liquid chromatography/mass spectrometry/mass spectrometry (LC/MS/MS) method. A mixed-effect linear model was used for comparison across gestational age and using postpartum as the reference group. RESULTS: The mean oral clearance (CL/F) based on whole-blood tacrolimus concentration was 39% higher during mid-pregnancy and late pregnancy compared with postpartum (47.4 ± 12.6 vs. 34.2 ± 14.8 L/h, P < 0.03). Tacrolimus-free fraction increased by 91% in plasma (f(P)) and by 100% in blood (f(B)) during pregnancy (P = 0.0007 and 0.002, respectively). Increased fP was inversely associated with serum albumin concentration (r = -0.7, P = 0.003), which decreased by 27% during pregnancy. Pregnancy-related changes in f(P) and f(B) contributed significantly to the observed gestational increase in tacrolimus whole-blood CL/F (r² = 0.36 and 0.47, respectively, P < 0.01). In addition, tacrolimus whole-blood CL/F was inversely correlated with both hematocrit and red blood cell counts, suggesting that binding of tacrolimus to erythrocytes restricts its availability for metabolism. Treating physicians increased tacrolimus dosages in study participants during pregnancy by an average of 45% to maintain tacrolimus whole-blood trough concentrations in the therapeutic range. This led to striking increases in unbound tacrolimus trough concentrations and unbound area under the concentration-time curve, by 112% and 173%, respectively, during pregnancy (P = 0.02 and 0.03, respectively). CONCLUSIONS: Tacrolimus pharmacokinetics are altered during pregnancy. Dose adjustment to maintain whole-blood tacrolimus concentration in the usual therapeutic range during pregnancy increases circulating free drug concentrations, which may impact clinical outcomes.


Asunto(s)
Inmunosupresores/farmacocinética , Tacrolimus/farmacocinética , Adulto , Biotransformación , Isótopos de Carbono , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/sangre , Inmunosupresores/orina , Tasa de Depuración Metabólica , Periodo Posparto , Embarazo , Primer Trimestre del Embarazo , Tercer Trimestre del Embarazo , Albúmina Sérica/análisis , Albúmina Sérica Humana , Tacrolimus/administración & dosificación , Tacrolimus/sangre , Tacrolimus/orina , Espectrometría de Masas en Tándem , Adulto Joven
11.
Clin Transl Sci ; 15(10): 2355-2365, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35981318

RESUMEN

Enhancing resiliency should elevate innovation and efficiency in biomedical research and development (R&D); however, compared with other professions, data on practice of resilience is lacking. Using the Benatti Resiliency Model (5 anchors: Well-Being, Self-Awareness, Brand, Connection, and Innovation), we surveyed professionals, including those in biomedical and pharmaceutical R&D. A structured LinkedIn questionnaire (March 16-May 23, 2021), surveyed each model anchor using five categories. One hundred fifty-eight participants (~6% student/trainee, 18%, 27%, and 49% in 1-5, 5-15 or >15 years post-terminal degree) took the survey (90 in biomedical and pharmaceutical R&D). Over 50% chose "always"/"often" across questions, except external influence or engagement. The question with one of the lowest "always" scores (~15%) was "I get feedback on my influence and impact in my career" in Brand, highlighting areas for leadership development and coaching. In the anchor of Well-being, nutrition and stress management also received some lowest "always" scores (~15% for both). Connection and Innovation scores trended slightly higher in biomedical and pharmaceutical R&D. No students/trainees chose "always" in Brand, indicating evolution of brand maturity over time. Self- and survey-assessed resiliency scores were associated (rs  = 0.37, p < 0.0001). Our survey yielded actionable insights on Resilience, including "best practices" through an open-ended question for one thing most useful to boost resilience in the survey and is the first application of the Benatti Model for crowdsourced research.


Asunto(s)
Liderazgo , Humanos , Encuestas y Cuestionarios , Preparaciones Farmacéuticas
12.
Clin Transl Sci ; 15(9): 2096-2104, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35611545

RESUMEN

Biologics are increasingly being co-developed in combination or as novel constructs like bispecific antibodies (BsAbs) with the goal of targeting multiple, non-redundant mechanisms of action. Rational design of combinations and dual-targeting approaches that consider disease complexities have the potential to improve efficacy and safety, to increase duration of clinical benefit, and to minimize clinical resistance mechanisms. Here we summarize examples of BsAbs and biologic combinations that have been approved by health authorities and present drug development considerations when deciding between these two strategies. These include an understanding of target biology, nonclinical safety risks, dose optimization strategies, the regulatory framework, pharmacokinetic, immunogenicity, and bioanalytical assay considerations. The disease biology, target dynamics, and pharmacology objectives were identified as important factors in early drug development to decide between a BsAb versus a combination. Nonclinical safety assessment and dose optimization strategies can also pose challenges for BsAb versus combinations. High unmet medical needs and lack of treatment options are often the common denominators for deciding to develop a BsAb or a combination. Future development of biologic triple combinations and BsAbs combinations with other biologics will further increase drug development complexities and hold promise for more effective treatment options for patients.


Asunto(s)
Anticuerpos Biespecíficos , Productos Biológicos , Neoplasias , Productos Biológicos/efectos adversos , Combinación de Medicamentos , Desarrollo de Medicamentos , Humanos , Neoplasias/tratamiento farmacológico
13.
Eur J Med Chem ; 236: 114330, 2022 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-35436670

RESUMEN

Pramlintide is an equipotent amylin analogue that reduces food intake and body weight in obese subjects and has been clinically approved as an adjunctive therapy for the treatment of adult diabetic patients. However, due to its extremely short half-life in vivo, a regimen of multiple daily administrations is required for achieving clinical effectiveness. Herein is described the development of prototypical long-acting pramlintide bioconjugates, in which pramlintide's disulfide-linked macrocycle was replaced by a cyclic thioether motif. This modification enabled stable chemical conjugation to a half-life extending antibody. In contrast to pramlintide (t1/2 < 0.75 h), bioconjugates 35 and 38 have terminal half-lives of ∼2 days in mice and attain significant exposure levels that are maintained up to 7 days. Single dose subcutaneous administration of 35 in lean mice, given 18-20 h prior to oral acetaminophen (AAP) administration, significantly reduced gastric emptying (as determined by plasma AAP levels). In a separate study, similar administration of 35 in fasted lean mice effected a reduction in food intake for up to 48 h. These data are consistent with durable amylinomimetic responses and provide the basis for further development of such long-acting amylinomimetic conjugates for the potential treatment of obesity and associated pathologies.


Asunto(s)
Agonistas de los Receptores de Amilina , Agonistas de los Receptores de Amilina/farmacología , Agonistas de los Receptores de Amilina/uso terapéutico , Amiloide , Animales , Peso Corporal , Humanos , Hipoglucemiantes/uso terapéutico , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Ratones , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico
14.
Anal Biochem ; 418(1): 126-33, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21784054

RESUMEN

Simultaneous and accurate measurement of circulating vitamin D metabolites is critical to studies of the metabolic regulation of vitamin D and its impact on health and disease. To that end, we have developed a specific liquid chromatography-tandem mass spectrometry (LC-MS/MS) method that permits the quantification of major circulating vitamin D(3) metabolites in human plasma. Plasma samples were subjected to a protein precipitation, liquid-liquid extraction, and Diels-Alder derivatization procedure prior to LC-MS/MS analysis. Importantly, in all human plasma samples tested, we identified a significant dihydroxyvitamin D(3) peak that could potentially interfere with the determination of 1α,25-dihydroxyvitamin D(3) [1α,25(OH)(2)D(3)] concentrations. This interfering metabolite has been identified as 4ß,25-dihydroxyvitamin D(3) [4ß,25(OH)(2)D(3)] and was found at concentrations comparable to 1α,25(OH)(2)D(3). Quantification of 1α,25(OH)(2)D(3) in plasma required complete chromatographic separation of 1α,25(OH)(2)D(3) from 4ß,25(OH)(2)D(3). An assay incorporating this feature was used to simultaneously determine the plasma concentrations of 25OHD(3), 24R,25(OH)(2)D(3), 1α,25(OH)(2)D(3), and 4ß,25(OH)(2)D(3) in healthy individuals. The LC-MS/MS method developed and described here could result in considerable improvement in quantifying 1α,25(OH)(2)D(3) as well as monitoring the newly identified circulating metabolite, 4ß,25(OH)(2)D(3).


Asunto(s)
24,25-Dihidroxivitamina D 3/análogos & derivados , Colecalciferol/sangre , 24,25-Dihidroxivitamina D 3/sangre , Cromatografía Liquida , Humanos , Espectrometría de Masas en Tándem
15.
Clin Pharmacol Ther ; 109(3): 716-727, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33002189

RESUMEN

Chimeric antigen receptor (CAR)-T cell therapy has achieved considerable success in treating B-cell hematologic malignancies. However, the challenges of extending CAR-T therapy to other tumor types, particularly solid tumors, remain appreciable. There are substantial variabilities in CAR-T cellular kinetics across CAR-designs, CAR-T products, dosing regimens, patient responses, disease types, tumor burdens, and lymphodepletion conditions. As a "living drug," CAR-T cellular kinetics typically exhibit four distinct phases: distribution, expansion, contraction, and persistence. The cellular kinetics of CAR-T may correlate with patient responses, but which factors determine CAR-T cellular kinetics remain poorly defined. Herein, we developed a cellular kinetic model to retrospectively characterize CAR-T kinetics in 217 patients from 7 trials and compared CAR-T kinetics across response status, patient populations, and tumor types. Based on our analysis results, CAR-T cells exhibited a significantly higher cell proliferation rate and capacity but a lower contraction rate in patients who responded to treatment. CAR-T cells proliferate to a higher degree in hematologic malignancies than in solid tumors. Within the assessed dose ranges (107 -109 cells), CAR-T doses were weakly correlated with CAR-T cellular kinetics and patient response status. In conclusion, the developed CAR-T cellular kinetic model adequately characterized the multiphasic CAR-T cellular kinetics and supported systematic evaluations of the potential influencing factors, which can have significant implications for the development of more effective CAR-T therapies.


Asunto(s)
Proliferación Celular , Inmunoterapia Adoptiva , Activación de Linfocitos , Modelos Inmunológicos , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Muerte Celular , Ensayos Clínicos como Asunto , Simulación por Computador , Humanos , Memoria Inmunológica , Cinética , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Estudios Retrospectivos , Linfocitos T/metabolismo
16.
MAbs ; 12(1): 1813962, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32967523

RESUMEN

Biotherapeutic drugs against tumor necrosis factor (TNF) are effective treatments for moderate to severe inflammatory bowel disease (IBD). Here, we evaluated CNTO 5048, an antimurine TNF surrogate monoclonal antibody (mAb), in a CD45RBhigh adoptive T cell transfer mouse colitis model, which allows examination of the early immunological events associated with gut inflammation and the therapeutic effects. The study was designed to quantitatively understand the effects of IBD on CNTO 5048 disposition, the ability of CNTO 5048 to neutralize pathogenic TNF at the colon under disease conditions, and the impact of dosing regimen on CNTO 5048 treatment effect. CNTO 5048 and TNF concentrations in both mice serum and colon homogenate were also measured. Free TNF concentrations in colon, but not in serum, were shown to correlate well with the colon pharmacodynamic readout, such as the summed histopathology score and neutrophil score. A minimal physiologically based pharmacokinetic (mPBPK) model was developed to characterize CNTO 5048 PK and disposition, as well as colon soluble TNF target engagement (TE). The mPBPK/TE model reasonably captured the observed data and provided a quantitative understanding of an anti-TNF mAb on its colon TNF suppression and therapeutic effect in a physiologically relevant IBD animal model. These results also provided insights into the potential benefits of using induction doses for the treatment of IBD patients.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino , Enfermedades Inflamatorias del Intestino , Modelos Biológicos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales de Origen Murino/farmacocinética , Anticuerpos Monoclonales de Origen Murino/farmacología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Ratas , Factor de Necrosis Tumoral alfa/inmunología
17.
Clin Pharmacol Ther ; 107(1): 85-88, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31750932

RESUMEN

Quantitative translational medicine (QTM) is envisioned as a multifaceted discipline that will galvanize the path from idea to medicine through quantitative translation across the discovery, development, regulatory, and utilization spectrum. Here, we summarize results of an American Society for Clinical Pharmacology and Therapeutics (ASCPT) survey on barriers relevant to the advancement of QTM and propose opportunities for its deployment. Importantly, we offer a call to action to break down these barriers through patient-centered stewardship, effective communication, cross-sector collaboration, and a modernized educational curriculum.


Asunto(s)
Farmacología Clínica , Investigación Biomédica Traslacional , Curriculum , Humanos , Farmacología Clínica/educación , Farmacología Clínica/estadística & datos numéricos , Sociedades Farmacéuticas , Encuestas y Cuestionarios , Investigación Biomédica Traslacional/estadística & datos numéricos
18.
MAbs ; 12(1): 1770018, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32544369

RESUMEN

Tumor necrosis factor (TNF) and interleukin (IL)-17A are pleiotropic cytokines implicated in the pathogenesis of several autoimmune diseases including rheumatoid arthritis (RA) and psoriatic arthritis (PsA). JNJ-61178104 is a novel human anti-TNF and anti-IL-17A monovalent, bispecific antibody that binds to both human TNF and human IL-17A with high affinities and blocks the binding of TNF and IL-17A to their receptors in vitro. JNJ-61178104 also potently neutralizes TNF and IL-17A-mediated downstream effects in multiple cell-based assays. In vivo, treatment with JNJ-61178104 resulted in dose-dependent inhibition of cellular influx in a human IL-17A/TNF-induced murine lung neutrophilia model and the inhibitory effects of JNJ-61178104 were more potent than the treatment with bivalent parental anti-TNF or anti-IL-17A antibodies. JNJ-61178104 was shown to engage its targets, TNF and IL-17A, in systemic circulation measured as drug/target complex formation in normal cynomolgus monkeys (cyno). Surprisingly, quantitative target engagement assessment suggested lower apparent in vivo target-binding affinities for JNJ-61178104 compared to its bivalent parental antibodies, despite their similar in vitro target-binding affinities. The target engagement profiles of JNJ-61178104 in humans were in general agreement with the predicted profiles based on cyno data, suggesting similar differences in the apparent in vivo target-binding affinities. These findings show that in vivo target engagement of monovalent bispecific antibody does not necessarily recapitulate that of the molar-equivalent dose of its bivalent parental antibody. Our results also offer valuable insights into the understanding of the pharmacokinetics/pharmacodynamics and target engagement of other bispecific biologics against dimeric and/or trimeric soluble targets in vivo.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Interleucina-17/inmunología , Trastornos Leucocíticos/inmunología , Pulmón/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Interleucina-17/antagonistas & inhibidores , Interleucina-17/metabolismo , Trastornos Leucocíticos/metabolismo , Trastornos Leucocíticos/prevención & control , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macaca fascicularis , Ratones , Inhibidores del Factor de Necrosis Tumoral/inmunología , Inhibidores del Factor de Necrosis Tumoral/farmacocinética , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
19.
Cell Metab ; 29(4): 837-843.e5, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30773465

RESUMEN

The gut hormone PYY3-36 reduces food intake in humans and exhibits at least additive efficacy in combination with GLP-1. However, the utility of PYY analogs as anti-obesity agents has been severely limited by emesis and rapid proteolysis, a profile similarly observed with native PYY3-36 in obese rhesus macaques. Here, we found that antibody conjugation of a cyclized PYY3-36 analog achieved high NPY2R selectivity, unprecedented in vivo stability, and gradual infusion-like exposure. These properties permitted profound reduction of food intake when administered to macaques for 23 days without a single emetic event in any animal. Co-administration with the GLP-1 receptor agonist liraglutide for an additional 5 days further reduced food intake with only one animal experiencing a single bout of emesis. This antibody-conjugated PYY analog therefore may enable the long-sought potential of GLP-1/PYY-based combination treatment to achieve robust, well-tolerated weight reduction in obese patients.


Asunto(s)
Anorexia/inducido químicamente , Péptido YY/química , Péptido YY/farmacología , Vómitos , Animales , Células CHO , Cricetulus , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Células HEK293 , Humanos , Liraglutida/farmacología , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Péptido YY/administración & dosificación , Vómitos/inducido químicamente
20.
MAbs ; 9(8): 1306-1316, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28898162

RESUMEN

The increased number of bispecific antibodies (BsAb) under therapeutic development has resulted in a need for mouse surrogate BsAbs. Here, we describe a one-step method for generating highly pure mouse BsAbs suitable for in vitro and in vivo studies. We identify two mutations in the mouse IgG2a and IgG2b Fc region: one that eliminates protein A binding and one that enhances protein A binding by 8-fold. We show that BsAbs harboring these mutations can be purified from the residual parental monoclonal antibodies in one step using protein A affinity chromatography. The structural basis for the effects of these mutations was analyzed by X-ray crystallography. While the mutation that disrupted protein A binding also inhibited FcRn interaction, a bispecific mutant in which one subunit retained the ability to bind protein A could still interact with FcRn. Pharmacokinetic analysis of the serum half-lives of the mutants showed that the mutant BsAb had a serum half-life comparable to a wild-type Ab. The results describe a rapid method for generating panels of mouse BsAbs that could be used in mouse studies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Receptores Fc/inmunología , Proteína Estafilocócica A/inmunología , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Cristalografía por Rayos X , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/inmunología , Proteínas Mutantes/metabolismo , Mutación , Unión Proteica/inmunología , Dominios Proteicos , Receptores Fc/metabolismo , Proteína Estafilocócica A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA