Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
AAPS J ; 24(6): 116, 2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-36376552

RESUMO

The aim of this research was to develop a reliable non-radiometric method to measure the residual blood in tissue without the need for perfusion or radiometric measurements in biodistribution studies. It was found that the perfusion method not only was ineffective in removing blood from tissue, but also introduced additional variability in the determination of tissue drug exposure and was not reproducible across studies. In addition, the use of hemoglobin as an endogenous protein and biomarker for tissue blood content was studied and it was found that hemoglobin measurement in tissue was not a reliable and effective approach for determination of residual blood level in tissue. To evaluate an alternative method for addressing the tissue blood level in biodistribution studies, animals were dosed with a Residual Blood Determinant Reagent (RBDR) 5 min prior to tissue harvesting. The level of RBDR, an exogenous protein, was measured in whole blood homogenate and in tissue lysate. Based on the level of the RBDR, the vascular blood volume (VBV) in tissue was calculated and then the tissue exposures were corrected based on the blood volumes. The tissue VBVs measured by the RBDR method were comparable with the literature values obtained by radiometric measurements.


Assuntos
Volume Sanguíneo , Hemoglobinas , Animais , Distribuição Tecidual , Hemoglobinas/metabolismo
2.
Bioanalysis ; 14(9): 581-588, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35548877

RESUMO

Aim: To develop a method for the quantitation of effector functionless mouse surrogate IgG1 drug molecules in mouse matrices. Materials & methods: A panel of antibodies that bound specifically to N297G mutation-containing mouse IgG molecules was generated in rats. The panel was screened to identify an antibody that could be used as both the capture and detection reagent in an electrochemiluminescent immunoassay. Results & conclusion: The quantitative assay developed with the N297G-specific antibody passed acceptance criteria across multiple IgG1 fragment crystallizable (Fc)-containing protein formats and provides accurate quantitation of the total levels of mouse surrogate protein Fc present in in vivo mouse serum samples. These results are useful in understanding drug integrity and the development of precise pharmacokinetic/pharmacodynamic relationships.


Assuntos
Anticorpos Monoclonais , Imunoglobulina G , Animais , Imunoensaio/métodos , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Testes Imunológicos , Camundongos , Ratos , Soro
3.
J Biol Chem ; 295(30): 10446-10455, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32518163

RESUMO

Transthyretin (TTR) is an abundant homotetrameric serum protein and was selected here for engineering higher-valency molecules because of its compact size, simple structure, and natural propensity to tetramerize. To demonstrate this utility, we fused TTR to the C terminus of conatumumab, an antibody that targets tumor necrosis factor-related apoptosis-inducing ligand receptor 2, as heavy chains to form antibody dimers and Fab heavy chains to form Fab tetramers. Moreover, we used constant heavy domain 3 heterodimerization substitutions to create TTR-mediated conatumumab tetramers. The conatumumab-TTR fusions displayed substantially enhanced potency in cell-based assays, as well as in murine tumor xenograft models. We conclude that antibody-TTR fusions may provide a powerful platform for multimerizing antibody and Fab fragments to enhance the capabilities of human therapeutics that benefit from target clustering and higher-order antigen-binding valency.


Assuntos
Anticorpos Monoclonais , Antineoplásicos Imunológicos , Fragmentos Fab das Imunoglobulinas , Neoplasias Experimentais , Pré-Albumina , Multimerização Proteica , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/farmacologia , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Pré-Albumina/genética , Pré-Albumina/farmacocinética , Pré-Albumina/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
MAbs ; 11(6): 1025-1035, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31185801

RESUMO

Accelerated development of monoclonal antibody (mAb) tool reagents is an essential requirement for the successful advancement of therapeutic antibodies in today's fast-paced and competitive drug development marketplace. Here, we describe a direct, flexible, and rapid nanofluidic optoelectronic single B lymphocyte antibody screening technique (NanOBlast) applied to the generation of anti-idiotypic reagent antibodies. Selectively enriched, antigen-experienced murine antibody secreting cells (ASCs) were harvested from spleen and lymph nodes. Subsequently, secreted mAbs from individually isolated, single ASCs were screened directly using a novel, integrated, high-content culture, and assay platform capable of manipulating living cells within microfluidic chip nanopens using structured light. Single-cell polymerase chain reaction-based molecular recovery on select anti-idiotypic ASCs followed by recombinant IgG expression and enzyme-linked immunosorbent assay (ELISA) characterization resulted in the recovery and identification of a diverse and high-affinity panel of anti-idiotypic reagent mAbs. Combinatorial ELISA screening identified both capture and detection mAbs, and enabled the development of a sensitive and highly specific ligand binding assay capable of quantifying free therapeutic IgG molecules directly from human patient serum, thereby facilitating important drug development decision-making. The ASC import, screening, and export discovery workflow on the chip was completed within 5 h, while the overall discovery workflow from immunization to recombinantly expressed IgG was completed in under 60 days.


Assuntos
Anticorpos Monoclonais Murinos/imunologia , Linfócitos B/imunologia , Imunoglobulina G/imunologia , Animais , Linfócitos B/citologia , Células CHO , Cricetulus , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos
5.
PLoS One ; 14(5): e0217061, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31120944

RESUMO

In this study we compared the pharmacokinetic profile of four unrelated antibodies, which do not bind to mammalian antigens, in IgG1 and IgG2 frameworks in both rats and non-human primates (NHP). This allowed for extensive cross comparison of the impact of antibody isotype, complementarity determining regions (CDR) and model species on pharmacokinetics without the confounding influence of antigen binding in the hosts. While antibody isotype had no significant impact on the pharmacokinetics, the CDRs do alter the profile, and there is an inverse correlation between the neonatal Fc receptor (FcRn) affinity and pharmacokinetic performance. Faster clearance rates were also associated with higher isoelectric points; however, although this panel of antibodies all possess basic isoelectric points, ranging from 8.44 to 9.18, they also have exceptional in vivo half-lives, averaging 369 hours, and low clearance rates, averaging 0.18 ml/h/kg in NHPs. This pattern of pharmacokinetic characteristics was conserved between rats and NHPs.


Assuntos
Anticorpos/metabolismo , Imunoglobulina G/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Células CHO , Regiões Determinantes de Complementaridade/imunologia , Cricetinae , Cricetulus , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Masculino , Camundongos , Farmacocinética , Primatas/imunologia , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Receptores Fc/imunologia
6.
ACS Chem Biol ; 14(4): 806-818, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30875193

RESUMO

Drug discovery research on new pain targets with human genetic validation, including the voltage-gated sodium channel NaV1.7, is being pursued to address the unmet medical need with respect to chronic pain and the rising opioid epidemic. As part of early research efforts on this front, we have previously developed NaV1.7 inhibitory peptide-antibody conjugates with tarantula venom-derived GpTx-1 toxin peptides with an extended half-life (80 h) in rodents but only moderate in vitro activity (hNaV1.7 IC50 = 250 nM) and without in vivo activity. We identified the more potent peptide JzTx-V from our natural peptide collection and improved its selectivity against other sodium channel isoforms through positional analogueing. Here we report utilization of the JzTx-V scaffold in a peptide-antibody conjugate and architectural variations in the linker, peptide loading, and antibody attachment site. We found conjugates with 100-fold improved in vitro potency relative to those of complementary GpTx-1 analogues, but pharmacokinetic and bioimaging analyses of these JzTx-V conjugates revealed a shorter than expected plasma half-life in vivo with accumulation in the liver. In an attempt to increase circulatory serum levels, we sought the reduction of the net +6 charge of the JzTx-V scaffold while retaining a desirable NaV in vitro activity profile. The conjugate of a JzTx-V peptide analogue with a +2 formal charge maintained NaV1.7 potency with 18-fold improved plasma exposure in rodents. Balancing the loss of peptide and conjugate potency associated with the reduction of net charge necessary for improved target exposure resulted in a compound with moderate activity in a NaV1.7-dependent pharmacodynamic model but requires further optimization to identify a conjugate that can fully engage NaV1.7 in vivo.


Assuntos
Imunoconjugados , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Peptídeos/química , Venenos de Aranha/química , Bloqueadores do Canal de Sódio Disparado por Voltagem , Animais , Anticorpos/química , Descoberta de Drogas , Humanos , Imunoconjugados/química , Imunoconjugados/farmacocinética , Masculino , Camundongos , Terapia de Alvo Molecular , Canal de Sódio Disparado por Voltagem NAV1.7/imunologia , Peptídeos/farmacocinética , Venenos de Aranha/farmacocinética , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacocinética
7.
Nat Commun ; 9(1): 2394, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29921922

RESUMO

Diminished growth factor signaling improves longevity in laboratory models, while a reduction in the somatotropic axis is favorably linked to human aging and longevity. Given the conserved role of this pathway on lifespan, therapeutic strategies, such as insulin-like growth factor-1 receptor (IGF-1R) monoclonal antibodies (mAb), represent a promising translational tool to target human aging. To this end, we performed a preclinical study in 18-mo-old male and female mice treated with vehicle or an IGF-1R mAb (L2-Cmu, Amgen Inc), and determined effects on aging outcomes. Here we show that L2-Cmu preferentially improves female healthspan and increases median lifespan by 9% (P = 0.03) in females, along with a reduction in neoplasms and inflammation (P ≤ 0.05). Thus, consistent with other models, targeting IGF-1R signaling appears to be most beneficial to females. Importantly, these effects could be achieved at advanced ages, suggesting that IGF-1R mAbs could represent a promising therapeutic candidate to delay aging.


Assuntos
Anticorpos Monoclonais/farmacologia , Longevidade/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Receptor IGF Tipo 1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/prevenção & controle , Receptor IGF Tipo 1/imunologia , Receptor IGF Tipo 1/metabolismo , Fatores Sexuais , Carga Tumoral/efeitos dos fármacos
8.
J Biol Chem ; 292(5): 1865-1875, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-27994062

RESUMO

IgG isotypes can differentially bind to Fcγ receptors and complement, making the selection of which isotype to pursue for development of a particular therapeutic antibody important in determining the safety and efficacy of the drug. IgG2 and IgG4 isotypes have significantly lower binding affinity to Fcγ receptors. Recent evidence suggests that the IgG2 isotype is not completely devoid of effector function, whereas the IgG4 isotype can undergo in vivo Fab arm exchange leading to bispecific antibody and off-target effects. Here an attempt was made to engineer an IgG1-based scaffold lacking effector function but with stability equivalent to that of the parent IgG1. Care was taken to ensure that both stability and lack of effector function was achieved with a minimum number of mutations. Among the Asn297 mutants that result in lack of glycosylation and thus loss of effector function, we demonstrate that the N297G variant has better stability and developability compared with the N297Q or N297A variants. To further improve the stability of N297G, we introduced a novel engineered disulfide bond at a solvent inaccessible location in the CH2 domain. The resulting scaffold has stability greater than or equivalent to that of the parental IgG1 scaffold. Extensive biophysical analyses and pharmacokinetic (PK) studies in mouse, rat, and monkey further confirmed the developability of this unique scaffold, and suggest that it could be used for all Fc containing therapeutics (e.g. antibodies, bispecific antibodies, and Fc fusions) requiring lack of effector function or elimination of binding to Fcγ receptors.


Assuntos
Substituição de Aminoácidos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/genética , Mutação de Sentido Incorreto , Animais , Humanos , Macaca fascicularis , Camundongos , Ratos
9.
Nat Commun ; 7: 11505, 2016 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-27230681

RESUMO

Inhibition of the Wnt antagonist sclerostin increases bone mass in patients with osteoporosis and in preclinical animal models. Here we show increased levels of the Wnt antagonist Dickkopf-1 (DKK-1) in animals treated with sclerostin antibody, suggesting a negative feedback mechanism that limits Wnt-driven bone formation. To test our hypothesis that co-inhibition of both factors further increases bone mass, we engineer a first-in-class bispecific antibody with single residue pair mutations in the Fab region to promote efficient and stable cognate light-heavy chain pairing. We demonstrate that dual inhibition of sclerostin and DKK-1 leads to synergistic bone formation in rodents and non-human primates. Furthermore, by targeting distinct facets of fracture healing, the bispecific antibody shows superior bone repair activity compared with monotherapies. This work supports the potential of this agent both for treatment and prevention of fractures and offers a promising therapeutic approach to reduce the burden of low bone mass disorders.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Fraturas Ósseas/tratamento farmacológico , Fraturas Ósseas/fisiopatologia , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Densidade Óssea , Modelos Animais de Doenças , Feminino , Fraturas Ósseas/genética , Fraturas Ósseas/metabolismo , Glicoproteínas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Macaca fascicularis , Masculino , Camundongos , Camundongos Knockout , Osteogênese/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Via de Sinalização Wnt/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
10.
Anal Biochem ; 470: 52-60, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25447458

RESUMO

Antibodies are critical tools for protein bioanalysis; their quality and performance dictate the caliber and robustness of ligand binding assays. After immunization, polyclonal B cells generate a diverse antibody repertoire against constant and variable regions of the therapeutic antibody immunogen. Herein we describe a comprehensive and multifactorial screening strategy to eliminate undesirable constant region-specific antibodies and select for anti-idiotypic antibodies with specificity for the unique variable region. Application of this strategy is described for the therapeutic antibody Mab-A case study. Five different factors were evaluated to select a final antibody pair for the quantification of therapeutics in biological matrices: (i) matrix effect in preclinical and clinical matrices, (ii) assay sensitivity with lower limit of quantification goal of single-digit ng/ml (low pM) at a signal-to-background ratio greater than 5, (iii) epitope distinction or nonbridging antibody pair, (iv) competition with target and inhibitory capacity enabling measurement of free drug, and (v) neutralizing bioactivity using bioassay. The selected antibody pair demonstrated superior assay sensitivity with no or minimal matrix effect in common biological samples, recognized two distinct binding epitopes on the therapeutic antibody variable region, and featured inhibitory and neutralizing effects with respect to quantification of free drug levels.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Bioensaio/métodos , Animais , Humanos , Indicadores e Reagentes , Ligantes , Limite de Detecção , Camundongos
11.
J Immunotoxicol ; 12(2): 164-73, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24990272

RESUMO

ORAI1 is the pore-forming component of calcium release-activated calcium (CRAC) channels. CRAC channels are the primary route for calcium ion (Ca(2+)) entry into T-cells following antigen stimulation. This Ca(2+) entry induces proliferation and cytokine production through activation of calcineurin and the nuclear factor of activated T-cells (NFAT) transcription factor along with subsequent cytokine-related genes. It was hypothesized that the in vivo inhibition of T-cell function by blocking ORAI1 or calcineurin would lead to similar functional consequences. To test this hypothesis the activity of 2C1.1, a fully human anti-ORAI1 monoclonal antibody, and cyclosporin A (CsA) were tested in vivo for their suppressive effect on T-cell-derived cytokine production and a T-cell-dependent antibody response (TDAR) using sheep red blood cells (SRBC) in cynomolgus monkeys. Despite showing similar inhibition of ex vivo interleukin (IL)-2 production by stimulated T-cells, both molecules exhibited different pharmacologic effects on the SRBC antibody response. CsA blocked the development of SRBC-specific antibodies, while 2C1.1 failed to inhibit the antigen-specific antibody response. These surprising observations suggest that full inhibition of the CRAC channel is required to inhibit a functional immune response, consistent with findings from human patients with loss of function mutations in ORAI1.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Canais de Cálcio/metabolismo , Ciclosporina/administração & dosagem , Macaca fascicularis , Linfócitos T/imunologia , Animais , Formação de Anticorpos , Calcineurina/metabolismo , Canais de Cálcio/imunologia , Bovinos , Células Cultivadas , Eritrócitos/imunologia , Humanos , Interleucina-2/metabolismo , Ativação Linfocitária , Masculino , Fatores de Transcrição NFATC/metabolismo , Proteína ORAI1 , Ovinos
12.
Bioanalysis ; 6(8): 1081-91, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24830892

RESUMO

BACKGROUND: The accuracy of highly sensitive biomarker methods is often confounded by the presence of various circulating endogenous factors in samples causing matrix effects. METHOD: This article outlines two different biomarker methods: hepcidin enzyme-linked immunosorbent assay (ELISA) for which an orthogonal assessment of ELISA to liquid chromatography-tandem mass spectrometry was performed to examine the potential matrix effect, and sclerostin ELISA to evaluate the matrix effect. RESULTS: Although the potential interfering effects of the endogenous hepcidin variants (prohepcidin and clipped) showed that these proteins had >30% immunoreactivity in ELISA, the hepcidin ELISA preferentially measures full-length hepcidin when the molar ratios of full-length to variants remain >1. The correlation of ELISA to liquid chromatography-tandem mass spectrometry results showed full-length hepcidin as the major form in diseased populations. CONCLUSION: A fit-for-for-purpose assessment of matrix effect/selectivity was also performed for each method. This article demonstrates the utility of a fit-for-purpose approach to assess the validity of biomarker methods in evaluating the interconnected parameters of matrix effects, sensitivity and selectivity.


Assuntos
Biomarcadores/sangue , Cromatografia Líquida/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Espectrometria de Massas em Tandem/métodos , Humanos , Sensibilidade e Especificidade
13.
Toxicol Pathol ; 42(3): 524-39, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23674391

RESUMO

We recently reported results that erythropoiesis-stimulating agent (ESA)-related thrombotic toxicities in preclinical species were not solely dependent on a high hematocrit (HCT) but also associated with increased ESA dose level, dose frequency, and dosing duration. In this article, we conclude that sequelae of an increased magnitude of ESA-stimulated erythropoiesis potentially contributed to thrombosis in the highest ESA dose groups. The results were obtained from two investigative studies we conducted in Sprague-Dawley rats administered a low (no thrombotic toxicities) or high (with thrombotic toxicities) dose level of a hyperglycosylated analog of recombinant human erythropoietin (AMG 114), 3 times weekly for up to 9 days or for 1 month. Despite similarly increased HCT at both dose levels, animals in the high-dose group had an increased magnitude of erythropoiesis measured by spleen weights, splenic erythropoiesis, and circulating reticulocytes. Resulting prothrombotic risk factors identified predominantly or uniquely in the high-dose group were higher numbers of immature reticulocytes and nucleated red blood cells in circulation, severe functional iron deficiency, and increased intravascular destruction of iron-deficient reticulocyte/red blood cells. No thrombotic events were detected in rats dosed up to 9 days suggesting a sustained high HCT is a requisite cofactor for development of ESA-related thrombotic toxicities.


Assuntos
Eritropoese/efeitos dos fármacos , Eritropoetina/farmacologia , Eritropoetina/toxicidade , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/toxicidade , Análise de Variância , Animais , Plaquetas , Eritrócitos , Eritropoetina/administração & dosagem , Hematócrito , Humanos , Ferro/sangue , Ferro/metabolismo , Masculino , Policitemia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Reticulócitos
14.
Toxicol Pathol ; 42(3): 540-54, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23674392

RESUMO

We previously reported an increased incidence of thrombotic toxicities in Sprague-Dawley rats administered the highest dose level of a hyperglycosylated analog of recombinant human erythropoietin (AMG 114) for 1 month as not solely dependent on high hematocrit (HCT). Thereafter, we identified increased erythropoiesis as a prothrombotic risk factor increased in the AMG 114 high-dose group with thrombotic toxicities, compared to a low-dose group with no toxicities but similar HCT. Here, we identified pleiotropic cytokines as prothrombotic factors associated with AMG 114 dose level. Before a high HCT was achieved, rats in the AMG 114 high, but not the low-dose group, had imbalanced hemostasis (increased von Willebrand factor and prothrombin time, decreased antithrombin III) coexistent with cytokines implicated in thrombosis: monocyte chemotactic protein 1 (MCP-1), MCP-3, tissue inhibitor of metalloproteinases 1, macrophage inhibitory protein-2, oncostatin M, T-cell-specific protein, stem cell factor, vascular endothelial growth factor, and interleukin-11. While no unique pathway to erythropoiesis stimulating agent-related thrombosis was identified, cytokines associated with increased erythropoiesis contributed to a prothrombotic intravascular environment in the AMG 114 high-dose group, but not in lower dose groups with a similar high HCT.


Assuntos
Citocinas/sangue , Citocinas/metabolismo , Eritropoese/efeitos dos fármacos , Eritropoetina/farmacologia , Proteínas Recombinantes/farmacologia , Animais , Eritropoetina/química , Hematócrito , Humanos , Masculino , Policitemia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Reticulócitos , Trombose
15.
Blood ; 122(17): 3054-61, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23945155

RESUMO

Iron maldistribution has been implicated in the etiology of many diseases including the anemia of inflammation (AI), atherosclerosis, diabetes, and neurodegenerative disorders. Iron metabolism is controlled by hepcidin, a 25-amino-acid peptide. Hepcidin is induced by inflammation and causes iron to be sequestered within cells of the reticuloendothelial system, suppressing erythropoiesis and blunting the activity of erythropoiesis stimulating agents (ESAs). For this reason, neutralization of hepcidin has been proposed as a therapeutic treatment of AI. The aim of the current work was to generate fully human anti-hepcidin antibodies (Abs) as a potential human therapeutic for the treatment of AI and other iron maldistribution disorders. An enzyme-linked immunosorbent assay was established using these Abs to identify patients likely to benefit from either ESAs or anti-hepcidin agents. Using human hepcidin knock-in mice, the mechanism of action of the Abs was shown to be due to an increase in available serum iron leading to enhanced red cell hemoglobinization. One of the Abs, 12B9m, was validated in a mouse model of AI and demonstrated to modulate serum iron in cynomolgus monkeys. The 12B9m Ab was deemed to be an appropriate candidate for use as a potential therapeutic to treat AI in patients with kidney disease or cancer.


Assuntos
Anemia Ferropriva/tratamento farmacológico , Anticorpos Neutralizantes/farmacologia , Eritrócitos/efeitos dos fármacos , Hemoglobinas/biossíntese , Ferro/sangue , Anemia Ferropriva/sangue , Anemia Ferropriva/patologia , Animais , Anticorpos Neutralizantes/biossíntese , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Ensaio de Imunoadsorção Enzimática , Eritrócitos/metabolismo , Eritrócitos/patologia , Eritropoese/efeitos dos fármacos , Feminino , Hematínicos/farmacologia , Humanos , Inflamação/prevenção & controle , Macaca fascicularis , Masculino , Camundongos
16.
Monoclon Antib Immunodiagn Immunother ; 32(3): 149-61, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23750472

RESUMO

Thrombopoietin and its cognate receptor, c-Mpl, are the primary molecular regulators of megakaryocytopoiesis and platelet production. To date the pattern of c-Mpl expression in human solid tumors and the distribution and biochemical properties of c-Mpl proteins in hematopoietic tissues are largely unknown. We have recently developed highly specific mouse monoclonal antibodies (MAb) against human c-Mpl. In this study we used these antibodies to demonstrate the presence of full-length and truncated human c-Mpl proteins in various megakaryocytic cell types, and their absence in over 100 solid tumor cell lines and in the 12 most common primary human tumor types. Quantitative assays showed a cell context-dependent distribution of full-length and truncated c-Mpl proteins. All forms of human c-Mpl protein were found to be modified with extensive N-linked glycosylation but different degrees of sialylation and O-linked glycosylation. Of note, different variants of full-length c-Mpl protein exhibiting differential glycosylation were expressed in erythromegakaryocytic leukemic cell lines and in platelets from healthy human donors. This work provides a comprehensive analysis of human c-Mpl mRNA and protein expression on normal and malignant hematopoietic and non-hematopoietic cells and demonstrates the multiple applications of several novel anti-c-Mpl antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Megacariócitos/metabolismo , Proteínas de Neoplasias/imunologia , Receptores de Trombopoetina/imunologia , Plaquetas , Linhagem Celular Tumoral , Proliferação de Células , Glicosilação , Humanos , Megacariócitos/imunologia , Neoplasias/imunologia , Neoplasias/metabolismo , Trombopoese , Trombopoetina
17.
Anal Chem ; 85(5): 2731-8, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23373459

RESUMO

Novel protein therapeutics have become increasingly important modalities for treating diseases. Such therapeutics include recombinant fusions of pharmacoactive polypeptides to half-life extenders such as monoclonal antibodies, fragments of antibodies, and albumin. Half-life extension can also be achieved via chemical attachment to polymers such as polyethylene glycol. Any of these therapeutics may be susceptible to biotransformation, most notably in vivo proteolytic truncation, and it is vital to understand this phenomenon during early drug development to ensure correct pharmacokinetic profiling and optimize the in vivo stability through re-engineering. In this paper, we describe an integrated approach that combines differential enzyme-linked immunosorbent assay (ELISA) with ligand-binding-mass spectrometry (LB-MS) to provide a thorough understanding of the biotransformation of novel protein therapeutics. Differential ELISA allows for a fast, high-throughput means to reveal gross in vivo proteolytic liabilities. Ensuing LB-MS analysis provides higher resolution details such as specific vulnerable loci to allow design refinement of the molecule. In this work, the power of the approach is elucidated by application to the optimization of a promising drug candidate, FGF21.


Assuntos
Ensaio de Imunoadsorção Enzimática/métodos , Fatores de Crescimento de Fibroblastos/metabolismo , Espectrometria de Massas/métodos , Biotransformação , Cromatografia Líquida , Fatores de Crescimento de Fibroblastos/uso terapêutico , Humanos , Ligantes
18.
AAPS J ; 15(2): 337-46, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23233266

RESUMO

We have recently developed a general liquid chromatography-tandem mass spectrometric (LC-MS/MS) method using a stable isotope-labeled (SIL) monoclonal antibody (mAb) as an internal standard (IS) for single-analyte quantification of mAb (Li et al. Anal Chem 84(3):1267-1273, 2012). The method offers an advantage over ligand binding assay in reducing the time and resources needed for bioanalytical support in preclinical stages of drug development. In this paper, we report another marked increase in assay efficiency for multi-analyte bioanalysis using unique surrogate peptides for each analyte and the strategic choice of the SIL-IS peptide. The method was qualified for the simultaneous determinations of four mAbs in rat plasma and applied to samples from discrete- and cassette-dosed rats. The pharmacokinetic parameters of the four mAbs of cassette dosing were comparable to those of discrete dosing and of enzyme-linked immunosorbent assay results. Although there may be limitations and special considerations for cassette-dosing of biologics, these results demonstrate the robust performance of the multi-analyte LC-MS/MS method allowing cassette-dosing that would ultimately reduce animal use and improve efficiency.


Assuntos
Anticorpos Monoclonais/sangue , Produtos Biológicos/sangue , Cromatografia Líquida , Espectrometria de Massas em Tandem , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Produtos Biológicos/administração & dosagem , Produtos Biológicos/farmacocinética , Biotransformação , Calibragem , Cromatografia Líquida/normas , Combinação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Injeções Subcutâneas , Fragmentos de Peptídeos/sangue , Controle de Qualidade , Ratos , Ratos Sprague-Dawley , Padrões de Referência , Espectrometria de Massas em Tandem/normas
19.
J Am Soc Nephrol ; 24(1): 66-76, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23184054

RESUMO

Wnt-modulator in surface ectoderm (WISE) is a secreted modulator of Wnt signaling expressed in the adult kidney. Activation of Wnt signaling has been observed in renal transplants developing interstitial fibrosis and tubular atrophy; however, whether WISE contributes to chronic changes is not well understood. Here, we found moderate to high expression of WISE mRNA in a rat model of renal transplantation and in kidneys from normal rats. Treatment with a neutralizing antibody against WISE improved proteinuria and graft function, which correlated with higher levels of ß-catenin protein in kidney allografts. In addition, treatment with the anti-WISE antibody reduced infiltration of CD68(+) macrophages and CD8(+) T cells, attenuated glomerular and interstitial injury, and decreased biomarkers of renal injury. This treatment reduced expression of genes involved in immune responses and in fibrogenic pathways. In summary, WISE contributes to renal dysfunction by promoting tubular atrophy and interstitial fibrosis.


Assuntos
Proteínas de Transporte/metabolismo , Transplante de Rim , Rim/metabolismo , Insuficiência Renal/prevenção & controle , Proteínas Wnt/metabolismo , Actinas/metabolismo , Animais , Anticorpos/uso terapêutico , Biomarcadores/urina , Caderinas/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Rim/imunologia , Testes de Função Renal , Masculino , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Insuficiência Renal/urina , beta Catenina/metabolismo
20.
Bioanalysis ; 4(20): 2513-23, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23157359

RESUMO

The majority of biotherapeutic drugs act on specific targets, which may serve as biomarkers to be evaluated for target engagement and validation. Together with subsequent pathway biomarkers, these data can provide proof-of-mechanism and understanding of the biological drug affect. A major task during early development is to predict, for the first first time in human clinical trials, the starting dose and simulate the PK/PD relationship. However, determinations of the biotherapeutic drug and target concentrations are not straightforward due to temporal changes of drug-target binding and challenges in developing reliable methods to measure the free and total drug and target. Herein, the bioanalysis of the target biomarker and the biotherapeutics in the context of PK/PD relevancy during drug development is reviewed. Binding of the target to the biotherapeutic will affect target clearance and drug disposition, resulting in nonlinear PK. Reliable and specific methods are crucial for the correct PK/PD modeling and interpretation.


Assuntos
Anticorpos Monoclonais/farmacocinética , Biomarcadores/análise , Animais , Anticorpos Monoclonais/imunologia , Descoberta de Drogas , Modelos Biológicos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...