Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Biopharm Drug Dispos ; 37(2): 75-92, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26461173

RESUMO

The mechanisms of absorption, distribution, metabolism and elimination of small and large molecule therapeutics differ significantly from one another and can be explored within the framework of a physiologically based pharmacokinetic (PBPK) model. This paper briefly reviews fundamental approaches to PBPK modeling, in which drug kinetics within tissues and organs are explicitly represented using physiologically meaningful parameters. The differences in PBPK models applied to small/large molecule drugs are highlighted, thus elucidating differences in absorption, distribution and elimination properties between these two classes of drugs in a systematic manner. The absorption of small and large molecules differs with respect to their common extravascular routes of delivery (oral versus subcutaneous). The role of the lymphatic system in drug distribution, and the involvement of tissues as sites of elimination (through catabolism and target mediated drug disposition) are unique features of antibody distribution and elimination that differ from small molecules, which are commonly distributed into the tissues but are eliminated primarily by liver metabolism. Fundamental differences exist in the ability to predict human pharmacokinetics based upon preclinical data due to differing mechanisms governing small and large molecule disposition. These differences have influence on the evolving utilization of PBPK modeling in the discovery and development of small and large molecule therapeutics.


Assuntos
Anticorpos Monoclonais/farmacocinética , Modelos Biológicos , Animais , Humanos
2.
Regul Toxicol Pharmacol ; 69(2): 226-33, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24755365

RESUMO

New challenges and opportunities in nonclinical safety testing of biologics were discussed at the 3rd European BioSafe Annual General Membership meeting in November 2013 in Berlin: (i)Approaches to refine use of non-human primates in non-clinical safety testing of biologics and current experience on the use of minipigs as alternative non-rodent species.(ii)Tissue distribution studies as a useful tool to support pharmacokinetic/pharmacodynamic (PKPD) assessment of biologics, in that they provide valuable mechanistic insights at drug levels at the site of action.(iii)Mechanisms of nonspecific toxicity of antibody drug conjugates (ADC) and ways to increase the safety margins.(iv)Although biologics toxicity typically manifests as exaggerated pharmacology there are some reported case studies on unexpected toxicity.(v)Specifics of non-clinical development approaches of noncanonical monoclonal antibodies (mAbs), like bispecifics and nanobodies.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Produtos Biológicos/efeitos adversos , Avaliação Pré-Clínica de Medicamentos/métodos , Segurança , Testes de Toxicidade , Animais , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/farmacocinética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Produtos Biológicos/imunologia , Produtos Biológicos/farmacocinética , Humanos , Modelos Animais , Primatas , Anticorpos de Domínio Único/efeitos adversos , Suínos , Porco Miniatura , Distribuição Tecidual
3.
Mol Pharm ; 10(5): 1581-95, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-23256608

RESUMO

Drug delivery across the brain-blood interfaces is a complex process involving physicochemical drug properties, transporters, enzymes, and barrier dysfunction in diseased conditions. Intact blood-brain barrier (BBB) limits the entry of potentially harmful compounds into the brain but may also reduce the CNS permeability of therapeutic agents. BBB permeability is typically assessed by measuring brain-to-plasma ratio in rodents (referred to as B/P ratio, BB, or Kp, often calculated as logBB), an approach that suffers significant limitations as discussed in the present review. Kp is not a permeability measurement but a partition coefficient mainly driven by the relative binding to plasma and brain tissue components including lipids, phospholipids, and proteins. Compounds with high Kp are often lipophilic with low free fraction available to mediate CNS activities. Efforts should be more concentrated on measuring pharmacologically relevant free drug concentrations at the target site. Using healthy rodents to predict brain penetration in patients might be biased due to species differences in BBB-related parameters such as transporter expression and functional activities. In addition, pathophysiological conditions such as aging, multiple sclerosis, and Alzheimer's and Parkinson's diseases have been described to affect BBB permeability, with barrier leakage and altered transporter activity. The impact of these species differences and disease states on drug delivery to the brain is largely overlooked. More data are needed to better understand their clinical implication in order to design more appropriate screening strategies and ultimately better mitigate the risk for failure in late stage development.


Assuntos
Barreira Hematoencefálica/metabolismo , Animais , Permeabilidade Capilar , Fármacos do Sistema Nervoso Central/administração & dosagem , Fármacos do Sistema Nervoso Central/sangue , Fármacos do Sistema Nervoso Central/farmacocinética , Doenças do Sistema Nervoso Central/tratamento farmacológico , Doenças do Sistema Nervoso Central/metabolismo , Sistemas de Liberação de Medicamentos , Humanos , Especificidade da Espécie
4.
Pharm Res ; 29(9): 2512-21, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22707361

RESUMO

PURPOSE: To compare the pharmacokinetics (PK) of MNRP1685A, a human monoclonal antibody (mAb) against neuropilin-1 (NRP1), in mice, rats, monkeys, and cancer patients from a Phase I study to model with parallel linear and nonlinear clearances. METHODS: Binding characteristics of MNRP1685A in different species were evaluated using surface plasmon resonance technology. PK profiles of MNRP1685A after single and/or multiple doses in different species were analyzed using population analysis. PK parameters were compared across species. RESULTS: MNRP1685A binds to NRP1 in all four species tested. Consistent with the wide expression of NRP1, MNRP1685A demonstrated pronounced non-linear PK over a wide dose range. PK profiles are best described by a two-compartment model with parallel linear and nonlinear clearances. Model-derived PK parameters suggest similar in-vivo target expression levels and binding affinity to target across all species tested. However, compared to typical human/humanized mAbs, non-specific clearance of MNRP1685A was faster in mice, rats, and humans (60.3, 19.4, and 8.5 ml/day/kg), but not in monkeys (3.22 ml/day/kg). CONCLUSIONS: Monkey PK properly predicted the target-mediated clearance of MNRP1685A but underestimated its non-specific clearance in humans. This unique PK property warrants further investigation of underlying mechanisms.


Assuntos
Anticorpos Monoclonais/farmacocinética , Neuropilina-1/imunologia , Animais , Anticorpos Monoclonais/imunologia , Humanos , Modelos Biológicos , Especificidade da Espécie
5.
J Pharmacokinet Pharmacodyn ; 39(2): 217-26, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22382554

RESUMO

MNRP1685A (anti-NRP1) is a fully human IgG1 monoclonal antibody against neuropilin-1 (NRP1), a protein necessary for blood vessel maturation. MNRP1685A binds to free membrane-bound NRP1 (mNRP1) and circulating NRP1 (cNRP1). Total cNRP1 increased in a dose-dependent manner following anti-NRP1 administration in mice, rats, and monkeys. The purpose of this study is to develop a mechanism-based model to simultaneously describe the kinetics of both unbound drug (MNRP1685A) and total cNRP1 in cynomolgus monkeys. Pharmacokinetic (PK) and pharmacodynamic (PD) profiles after single- or multiple-dose administrations were well described by the two-target quasi-steady-state (QSS) model. The estimated nonspecific clearance was 3.26 mL/day/kg and central compartment volume was 38.2 mL/kg. The maximum elimination rate for mNRP1-mediated disposition was 98.8 nM/day. The synthesis rate (3.8 nM/day), degradation rate constant (1.53 day(-1)), and complex elimination rate constant (0.260 day(-1)) for cNRP1 were also derived from the model. QSS constants were 6.94 nM for mNRP1 and 2.8 nM for cNRP1. The results suggest that cNRP1 has minimal effect on MNRP1685A PK while mNRP1 plays a major role in the target-mediated drug disposition. This finding is favorable as the desired pharmacological target is mNRP1, rather than cNRP1. The two-target QSS model provides mechanistic understanding of the nonlinear PK of MNRP1685A. Based on the model prediction, the free drug concentrations to maintain free mNRP1 and cNRP1 below 10% of baseline level are 10 and 20 µg/mL, respectively. This serves as a target concentration for clinical dose selection, assuming cynomolgus monkeys are predictive for humans.


Assuntos
Anticorpos Monoclonais/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Modelos Biológicos , Neuropilina-1/antagonistas & inibidores , Animais , Anticorpos Monoclonais/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/farmacologia , Macaca fascicularis , Masculino , Neuropilina-1/metabolismo , Farmacocinética , Ovinos
6.
Bioconjug Chem ; 22(10): 1994-2004, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-21913715

RESUMO

Antibody-drug conjugates (ADCs) are designed to combine the exquisite specificity of antibodies to target tumor antigens with the cytotoxic potency of chemotherapeutic drugs. In addition to the general chemical stability of the linker, a thorough understanding of the relationship between ADC composition and biological disposition is necessary to ensure that the therapeutic window is not compromised by altered pharmacokinetics (PK), tissue distribution, and/or potential organ toxicity. The six-transmembrane epithelial antigen of prostate 1 (STEAP1) is being pursued as a tumor antigen target. To assess the role of ADC composition in PK, we evaluated plasma and tissue PK profiles in rats, following a single dose, of a humanized anti-STEAP1 IgG1 antibody, a thio-anti-STEAP1 (ThioMab) variant, and two corresponding thioether-linked monomethylauristatin E (MMAE) drug conjugates modified through interchain disulfide cysteine residues (ADC) and engineered cysteines (TDC), respectively. Plasma PK of total antibody measured by enzyme-linked immunosorbent assay (ELISA) revealed ∼45% faster clearance for the ADC relative to the parent antibody, but no apparent difference in clearance between the TDC and unconjugated parent ThioMab. Total antibody clearances of the two unconjugated antibodies were similar, suggesting minimal effects on PK from cysteine mutation. An ELISA specific for MMAE-conjugated antibody indicated that the ADC cleared more rapidly than the TDC, but total antibody ELISA showed comparable clearance for the two drug conjugates. Furthermore, consistent with relative drug load, the ADC had a greater magnitude of drug deconjugation than the TDC in terms of free plasma MMAE levels. Antibody conjugation had a noticeable, albeit minor, impact on tissue distribution with a general trend toward increased hepatic uptake and reduced levels in other highly vascularized organs. Liver uptakes of ADC and TDC at 5 days postinjection were 2-fold and 1.3-fold higher, respectively, relative to the unmodified antibodies. Taken together, these results indicate that the degree of overall structural modification in anti-STEAP1-MMAE conjugates has a corresponding level of impact on both PK and tissue distribution.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Antígenos de Neoplasias/imunologia , Imunoconjugados/química , Imunoconjugados/farmacocinética , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Oxirredutases/imunologia , Animais , Anticorpos Monoclonais/sangue , Dissulfetos/química , Humanos , Imunoconjugados/sangue , Masculino , Modelos Moleculares , Oligopeptídeos/sangue , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
7.
Toxicol Appl Pharmacol ; 250(2): 194-212, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21034759

RESUMO

A general toxicity of basic drugs is related to phospholipidosis in tissues. Therefore, it is essential to predict the tissue distribution of basic drugs to facilitate an initial estimate of that toxicity. The objective of the present study was to further assess the original prediction method that consisted of using the binding to red blood cells measured in vitro for the unbound drug (RBCu) as a surrogate for tissue distribution, by correlating it to unbound tissue:plasma partition coefficients (Kpu) of several tissues, and finally to predict volume of distribution at steady-state (V(ss)) in humans under in vivo conditions. This correlation method demonstrated inaccurate predictions of V(ss) for particular basic drugs that did not follow the original correlation principle. Therefore, the novelty of this study is to provide clarity on the actual hypotheses to identify i) the impact of pharmacological mode of action on the generic correlation of RBCu-Kpu, ii) additional mechanisms of tissue distribution for the outlier drugs, iii) molecular features and properties that differentiate compounds as outliers in the original correlation analysis in order to facilitate its applicability domain alongside the properties already used so far, and finally iv) to present a novel and refined correlation method that is superior to what has been previously published for the prediction of human V(ss) of basic drugs. Applying a refined correlation method after identifying outliers would facilitate the prediction of more accurate distribution parameters as key inputs used in physiologically based pharmacokinetic (PBPK) and phospholipidosis models.


Assuntos
Lipidoses/induzido quimicamente , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Fosfolipídeos/metabolismo , Animais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Ratos , Distribuição Tecidual
8.
J Pharmacokinet Pharmacodyn ; 38(5): 581-93, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21779940

RESUMO

The volume of distribution at steady state (Vss) of therapeutic proteins is usually assessed by non-compartmental or compartmental pharmacokinetic (PK) analysis wherein errors may arise due to the elimination of therapeutic proteins from peripheral tissues that are not in rapid equilibrium with the sampling compartment (usually blood). Here we explored another potential source of error in the estimation of Vss that is linked to the heterogeneity of therapeutic proteins which may consist of components (e.g. glycosylation variants) with different elimination rates. PK simulations were performed with such hypothetical binary protein mixtures where elimination was assumed to be exclusively from the central compartment. The simulations demonstrated that binary mixtures containing a rapid-elimination component can give rise to pronounced bi-phasic concentration-time profiles. Apparent Vss observed with both non-compartmental and 2-compartmental PK analysis, increased with increasing fraction as well as with increasing elimination rate k ( 10 ) of the rapid-elimination component. Simulation results were complemented by PK analysis of an in vivo study in cynomolgus monkeys with different lots of lenercept, a tumor necrosis factor receptor-immunoglobulin G1 fusion protein, with different heterogeneities. The comparative Vss data for the three lenercept lots with different amounts of rapidly cleared components were consistent with the outcome of our simulations. Both lots with a higher fraction of rapidly cleared components had a statistically significant higher Vss as compared to the reference lot. Overall our study demonstrates that Vss of a therapeutic protein may be overestimated in proteins with differently eliminated components.


Assuntos
Software , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Simulação por Computador , Composição de Medicamentos , Glicosilação , Humanos , Cadeias gama de Imunoglobulina/química , Cadeias gama de Imunoglobulina/uso terapêutico , Macaca fascicularis , Taxa de Depuração Metabólica , Polissacarídeos/farmacocinética , Estrutura Quaternária de Proteína , Receptores do Fator de Necrose Tumoral/química , Receptores do Fator de Necrose Tumoral/uso terapêutico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Fatores de Tempo
9.
Bioconjug Chem ; 21(12): 2153-63, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21053952

RESUMO

Antibody pharmacokinetics and pharmacodynamics are often governed by biological processes such as binding to antigens and other cognate receptors. Emphasis must also be placed, however, on fundamental physicochemical properties that define antibodies as complex macromolecules, including shape, size, hydrophobicity, and charge. Electrostatic interactions between anionic cell membranes and the predominantly positive surface charge of most antibodies can influence blood concentration and tissue disposition kinetics in a manner that is independent of antigen recognition. In this context, the deliberate modification of antibodies by chemical means has been exploited as a valuable preclinical research tool to investigate the relationship between net molecular charge and biological disposition. Findings from these exploratory investigations may be summarized as follows: (I) shifts in isoelectric point of approximately one pI unit or more can produce measurable changes in tissue distribution and kinetics, (II) increases in net positive charge generally result in increased tissue retention and increased blood clearance, and (III) decreases in net positive charge generally result in decreased tissue retention and increased whole body clearance. Understanding electrostatic interactions between antibodies and biological matrices holds relevance in biotechnology, especially with regard to the development of immunoconjugates. The guiding principles and knowledge gained from preclinical evaluation of chemically modified antibodies will be discussed and placed in the context of therapeutic antibodies that are currently marketed or under development, with a particular emphasis on pharmacokinetic and disposition properties.


Assuntos
Anticorpos/química , Anticorpos/metabolismo , Imunoconjugados/química , Imunoconjugados/farmacocinética , Animais , Ânions/metabolismo , Anticorpos/imunologia , Anticorpos/farmacologia , Antígenos/imunologia , Cátions/metabolismo , Membrana Celular/imunologia , Membrana Celular/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imunoconjugados/imunologia , Imunoconjugados/metabolismo , Ponto Isoelétrico , Camundongos , Modelos Moleculares , Terapia de Alvo Molecular , Neoplasias/imunologia , Neoplasias/terapia , Ligação Proteica , Engenharia de Proteínas/métodos , Ratos , Eletricidade Estática , Distribuição Tecidual/imunologia
10.
Drug Metab Dispos ; 38(7): 1029-38, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20406853

RESUMO

Factors determining the pharmacokinetics of 2-chloro-N-(4-chloro-3-(pyridine-2-yl)phenyl)-4-(methylsulfonyl)benzamide (GDC-0449) were investigated using preclinical studies and physiologically based pharmacokinetic (PBPK) modeling. Multiple-dose studies where dogs were given twice-daily oral doses of either 7.5 or 25 mg/kg GDC-0449 showed less than dose-proportional increases in exposure on day 1. At steady state, exposures were comparable between the two dose groups. Oral administration of activated charcoal to dogs receiving oral or intravenous GDC-0449 (25 mg) showed a more rapid decrease in plasma concentrations, suggesting that the concentration gradient driving intestinal membrane permeation was reversible. The biliary clearance of GDC-0449 in dogs was low (0.04 ml/min/kg) and did not account for the majority of the estimated systemic clearance (approximately 19% of systemic clearance). Likewise, in vitro studies using sandwich-cultured human hepatocytes showed negligible biliary excretion. The effect of particle size on oral absorption was shown in a single-dose study where 150 mg of GDC-0449 of two particle sizes was administered. An oral PBPK model was used to investigate mechanisms determining the oral pharmacokinetics of GDC-0449. The overall oral absorption of GDC-0449 appears to depend on the interplay between the dissolution and intestinal membrane permeation processes. A unique feature of GDC-0449 distinguishing it from other Biopharmaceutical Classification System II compounds was that incorporation of the effects of solubility rate-limited absorption and nonsink permeation on the intestinal membrane permeation process was necessary to describe its pharmacokinetic behavior.


Assuntos
Anilidas/química , Anilidas/farmacocinética , Proteínas Hedgehog/antagonistas & inibidores , Piridinas/química , Piridinas/farmacocinética , Administração Oral , Anilidas/administração & dosagem , Animais , Carvão Vegetal/farmacologia , Simulação por Computador , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Hepatócitos/metabolismo , Humanos , Injeções Intravenosas , Absorção Intestinal , Masculino , Tamanho da Partícula , Piridinas/administração & dosagem , Solubilidade
11.
Drug Metab Dispos ; 38(4): 600-5, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20071453

RESUMO

The neonatal Fc receptor (FcRn) plays a critical role in maintaining homeostasis of IgG antibodies. Recent studies have shown that the FcRn-IgG interaction can be modulated to alter the pharmacokinetics of the antibody. This has been achieved by altering amino acid residues in the FcRn-binding domain of the antibody, resulting in a change in the pH-dependent binding affinity of the antibody to FcRn. The purpose of this study was to examine the impact of the pH-dependent FcRn binding affinity on the pharmacokinetics of the antibody with changes in the Asn434 residue. Two anti-tumor necrosis factor-alpha monoclonal antibody (mAb) FcRn variants (N434A and N434H) were engineered, and pharmacokinetic studies of the two FcRn variants together with the wild type (WT) were conducted in mice and cynomolgus monkeys. N434A, which had binding properties to murine FcRn similar to those of the WT, had the same pharmacokinetic profile as the WT in mice. N434H, with the highest binding affinity to murine FcRn at pH 7.4, had a faster clearance (16.1 ml/day/kg) and a lower bioavailability (61.3%) compared with the WT (5.07 ml/day/kg, 73.2%) and N434A (5.90 ml/day/kg, 72.4%) in mice. N434A and N434H, which had higher binding affinity at pH 6.0 to monkey FcRn with comparable affinity at pH 7.4, had significantly higher areas under the serum concentration-time curve from time 0 to day 7 than the WT (749 +/- 71.9 and 819 +/- 81.5 versus 592 +/- 56.8 microg/ml . day) in monkeys. Thus, increasing the binding affinity of mAbs to FcRn at pH 6.0 while keeping a low binding affinity at pH 7.4 improves the pharmacokinetics of these molecules.


Assuntos
Anticorpos Bloqueadores/metabolismo , Anticorpos Monoclonais/farmacocinética , Receptores Fc/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Bloqueadores/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Disponibilidade Biológica , Humanos , Concentração de Íons de Hidrogênio , Injeções Intravenosas , Injeções Subcutâneas , Macaca fascicularis , Camundongos , Camundongos SCID , Especificidade da Espécie
12.
Mol Pharm ; 7(5): 1848-57, 2010 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-20704296

RESUMO

Identification of clinically predictive models of disposition kinetics for antibody therapeutics is an ongoing pursuit in drug development. To encourage translation of drug candidates from early research to clinical trials, clinical diagnostic agents may be used to characterize antibody disposition in physiologically relevant preclinical models. TechneScan PYP was employed to measure tissue vascular volumes (V(v)) in healthy mice. Two methods of red blood cell (RBC) labeling were compared: a direct in vivo method that is analogous to a clinical blood pool imaging protocol, and an indirect method in which radiolabeled blood was transfused from donor mice into recipient mice. The indirect method gave higher precision in RBC labeling yields, lower V(v) values in most tissues, and lower (99m)Tc uptake in kidneys and bladder by single photon emission computed tomographic (SPECT) imaging relative to the direct method. Furthermore, the relative influence of each method on the calculated area under the first 7 days of the concentration-time curve (AUC(0-7)) of an IgG in nude mice was assessed using a physiologically based pharmacokinetic model. The model was sensitive to the source of V(v) values, whether obtained from the literature or measured by either method, when used to predict experimental AUC(0-7) values for radiolabeled trastuzumab in healthy murine tissues. In summary, a novel indirect method for preclinical determination of V(v) offered higher precision in RBC labeling efficiency and lower renal uptake of (99m)Tc than the direct method. In addition, these observations emphasize the importance of obtaining accurate physiological parameter values for modeling antibody uptake.


Assuntos
Volume Sanguíneo , Compostos Radiofarmacêuticos , Pirofosfato de Tecnécio Tc 99m , Animais , Anticorpos Monoclonais Humanizados/farmacocinética , Vasos Sanguíneos/anatomia & histologia , Química Farmacêutica , Descoberta de Drogas , Eritrócitos/metabolismo , Feminino , Camundongos , Camundongos Nus , Modelos Biológicos , Imagem Multimodal , Farmacocinética , Tomografia por Emissão de Pósitrons , Distribuição Tecidual , Tomografia Computadorizada por Raios X , Trastuzumab
13.
Expert Opin Drug Metab Toxicol ; 4(9): 1143-52, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18721109

RESUMO

BACKGROUND: Whole-body physiologically-based pharmacokinetic (WB-PBPK) models mathematically describe an organism as a closed circulatory system consisting of compartments that represent the organs important for compound absorption, distribution, metabolism and elimination. OBJECTIVES: To review the current state of WB-PBPK model use in the clinical phases of drug development. METHODS: A qualitative description of the WB-PBPK model structure is included along with a review of the varying methods available for input parameterisation. Current and potential WB-PBPK model application in clinical development is discussed. CONCLUSIONS: This modelling tool is at present used for small and large molecule drug development primarily as a means to scale pharmacokinetics from animals to humans based on physiology. The pharmaceutical industry is active in employing these models to clinical drug development although the applications in use now are narrow in comparison to the potential. Expanded integration of WB-PBPK models into the drug development process will only be achieved with staff training, managerial will, success stories and regulatory agency openness.


Assuntos
Desenho de Fármacos , Modelos Biológicos , Farmacocinética , Indústria Farmacêutica/economia , Indústria Farmacêutica/organização & administração , Humanos
14.
J Immunol Res ; 2016: 2342187, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27579329

RESUMO

Biologics have emerged as a powerful and diverse class of molecular and cell-based therapies that are capable of replacing enzymes, editing genomes, targeting tumors, and more. As this complex array of tools arises a distinct set of challenges is rarely encountered in the development of small molecule therapies. Biotherapeutics tend to be big, bulky, polar molecules comprised of protein and/or nucleic acids. Compared to their small molecule counterparts, they are fragile, labile, and heterogeneous. Their biodistribution is often limited by hydrophobic barriers which often restrict their administration to either intravenous or subcutaneous entry routes. Additionally, their potential for immunogenicity has proven to be a challenge to developing safe and reliably efficacious drugs. Our discussion will emphasize immunogenicity in the context of therapeutic proteins, a well-known class of biologics. We set out to describe what is known and unknown about the mechanisms underlying the interplay between antigenicity and immune response and their effect on the safety, efficacy, pharmacokinetics, and pharmacodynamics of these therapeutic agents.


Assuntos
Produtos Biológicos/imunologia , Produtos Biológicos/farmacologia , Proteínas/imunologia , Proteínas/farmacologia , Animais , Produtos Biológicos/efeitos adversos , Produtos Biológicos/farmacocinética , Simulação por Computador , Gerenciamento Clínico , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/farmacologia , Imunomodulação , Modelos Biológicos , Modelos Imunológicos , Engenharia de Proteínas , Proteínas/efeitos adversos , Proteínas/farmacocinética , Fatores de Risco , Resultado do Tratamento
15.
J Med Chem ; 47(6): 1487-513, 2004 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-14998336

RESUMO

Cyclothialidine (1, Ro 09-1437) is a potent DNA gyrase inhibitor that was isolated from Streptomyces filipinensis NR0484 and is a member of a new family of natural products. It acts by competitively inhibiting the ATPase activity exerted by the B subunit of DNA gyrase but barely exhibits any growth inhibitory activity against intact bacterial cells, presumably due to insufficient permeation of the cytoplasmic membrane. To explore the antibacterial potential of 1, we developed a flexible synthetic route allowing for the systematic modification of its structure. From a first set of analogues, structure-activity relationships (SAR) were established for different substitution patterns, and the 14-hydroxylated, bicyclic core (X) of 1 seemed to be the structural prerequisite for DNA gyrase inhibitory activity. The variation of the lactone ring size, however, revealed that activity can be found among 11- to 16-membered lactones, and even seco-analogues were shown to maintain some enzyme inhibitory properties, thereby reducing the minimal structural requirements to a rather simple, hydroxylated benzyl sulfide (XI). On the basis of these "minimal structures" a modification program afforded a number of inhibitors that showed in vitro activity against Gram-positive bacteria. The best activities were displayed by 14-membered lactones, and representatives of this subclass exhibit excellent and broad in vitro antibacterial activity against Gram-positive pathogens, including Staphylococcus aureus, Streptococcus pyogenes, and Enterococcus faecalis, and overcome resistance against clinically used drugs. By improving the pharmacokinetic properties of the most active compounds (94, 97), in particular by lowering their lipophilic properties, we were able to identify congeners of cyclothialidine (1) that showed efficacy in vivo.


Assuntos
Antibacterianos/síntese química , Lactamas/síntese química , Lactonas/síntese química , Oxidiazóis/síntese química , Peptídeos Cíclicos/síntese química , Inibidores da Topoisomerase II , Animais , Antibacterianos/química , Antibacterianos/farmacologia , DNA Girase/química , Farmacorresistência Bacteriana Múltipla , Bactérias Gram-Positivas/efeitos dos fármacos , Células HeLa , Humanos , Lactamas/química , Lactamas/farmacologia , Lactonas/química , Lactonas/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Oxidiazóis/química , Oxidiazóis/farmacologia , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Subunidades Proteicas/antagonistas & inibidores , Infecções Estafilocócicas/tratamento farmacológico , Estereoisomerismo , Relação Estrutura-Atividade , Testes de Toxicidade
16.
Biochem Pharmacol ; 64(11): 1637-50, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12429353

RESUMO

The influence of the isolation procedure of hepatocytes, extracellular matrix (ECM) configuration and incubation medium supplementation by dexamethasone (DEX) on the cell morphology and on the gene expression of membrane transporters was examined in rat hepatocytes. The mRNA levels were determined using oligonucleotide microarrays, in liver, in suspension and in primary culture in monolayer (CPC), and in collagen gels sandwich (SPC) in absence and presence of DEX (100 and 1000 nM). The results indicated pronounced morphological differences between CPC and SPC in response to DEX demonstrating that the hepatocytes re-formed, as in vivo, multicellular arrays with extensive bile canalicular network only in SPC in presence of DEX. The mRNA levels of membrane transporters were not affected significantly during isolation procedure. However, plating hepatocytes in CPC resulted in a decrease of major basolateral transporters mRNA level whereas mRNA levels of mdr1b and mrp3 were increased (>100-fold). Similar observations were made in SPC in the absence of DEX demonstrating that the ECM configuration alone did not play a critical role in the regulation of membrane transporters. However, adding DEX to the incubation medium in SPC resulted in an up-regulation of mdr2, oatp2 and mrp2 in a concentration-dependent way for the two latter genes, whereas mdr1b and mrp3 expression were maintained to their baseline liver levels. These data suggested therefore that the combination of ECM and DEX supplementation is essential for the formation of the bile canalicular network and is a determinant factor in the regulation of membrane transporters in cultured rat hepatocytes.


Assuntos
Dexametasona/farmacologia , Matriz Extracelular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/biossíntese , Animais , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Quimiocinas CC/biossíntese , Hepatócitos/fisiologia , Proteínas Inflamatórias de Macrófagos/biossíntese , Proteínas de Membrana Transportadoras/biossíntese , Ratos
17.
Toxicol Lett ; 138(1-2): 29-49, 2003 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-12559691

RESUMO

The present paper proposes a modeling and simulation strategy for the prediction of pharmacokinetics (PK) of drug candidates by using currently available in silico and in vitro based prediction tools for absorption, distribution, metabolism and excretion (ADME). These methods can be used to estimate specific ADME parameters (such as rate and extent of absorption into portal vein, volume of distribution, metabolic clearance in the liver). They can also be part of a physiologically based pharmacokinetic (PBPK) model to simulate concentration-time profiles in tissues and plasma resulting from the overall PK after intravenous or oral administration. Since the ADME prediction tools are built only on commonly generated in silico and in vitro data, they can be applied already in early drug discovery, prior to any in vivo study. With the suggested methodology, the following advantages of the mechanistic PBPK modeling framework can now be utilized to explore potential clinical candidates already in drug discovery: (i) prediction of plasma (blood) and tissue PK of drug candidates prior to in vivo experiments, (ii) supporting a better mechanistic understanding of PK properties, as well as helping the development of more rationale PK-PD relationships from tissue kinetic data predicted, and hence facilitating a more rational decision during clinical candidate selection, and (iii) the extrapolation across species, routes of administration and dose levels.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Avaliação de Medicamentos/métodos , Drogas em Investigação/farmacocinética , Drogas em Investigação/toxicidade , Modelos Biológicos , Animais , Drogas em Investigação/classificação , Humanos , Relação Quantitativa Estrutura-Atividade , Ratos , Solubilidade , Especificidade da Espécie
18.
J Pharm Sci ; 91(5): 1358-70, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11977112

RESUMO

Many in vitro data on physicochemical properties and specific absorption, distribution, metabolism, and elimination (ADME) processes are already available at early stages of drug discovery. These data about new drug candidates could be integrated/connected in physiologically based pharmacokinetic (PBPK) models to estimate a priori the overall plasma and tissue kinetic behaviors under in vivo conditions. The objective of the present study was to illustrate that generic PBPK models integrating such data can be developed in drug discovery prior to any in vivo studies. This approach was illustrated with three example compounds, including two lipophilic bases (diazepam, propranolol) and one neutral more hydrophilic drug (ethoxybenzamide). Distribution and liver metabolism were the processes integrated in the generic rat PBPK models of disposition. Tissue:plasma partition coefficients (P(t:p)s) used for description of distribution were estimated from established tissue composition-based equations, which need only in vitro data on drug lipophilicity and plasma protein binding as sole input parameters. Furthermore, data on intrinsic clearance (CL(int)) determined in vitro with hepatocytes were scaled to the in vivo situation to estimate hepatic metabolic clearance. These prediction approaches were both incorporated in the PBPK models to enable automated estimation of distribution and liver metabolism for each drug studied. The generic PBPK models suggested can simulate a priori concentration-time profiles of plasma and several tissues after intravenous administrations to rat. The results indicate that most of the simulated concentration-time profiles of plasma and 10 tissues are in reasonable agreement with the corresponding experimental data determined in vivo (less than a factor of two). However, some more relevant deviations were observed for specific tissues (brain and gut for diazepam; liver and gut for ethoxybenzamide; lung for propranolol) because of important ADME processes were probably neglected in the PBPK models of these drugs. In this context, generic PBPK models were also used for mechanistic evaluations of pharmacokinetics for generating research hypotheses to understand these deviations. Overall, the present generic and integrative PBPK approach of drug disposition suggested as a tool for a priori simulations and mechanistic evaluations of pharmacokinetics has the potential to improve the selection and optimization of new drug candidates.


Assuntos
Benzamidas/farmacocinética , Etil-Éteres/farmacocinética , Farmacocinética , Animais , Benzamidas/sangue , Diazepam/sangue , Diazepam/farmacocinética , Etil-Éteres/sangue , Taxa de Depuração Metabólica , Modelos Biológicos , Propranolol/sangue , Propranolol/farmacocinética , Ratos , Fatores de Tempo , Distribuição Tecidual
19.
J Pharm Sci ; 91(1): 129-56, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11782904

RESUMO

In drug discovery and nonclinical development the volume of distribution at steady state (V(ss)) of each novel drug candidate is commonly determined under in vivo conditions. Therefore, it is of interest to predict V(ss) without conducting in vivo studies. The traditional description of V(ss) corresponds to the sum of the products of each tissue:plasma partition coefficient (P(t:p)) and the respective tissue volume in addition to the plasma volume. Because data on volumes of tissues and plasma are available in the literature for mammals, the other input parameters needed to estimate V(ss) are the P(t:p)'s, which can potentially be predicted with established tissue composition-based equations. In vitro data on drug lipophilicity and plasma protein binding are the input parameters used in these equations. Such a mechanism-based approach would be particularly useful to provide first-cut estimates of V(ss) prior to any in vivo studies and to explore potential unexpected deviations between sets of predicted and in vivo V(ss) data, when the in vivo data become available during the drug development process. The objective of the present study was to use tissue composition-based equations to predict rat and human V(ss) prior to in vivo studies for 123 structurally unrelated compounds (acids, bases, and neutrals). The predicted data were compared with in vivo data obtained from the literature or at Roche. Overall, the average ratio of predicted-to-experimental rat and human V(ss) values was 1.06 (SD = 0.817, r = 0.78, n = 147). In fact, 80% of all predicted values were within a factor of two of the corresponding experimental values. The drugs can therefore be separated into two groups. The first group contains 98 drugs for which the predicted V(ss) were within a factor of two of those experimentally determined (average ratio of 1.01, SD = 0.39, r = 0.93, n = 118), and the second group includes 25 other drugs for which the predicted and experimental V(ss) differ by a factor larger than two (average ratio of 1.32, SD = 1.74, r = 0.42, n = 29). Thus, additional relevant distribution processes were neglected in predicting V(ss) of drugs of the second group. This was true especially in the case of some cationic-amphiphilic bases. The present study is the first attempt to develop and validate a mechanistic distribution model for predicting rat and human V(ss) of drugs prior to in vivo studies.


Assuntos
Modelos Biológicos , Modelos Químicos , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/metabolismo , Farmacocinética , Distribuição Tecidual , Animais , Fenômenos Químicos , Físico-Química , Humanos , Ratos
20.
J Pharm Sci ; 92(10): 1990-2007, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14502539

RESUMO

The objective of this study was to use in synergy physiologically based and empirical approaches to estimate the drug-specific input parameters of PBPK models of disposition to simulate the plasma concentration-time profile of epiroprim in human. The estimated input parameters were the tissue:plasma partition coefficients (Pt:p) for distribution and the blood clearance (CL) for the in vivo conditions. Epiroprim represents a challenge for such methods, because it shows large interspecies differences in its pharmacokinetic properties. Two approaches were used to predict the human Pt:p values: the tissue composition model (TCM) and the "Arundel approach" based on the volume of distribution at steady state (Vdss) determined in vivo in the rat. CL in human was predicted by (1) conventional allometric scaling of in vivo animal clearances (CAS), (2) physiologically based direct scaling up of in vitro hepatocyte data (DSU), and (3) allometric scaling of animal intrinsic in vivo blood CL normalized by the ratios of animal:human intrinsic clearances determined in vitro with hepatocytes (NAS). The performance of prediction was assessed by comparing separately the above pharmacokinetic parameters (Vdss estimated from the Pt:p values and blood CL) with the corresponding in vivo data obtained from the plasma kinetic profiles. These input parameters were used in PBPK models, and the resulting plasma concentration-time profiles of epiroprim were compared with those observed in rat and human. Previously to the construction of the human PBPK model, a model for the rat was also developed to gain more confidence on the model structure and assumptions. Overall, using the TCM and the NAS for the parameterization of the distribution and clearance, respectively, the PBPK model gave the more accurate predictions of epiroprim's disposition in human. This study represents therefore an attractive approach, which may potentially help the clinical candidate selection.


Assuntos
Trimetoprima/análogos & derivados , Trimetoprima/farmacocinética , Animais , Peso Corporal , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Cães , Hepatócitos/metabolismo , Humanos , Macaca , Masculino , Camundongos , Modelos Biológicos , Especificidade da Espécie , Fatores de Tempo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA