Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 270
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Cell ; 176(3): 479-490.e12, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30639100

RESUMO

The angiotensin II (AngII) type 1 receptor (AT1R) is a critical regulator of cardiovascular and renal function and is an important model for studies of G-protein-coupled receptor (GPCR) signaling. By stabilizing the receptor with a single-domain antibody fragment ("nanobody") discovered using a synthetic yeast-displayed library, we determined the crystal structure of active-state human AT1R bound to an AngII analog with partial agonist activity. The nanobody binds to the receptor's intracellular transducer pocket, stabilizing the large conformational changes characteristic of activated GPCRs. The peptide engages the AT1R through an extensive interface spanning from the receptor core to its extracellular face and N terminus, remodeling the ligand-binding cavity. Remarkably, the mechanism used to propagate conformational changes through the receptor diverges from other GPCRs at several key sites, highlighting the diversity of allosteric mechanisms among GPCRs. Our structure provides insight into how AngII and its analogs stimulate full or biased signaling, respectively.


Assuntos
Receptor Tipo 1 de Angiotensina/metabolismo , Anticorpos de Domínio Único/farmacologia , Angiotensina II , Bloqueadores do Receptor Tipo 1 de Angiotensina II/metabolismo , Arrestinas/metabolismo , Células HEK293 , Humanos , Fragmentos de Imunoglobulinas/farmacologia , Conformação Proteica , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anticorpos de Domínio Único/metabolismo , beta-Arrestinas/metabolismo
2.
Trends Biochem Sci ; 43(7): 533-546, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29735399

RESUMO

G protein-coupled receptors (GPCRs) recognize a diverse array of extracellular stimuli, and they mediate a broad repertoire of signaling events involved in human physiology. Although the major effort on targeting GPCRs has typically been focused on their extracellular surface, a series of recent developments now unfold the possibility of targeting them from the intracellular side as well. Allosteric modulators binding to the cytoplasmic surface of GPCRs have now been described, and their structural mechanisms are elucidated by high-resolution crystal structures. Furthermore, pepducins, aptamers, and intrabodies targeting the intracellular face of GPCRs have also been successfully utilized to modulate receptor signaling. Moreover, small molecule compounds, aptamers, and synthetic intrabodies targeting ß-arrestins have also been discovered to modulate GPCR endocytosis and signaling. Here, we discuss the emerging paradigm of intracellular targeting of GPCRs, and outline the current challenges, potential opportunities, and future outlook in this particular area of GPCR biology.


Assuntos
Endocitose , Modelos Moleculares , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Regulação Alostérica/efeitos dos fármacos , Animais , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/metabolismo , Aptâmeros de Nucleotídeos/farmacologia , Sítios de Ligação , Endocitose/efeitos dos fármacos , Humanos , Fragmentos de Imunoglobulinas/química , Fragmentos de Imunoglobulinas/metabolismo , Fragmentos de Imunoglobulinas/farmacologia , Ligantes , Lipopeptídeos/química , Lipopeptídeos/metabolismo , Lipopeptídeos/farmacologia , Conformação Proteica , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/química , Transdução de Sinais/efeitos dos fármacos
3.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34681796

RESUMO

Neutrophil elastase (NE) is a serine protease released during neutrophil maturation. High levels of NE are related to lung tissue damage and poor prognosis in cancer; thus, NE is a potential target for therapeutic immunotherapy for multiple lung diseases and cancers. Here, we isolate and characterize two high-affinity, specific, and noncompetitive anti-NE antibodies Fab 1C10 and VH 1D1.43 from two large phage-displayed human Fab and VH libraries. After fusion with human IgG1 Fc, both of them (VH-Fc 1D1.43 and IgG1 1C10) inhibit NE enzymatic activity with VH-Fc 1D1.43 showing comparable inhibitory effects to that of the small molecule NE inhibitor SPCK and IgG1 1C10 exhibiting even higher (2.6-fold) activity than SPCK. Their epitopes, as mapped by peptide arrays combined with structural modeling, indicate different mechanisms for blocking NE activity. Both VH-Fc and IgG1 antibodies block NE uptake by cancer cells and fibroblast differentiation. VH-Fc 1D1.43 and IgG1 1C10 are promising for the antibody-based immunotherapy of cancer and inflammatory diseases.


Assuntos
Inflamação/tratamento farmacológico , Elastase de Leucócito/imunologia , Neoplasias/tratamento farmacológico , Proteínas Secretadas Inibidoras de Proteinases/uso terapêutico , Células Cultivadas , Mapeamento de Epitopos , Humanos , Domínios de Imunoglobulina/fisiologia , Fragmentos de Imunoglobulinas/química , Fragmentos de Imunoglobulinas/farmacologia , Fragmentos de Imunoglobulinas/uso terapêutico , Imunoterapia/métodos , Inflamação/imunologia , Elastase de Leucócito/antagonistas & inibidores , Masculino , Modelos Moleculares , Terapia de Alvo Molecular , Neoplasias/imunologia , Células PC-3 , Estrutura Secundária de Proteína , Proteínas Secretadas Inibidoras de Proteinases/química , Proteínas Secretadas Inibidoras de Proteinases/farmacologia
4.
Pharmacol Rev ; 68(3): 603-30, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27329045

RESUMO

Proteinase 3 (PR3) has received great scientific attention after its identification as the essential antigenic target of antineutrophil cytoplasm antibodies in Wegener's granulomatosis (now called granulomatosis with polyangiitis). Despite many structural and functional similarities between neutrophil elastase (NE) and PR3 during biosynthesis, storage, and extracellular release, unique properties and pathobiological functions have emerged from detailed studies in recent years. The development of highly sensitive substrates and inhibitors of human PR3 and the creation of PR3-selective single knockout mice led to the identification of nonredundant roles of PR3 in cell death induction via procaspase-3 activation in cell cultures and in mouse models. According to a study in knockout mice, PR3 shortens the lifespan of infiltrating neutrophils in tissues and accelerates the clearance of aged neutrophils in mice. Membrane exposure of active human PR3 on apoptotic neutrophils reprograms the response of macrophages to phagocytosed neutrophils, triggers secretion of proinflammatory cytokines, and undermines immune silencing and tissue regeneration. PR3-induced disruption of the anti-inflammatory effect of efferocytosis may be relevant for not only granulomatosis with polyangiitis but also for other autoimmune diseases with high neutrophil turnover. Inhibition of membrane-bound PR3 by endogenous inhibitors such as the α-1-protease inhibitor is comparatively weaker than that of NE, suggesting that the adverse effects of unopposed PR3 activity resurface earlier than those of NE in individuals with α-1-protease inhibitor deficiency. Effective coverage of PR3 by anti-inflammatory tools and simultaneous inhibition of both PR3 and NE should be most promising in the future.


Assuntos
Anti-Inflamatórios/farmacologia , Fragmentos de Imunoglobulinas/farmacologia , Fragmentos de Imunoglobulinas/uso terapêutico , Mieloblastina/antagonistas & inibidores , Neutrófilos/enzimologia , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Apoptose/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Terapia de Alvo Molecular , Mieloblastina/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia
5.
Immunopharmacol Immunotoxicol ; 40(5): 368-374, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30183441

RESUMO

OBJECTIVES: Angiogenesis targeting is an attractive approach for cancer treatment. Delta-like ligand 4 (DLL4) plays a pivotal role in neovascular development and its inhibitors have recently entered clinical trials for solid tumors. The aim of this study was to evaluate the possibilities of using anti-DLL4 antibody fragment as an angiogenesis maturation inhibitor. MATERIALS AND METHODS: In this study, a DLL4-specific Nanobody, named 3Nb3, was selected and assessed by western blotting and internalization assays. Functional assessments included MTT, apoptosis, and chicken chorioallantoic membrane (CAM) assays. RESULTS: Based on the results, 3Nb3 specifically binds to DLL4 and internalizes into MKN cell. Furthermore, 3Nb3 significantly inhibited the proliferation of cells and also neovascularization in the CAM. CONCLUSIONS: These data demonstrated the potential of Nanobody for application in targeting DLL4. Our findings may provide a basis for the development of novel therapeutic techniques to inhibit growth and neovascularization of tumors.


Assuntos
Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Proliferação de Células/efeitos dos fármacos , Fragmentos de Imunoglobulinas/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas de Membrana/imunologia , Neovascularização Patológica/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Embrião de Galinha , Membrana Corioalantoide/efeitos dos fármacos , Células HEK293 , Humanos
6.
J Biol Chem ; 291(1): 447-61, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26546677

RESUMO

Exogenous cytokine therapy can induce systemic toxicity, which might be prevented by activating endogenously produced cytokines in local cell niches. Here we developed antibody-based activators of cytokine signaling (AcCS), which recognize cytokines only when they are bound to their cell surface receptors. AcCS were developed for type I interferons (IFNs), which induce cellular activities by binding to cell surface receptors IFNAR1 and IFNAR2. As a potential alternative to exogenous IFN therapy, AcCS were shown to potentiate the biological activities of natural IFNs by ∼100-fold. Biochemical and structural characterization demonstrates that the AcCS stabilize the IFN-IFNAR2 binary complex by recognizing an IFN-induced conformational change in IFNAR2. Using IFN mutants that disrupt IFNAR1 binding, AcCS were able to enhance IFN antiviral potency without activating antiproliferative responses. This suggests AcCS can be used to manipulate cytokine signaling for basic science and possibly for therapeutic applications.


Assuntos
Citocinas/imunologia , Fragmentos de Imunoglobulinas/imunologia , Receptores de Citocinas/imunologia , Transdução de Sinais , Antivirais/química , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fragmentos de Imunoglobulinas/farmacologia , Interferon-alfa/farmacologia , Cinética , Mutação/genética , Fosforilação , Conformação Proteica , Receptor de Interferon alfa e beta/química , Receptor de Interferon alfa e beta/metabolismo , Reprodutibilidade dos Testes , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Pharm Res ; 34(10): 2131-2141, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28681164

RESUMO

PURPOSE: To establish a continuous relationship between the size of various antibody fragments and their systemic clearance (CL) in mice. METHODS: Two different orthogonal approaches have been used to establish the relationship. First approach uses CL values estimated by non-compartmental analysis (NCA) to establish a correlation with protein size. The second approach simultaneously characterizes the PK data for all the proteins using a 2-compartment model to establish a relationship between protein size and pharmacokinetic (PK) parameters. RESULTS: Simple mathematical functions (e.g. sigmoidal, power law) were able to characterize the CL vs. protein size relationship generated using the investigated proteins. The relationship established in mouse was used to predict rat, rabbit, monkey, and human relationships using allometric scaling. The predicted relationships were found to capture the available spares data from each species reasonably well. CONCLUSIONS: The CL vs. protein size relationship is important for establishing a robust quantitative structure-PK relationship (QSPKR) for protein therapeutics. The relationship presented here can help in a priori predicting plasma exposure of therapeutic proteins, and together with our previously established relationship between plasma and tissue concentrations of proteins, it can predict the tissue exposure of non-binding proteins simply based on molecular weight/radius and dose.


Assuntos
Anticorpos Monoclonais/farmacologia , Fragmentos de Imunoglobulinas/farmacologia , Modelos Biológicos , Animais , Anticorpos Monoclonais/química , Haplorrinos , Humanos , Fragmentos de Imunoglobulinas/química , Cinética , Camundongos , Estrutura Molecular , Peso Molecular , Coelhos , Ratos , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade
8.
Nature ; 469(7329): 175-80, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21228869

RESUMO

G protein coupled receptors (GPCRs) exhibit a spectrum of functional behaviours in response to natural and synthetic ligands. Recent crystal structures provide insights into inactive states of several GPCRs. Efforts to obtain an agonist-bound active-state GPCR structure have proven difficult due to the inherent instability of this state in the absence of a G protein. We generated a camelid antibody fragment (nanobody) to the human ß(2) adrenergic receptor (ß(2)AR) that exhibits G protein-like behaviour, and obtained an agonist-bound, active-state crystal structure of the receptor-nanobody complex. Comparison with the inactive ß(2)AR structure reveals subtle changes in the binding pocket; however, these small changes are associated with an 11 Å outward movement of the cytoplasmic end of transmembrane segment 6, and rearrangements of transmembrane segments 5 and 7 that are remarkably similar to those observed in opsin, an active form of rhodopsin. This structure provides insights into the process of agonist binding and activation.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/química , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Fragmentos de Imunoglobulinas/química , Fragmentos de Imunoglobulinas/imunologia , Nanoestruturas/química , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/imunologia , Agonistas de Receptores Adrenérgicos beta 2/metabolismo , Animais , Sítios de Ligação , Camelídeos Americanos , Cristalografia por Raios X , Agonismo Inverso de Drogas , Humanos , Fragmentos de Imunoglobulinas/metabolismo , Fragmentos de Imunoglobulinas/farmacologia , Ligantes , Modelos Moleculares , Movimento/efeitos dos fármacos , Opsinas/agonistas , Opsinas/química , Opsinas/metabolismo , Propanolaminas/química , Propanolaminas/metabolismo , Propanolaminas/farmacologia , Conformação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Proteínas Virais/química , Proteínas Virais/metabolismo
9.
Pharm Res ; 32(7): 2439-49, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25690340

RESUMO

PURPOSE: Many genetic disorders, including chondrodysplasias, and acquired disorders impair growth plate function, resulting in short and sometimes malformed bones. There are multiple endocrine and paracrine factors that promote chondrogenesis at the growth plate, which could potentially be used to treat these disorders. Targeting these growth factors specifically to the growth plate might augment the therapeutic skeletal effect while diminishing undesirable effects on non-target tissues. METHODS: Using yeast display technology, we selected single-chain variable antibody fragments that bound to human and mouse matrilin-3, an extracellular matrix protein specifically expressed in cartilage tissue. The ability of the selected antibody fragments to bind matrilin-3 and to bind cartilage tissue in vitro and in vivo was assessed by ELISA and immunohistochemistry. RESULTS: We identified antibody fragments that bound matrilin-3 with high affinity and also bound with high tissue specificity to cartilage homogenates and to cartilage structures in mouse embryo sections. When injected intravenously in mice, the antibody fragments specifically homed to cartilage. CONCLUSIONS: Yeast display successfully selected antibody fragments that are able to target cartilage tissue in vivo. Coupling these antibodies to chondrogenic endocrine and paracrine signaling molecules has the potential to open up new pharmacological approaches to treat childhood skeletal growth disorders.


Assuntos
Lâmina de Crescimento/efeitos dos fármacos , Fragmentos de Imunoglobulinas/farmacologia , Proteínas Matrilinas/metabolismo , Anticorpos de Cadeia Única/farmacologia , Animais , Especificidade de Anticorpos , Clonagem Molecular , Lâmina de Crescimento/embriologia , Lâmina de Crescimento/metabolismo , Células HEK293 , Humanos , Fragmentos de Imunoglobulinas/administração & dosagem , Fragmentos de Imunoglobulinas/toxicidade , Imuno-Histoquímica , Proteínas Matrilinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteínas Recombinantes/metabolismo , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/toxicidade , Leveduras/genética
10.
Neurobiol Dis ; 56: 74-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23607939

RESUMO

Approximately 10% of amyotrophic lateral sclerosis (ALS) cases are familial (known as FALS) with an autosomal dominant inheritance pattern, and ~25% of FALS cases are caused by mutations in Cu/Zn superoxide dismutase (SOD1). There is convincing evidence that mutant SOD1 (mtSOD1) kills motor neurons (MNs) because of a gain-of-function toxicity, most likely related to aggregation of mtSOD1. A number of recent reports have suggested that antibodies can be used to treat mtSOD1-induced FALS. To follow up on the use of antibodies as potential therapeutics, we generated single chain fragments of variable region antibodies (scFvs) against SOD1, and then expressed them as 'intrabodies' within a motor neuron cell line. In the present study, we describe isolation of human scFvs that interfere with mtSOD1 in vitro aggregation and toxicity. These scFvs may have therapeutic potential in sporadic ALS, as well as FALS, given that sporadic ALS may also involve abnormalities in the SOD1 protein or activity.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Fragmentos de Imunoglobulinas/farmacologia , Superóxido Dismutase/efeitos dos fármacos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Bacteriófago M13/genética , Biotinilação , Western Blotting , Morte Celular/efeitos dos fármacos , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Humanos , Superóxido Dismutase/genética , Superóxido Dismutase-1
11.
Cancer Immunol Immunother ; 62(4): 747-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23242316

RESUMO

Despite the well-established antitumor activity of CD1d-restricted invariant natural killer T lymphocytes (iNKT), their use for cancer therapy has remained challenging. This appears to be due to their strong but short-lived activation followed by long-term anergy after a single administration of the CD1d agonist ligand alpha-galactosylceramide (αGC). As a promising alternative, we obtained sustained mouse iNKT cell responses associated with prolonged antitumor effects through repeated administrations of tumor-targeted recombinant sCD1d-antitumor scFv fusion proteins loaded with αGC. Here, we demonstrate that CD1d fusion proteins bound to tumor cells via the antibody fragment specific for a tumor-associated antigen, efficiently activate human iNKT cell lines leading to potent tumor cell lysis. The importance of CD1d tumor targeting was confirmed in tumor-bearing mice in which only the specific tumor-targeted CD1d fusion protein resulted in tumor inhibition of well-established aggressive tumor grafts. The therapeutic efficacy correlated with the repeated activation of iNKT and natural killer cells marked by their release of TH1 cytokines, despite the up-regulation of the co-inhibitory receptor PD-1. Our results demonstrate the superiority of providing the superagonist αGC loaded on recombinant CD1d proteins and support the use of αGC/sCD1d-antitumor fusion proteins to secure a sustained human and mouse iNKT cell activation, while targeting their cytotoxic activity and cytokine release to the tumor site.


Assuntos
Antígenos CD1d/farmacologia , Fragmentos de Imunoglobulinas/farmacologia , Imunoterapia Adotiva/métodos , Células T Matadoras Naturais/efeitos dos fármacos , Células T Matadoras Naturais/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Antígenos CD1d/imunologia , Linhagem Celular Tumoral , Feminino , Galactosilceramidas/imunologia , Humanos , Fragmentos de Imunoglobulinas/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/imunologia
12.
Arch Biochem Biophys ; 526(2): 146-53, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22387378

RESUMO

Therapeutic monoclonal antibodies (mAbs) are mostly used in cancer, as anti-infectious agents and as immunomodulatory drugs, and are amongst the most active area of research and development in the pharmaceutical industry. This class of drugs comprises unconjugated antibodies or antibody fragments, antibody-drug conjugates, radio-immunoconjugates and bispecific/trispecific molecules. A better understanding of the mechanism of action of successful mAbs is fundamental for the selection of more active and less toxic mAbs of new generation. Furthermore reliable screening of new compounds at an early stage of preclinical development, for both efficacy and toxicity, should allow the selection of the best molecules at an early stage, and improve the rate of success of this class of drugs. Here we review the major methods that are employed for testing the activity of therapeutic mAbs in vitro and in vivo in small animal models and point out to some of the pitfalls in these assays.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Avaliação Pré-Clínica de Medicamentos/métodos , Imunoconjugados/uso terapêutico , Fragmentos de Imunoglobulinas/uso terapêutico , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Morte Celular/efeitos dos fármacos , Humanos , Sistema Imunitário/efeitos dos fármacos , Imunoconjugados/farmacologia , Fragmentos de Imunoglobulinas/farmacologia , Fagocitose/efeitos dos fármacos
13.
Thromb Haemost ; 122(1): 57-66, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33882608

RESUMO

Tissue plasminogen activator (tPA) is used clinically because it has a higher binding specificity for insoluble fibrin (IF) than urokinase (UK), but even pro-tPA has catalytic activity against substrates other than IF. UK has the advantage that it is specifically activated on IF; however, it binds IF weakly. Previously, we established a monoclonal antibody (mAb) that recognizes a pit structure formed only in IF. Here, we developed a new mAb against the pit, 1101, that does not affect coagulation or fibrinolysis, and prepared a fusion protein of UK with humanized 1101 Fab to transport UK selectively to IF. In IF-containing lesions, UK is cleaved by plasmin at two sites, Lys158/Ile159 and Lys135/Lys136. Cleavage of the former leads to activation of UK; however, because activated UK is linked by S-S bonds before and after cleavage, it is not released from the fusion. Cleavage at the latter site causes UK to leave the fusion protein; hence, we mutated Lys135/Lys136 to Gly135/Gly136 to prevent release of UK. This engineered UK-antibody fusion, AMU1114, significantly decreased the reduction of plasma plasminogen levels in vivo relative to UK. In a photochemically induced mouse model of thrombus, the vascular patency rate was 0% (0/10) in the control, 50% (5/10) in the tPA treatment group, and 90% (9/10) in the AMU1114 treatment group. Although no death was observed 1 hour after administration of each thrombolytic agent, some mice died within 24 hours in all treatment groups, including control. These data indicate the need for further basic studies of AMU1114.


Assuntos
Fibrina/efeitos dos fármacos , Fragmentos de Imunoglobulinas/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Animais , Modelos Animais de Doenças , Fibrina/metabolismo , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico , Fragmentos de Imunoglobulinas/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL/sangue , Camundongos Endogâmicos C57BL/metabolismo , Ativador de Plasminogênio Tecidual/farmacologia , Ativador de Plasminogênio Tecidual/uso terapêutico , Ativador de Plasminogênio Tipo Uroquinase/uso terapêutico
14.
CPT Pharmacometrics Syst Pharmacol ; 11(1): 68-81, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34713987

RESUMO

PB2452, a neutralizing monoclonal antibody fragment that binds the antiplatelet drug ticagrelor with high affinity, is being developed as a ticagrelor reversal agent. To identify a clinically useful intravenous (i.v.) reversal regimen, a semimechanistic exposure-response model was developed during the PB2452 first-in-human phase I study. From a randomized, double-blind, placebo-controlled, single-dose trial to evaluate the safety, efficacy, and pharmacokinetics (PKs) of PB2452 in 61 healthy volunteers pretreated with ticagrelor, sequential dose cohort data were used to build and refine an exposure-response model that combined population PK models for ticagrelor (TICA), ticagrelor active metabolite (TAM), and PB2452, and related their binding relationships to the PK of uncomplexed TICA and TAM which is predictive of platelet inhibition. Platelet function was assessed by multiple assays. The model was developed using Bayesian methods in NONMEM. Human PK and pharmacodynamic data from sequential dose cohorts were used to initially define and then refine model parameters. Model simulations indicated that an initial i.v. bolus of PB2452, followed by a high-rate infusion, and then a slower-rate infusion would provide immediate and sustained reversal of the antiplatelet effects of ticagrelor. Based on model predictions, a 6 g i.v. bolus followed by 6 g infused over 4 h and then 6 g over 12 h was identified and tested in study subjects and shown to provide complete reversal within 5 min of infusion onset that was sustained for 20-24 h. The model is predictive of the reversal profile of PB2452 and will inform future trials of PB2452.


Assuntos
Reversão da Anticoagulação/métodos , Anticorpos Amplamente Neutralizantes/administração & dosagem , Fragmentos de Imunoglobulinas/administração & dosagem , Fragmentos de Imunoglobulinas/farmacologia , Ticagrelor/antagonistas & inibidores , Adolescente , Adulto , Teorema de Bayes , Plaquetas/efeitos dos fármacos , Anticorpos Amplamente Neutralizantes/farmacologia , Anticorpos Amplamente Neutralizantes/uso terapêutico , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Fragmentos de Imunoglobulinas/uso terapêutico , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Inibidores da Agregação Plaquetária/farmacologia , Ticagrelor/administração & dosagem , Ticagrelor/farmacocinética , Ticagrelor/farmacologia , Ticagrelor/uso terapêutico , Adulto Jovem
15.
J Clin Invest ; 118(3): 994-1005, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18259610

RESUMO

Invariant NKT (iNKT) cells are potent activators of DCs, NK cells, and T cells, and their antitumor activity has been well demonstrated. A single injection of the high-affinity CD1d ligand alpha-galactosylceramide (alphaGalCer) leads to short-lived iNKT cell activation followed, however, by long-term anergy, limiting its therapeutic use. In contrast, we demonstrated here that when alphaGalCer was loaded on a recombinant soluble CD1d molecule (alphaGalCer/sCD1d), repeated injections led to sustained iNKT and NK cell activation associated with IFN-gamma secretion as well as DC maturation in mice. Most importantly, when alphaGalCer/sCD1d was fused to a HER2-specific scFv antibody fragment, potent inhibition of experimental lung metastasis and established s.c. tumors was obtained when systemic treatment was started 2-7 days after the injection of HER2-expressing B16 melanoma cells. In contrast, administration of free alphaGalCer at this time had no effect. The antitumor activity of the CD1d-anti-HER2 fusion protein was associated with HER2-specific tumor localization and accumulation of iNKT, NK, and T cells at the tumor site. Targeting iNKT cells to the tumor site thus may activate a combined innate and adaptive immune response that may prove to be effective in cancer immunotherapy.


Assuntos
Antígenos CD1/farmacologia , Antineoplásicos/farmacologia , Galactosilceramidas/farmacologia , Fragmentos de Imunoglobulinas/farmacologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Receptor ErbB-2/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Animais , Antígenos CD1d , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Feminino , Interferon gama/biossíntese , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor ErbB-2/imunologia
16.
Clin Exp Immunol ; 165(3): 285-91, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21649648

RESUMO

While antibody-based therapeutics have become firmly established as front-line drugs, the use of antibodies as research tools in small molecule drug discovery is still in its infancy. In this review we focus on the use of antibody fragments as crystallization chaperones to aid the structural determination of otherwise 'uncrystallizable' or 'undruggable' target proteins. We also highlight a potential application for this technology, in which antibody-mediated structures may be used to inform the design of new chemical entities.


Assuntos
Cristalografia/métodos , Fragmentos de Imunoglobulinas/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Descoberta de Drogas/métodos , Fragmentos de Imunoglobulinas/química
17.
J Surg Res ; 170(2): 169-78, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21601881

RESUMO

BACKGROUND: Intact antibodies are poor imaging agents due to a long serum half-life (10-20 d) preventing adequate contrast between the tumor and surrounding blood. Smaller engineered antibody fragments overcome this problem by exhibiting shorter serum half-lives (4-20 h).The diabody (55 kDa) is the smallest antibody fragment, which retains the bivalency of the intact antibody. Our goal was to develop and characterize the anti-CA19-9 diabody fragment and determine its ability to provide antigen specific imaging of pancreas cancer. METHODS: The diabody DNA construct was created by isolation of the variable region genes of the intact anti-CA19-9 antibody. Diabody expression was carried out in NS0 cells and purified using HPLC from supernatant. Specific antigen binding was confirmed with flow cytometry and immunofluorescence. Radiolabeled diabody was injected into mice harboring an antigen positive xenograft (BxPC3 or Capan-2) and a negative xenograft (MiaPaca-2). MicroCT and MicroPET were performed at successive time intervals after injection. Radioactivity was measured in blood and tumor to provide objective confirmation of the microPET images. RESULTS: Immunofluorescence and flow cytometry showed specific binding of the anti-CA19-9 diabody. Pancreas xenograft imaging of BxPC3/MiaPaca-2 and Capan-2/MiaPaca-2 models with the anti-CA19-9 diabody demonstrated an average tumor:blood ratio of 5.0 and 2.0, respectively, and an average positive:negative tumor ratio of 11 and 6, respectively. With respect to the tumor:blood ratio, these data indicate five times and two times more radioactivity in the tumor than in the blood yielding adequate contrast between tumor tissue and background (i.e., blood) to create the representative microPET images. CONCLUSIONS: We successfully engineered a functional diabody against CA19-9, a tumor antigen present on the vast majority of pancreas cancers. Additionally, we demonstrate high contrast antigen specific microPET imaging of pancreas cancer in xenograft models.


Assuntos
Anticorpos Monoclonais , Antígeno CA-19-9/imunologia , Fragmentos de Imunoglobulinas , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Animais , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/farmacocinética , Especificidade de Anticorpos , Antígeno CA-19-9/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Hibridomas , Fragmentos de Imunoglobulinas/sangue , Fragmentos de Imunoglobulinas/farmacologia , Radioisótopos do Iodo/farmacocinética , Camundongos , Mieloma Múltiplo , Neoplasias Pancreáticas/imunologia , Engenharia de Proteínas/métodos , Distribuição Tecidual , Transplante Heterólogo
18.
Appl Microbiol Biotechnol ; 90(2): 553-64, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21305279

RESUMO

Phage-display and competitive panning elution leads to the identification of minimum-sized antigen binders together with conventional antibodies from a mouse cDNA library constructed from HM-1 killer toxin neutralizing monoclonal antibody (nmAb-KT). Antigen-specific altered camelid-like single-domain heavy chain antibody (scFv K2) and a conventional antibody (scFv K1) have been isolated against the idiotypic antigen nmAb-KT. The objectives of the study were to examine (1) their properties as compared to conventional antibodies and also (2) their antifungal activity against different pathogenic and non-pathogenic fungal species. The alternative small antigen-binder, i.e., the single-domain heavy chain antibody, was originated from a conventional mouse scFv phage library through somatic hyper-mutation while selection against antigen. This single-domain antibody fragment was well expressed in bacteria and specifically bound with the idiotypic antigen nmAb-KT and had a high stability and solubility. Experimental data showed that the binding affinity for this single-domain antibody was 272-fold higher (K(d)=1.07×10(-10) M) and antifungal activity was three- to fivefold more efficient (IC(50)=0.46×10(-6) to 1.17×10(-6) M) than that for the conventional antibody (K(d)=2.91×10(-8) M and IC(50)=2.14×10(-6) to 3.78×10(-6) M). The derived single-domain antibody might be an ideal scaffold for anti-idiotypic antibody therapy and the development of smaller peptides or peptide mimetic drugs due to their less complex antigen-binding site. We expect that such single-domain synthetic antibodies will find their way into a number of biotechnological or medical applications.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Antifúngicos/química , Antifúngicos/farmacologia , Fatores Matadores de Levedura/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Anti-Idiotípicos/metabolismo , Anticorpos Antifúngicos/isolamento & purificação , Anticorpos Monoclonais/metabolismo , Antifúngicos/química , Antifúngicos/imunologia , Western Blotting , Candida albicans/efeitos dos fármacos , Cryptococcus neoformans/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/metabolismo , Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/metabolismo , Fragmentos de Imunoglobulinas/farmacologia , Concentração Inibidora 50 , Camundongos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Saccharomyces cerevisiae/efeitos dos fármacos , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo
19.
Drug Deliv ; 28(1): 1443-1454, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34236266

RESUMO

Alpha folate receptor (FRα) is currently under investigation as a target for the treatment of patients with non-small-cell lung cancer (NSCLC), since it is highly expressed in tumor cells but is largely absent in normal tissue. In this study, a novel human variable domain of a heavy-chain (VH) antibody fragment specific to FRα was enriched and selected by phage bio-planning. The positive phage clone (3A102 VH) specifically bound to FRα and also cross-reacted with FRß, as tested by ELISA. Clone 3A102 VH was then successfully expressed as a soluble protein in an E. coli shuffle strain. The obtained soluble 3A102 VH demonstrated a high affinity for FRα with affinity constants (Kaff) values around 7.77 ± 0.25 × 107 M-1, with specific binding against both FRα expressing NSCLC cells and NSCLC patient-derived primary cancer cells, as tested by cell ELISA. In addition, soluble 3A102 VH showed the potential desired property of a targeting molecule by being internalized into FRα-expressing cells, as observed by confocal microscopy. This study inspires the use of phage display to develop human VH antibody (Ab) fragments that might be well suited for drug targeted therapy of NSCLC and other FRα-positive cancer cells.


Assuntos
Bacteriófagos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptor 1 de Folato/antagonistas & inibidores , Fragmentos de Imunoglobulinas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Linhagem Celular Tumoral , Humanos , Fragmentos de Imunoglobulinas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
20.
ACS Chem Biol ; 16(9): 1757-1769, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34406751

RESUMO

Cysteine-rich knob domains found in the ultralong complementarity determining regions of a subset of bovine antibodies are capable of functioning autonomously as 3-6 kDa peptides. While they can be expressed recombinantly in cellular systems, in this paper we show that knob domains are also readily amenable to a chemical synthesis, with a co-crystal structure of a chemically synthesized knob domain in complex with an antigen showing structural equivalence to the biological product. For drug discovery, following the immunization of cattle, knob domain peptides can be synthesized directly from antibody sequence data, combining the power and diversity of the bovine immune repertoire with the ability to rapidly incorporate nonbiological modifications. We demonstrate that, through rational design with non-natural amino acids, a paratope diversity can be massively expanded, in this case improving the efficacy of an allosteric peptide. As a potential route to further improve stability, we also performed head-to-tail cyclizations, exploiting the proximity of the N and C termini to synthesize functional, fully cyclic antibody fragments. Lastly, we highlight the stability of knob domains in plasma and, through pharmacokinetic studies, use palmitoylation as a route to extend the plasma half-life of knob domains in vivo. This study presents an antibody-derived medicinal chemistry platform, with protocols for solid-phase synthesis of knob domains, together with the characterization of their molecular structures, in vitro pharmacology, and pharmacokinetics.


Assuntos
Regiões Determinantes de Complementaridade/química , Fragmentos de Imunoglobulinas/química , Peptídeos Cíclicos/síntese química , Sequência de Aminoácidos , Animais , Bovinos , Fragmentos de Imunoglobulinas/sangue , Fragmentos de Imunoglobulinas/farmacologia , Masculino , Modelos Moleculares , Peptídeos Cíclicos/sangue , Peptídeos Cíclicos/farmacocinética , Ligação Proteica , Domínios Proteicos , Dobramento de Proteína , Ratos Sprague-Dawley , Técnicas de Síntese em Fase Sólida , Espectrometria de Massas em Tandem , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA