Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
1.
Br J Clin Pharmacol ; 88(1): 323-335, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34197653

RESUMO

AIMS: In the UNITI endoscopy sub-study, only 17.4% of patients with Crohn's disease (CD) on ustekinumab achieved endoscopic response and 10.9% of patients achieved endoscopic remission at week (w)44. We aimed to evaluate the impact of alternative ustekinumab dosage regimens on endoscopic outcomes based on population pharmacokinetic-pharmacodynamic (popPK-PD) modelling and simulation analysis. METHODS: Real-world data were obtained from 83 patients with moderate-to-severe CD (95% biological-refractory) enrolled in a prospective cohort study receiving intravenous ustekinumab (~6 mg/kg) followed by every eight-week (q8w) subcutaneous maintenance therapy (90 mg). Three sequential models were developed: a two-compartment popPK model linking ustekinumab dose to ustekinumab exposure, an indirect response popPK-PD model describing the effect of ustekinumab exposure on fecal calprotectin (fCal), and a logistic regression outcome model linking fCal to endoscopic outcomes. RESULTS: Ustekinumab clearance increased with decreasing serum albumin and increasing bodyweight. fCal decreased with increasing ustekinumab exposure. The probability of endoscopic response at w24 increased from 10.0% to 17.9% with fCal at w8 decreasing from 1800 µg/g to 694 µg/g (EC50 ). The probability of endoscopic remission at w24 increased from 2.1% to 10.0% with fCal at w8 decreasing from 1800 µg/g to 214 µg/g (EC50 ). Simulation-based comparison of q8w and q4w maintenance dosing regimens predicted 16.7% and 22.2% endoscopic response rates, respectively. Endoscopic remission rates were estimated to be 4.2% on q8w dosing and 6.7% on q4w dosing. CONCLUSIONS: The developed models can guide clinical trial design and support model-informed dose optimization (stratified or individualized dosing) to improve endoscopic outcomes.


Assuntos
Doença de Crohn , Ustekinumab , Doença de Crohn/tratamento farmacológico , Fezes , Humanos , Complexo Antígeno L1 Leucocitário , Estudos Prospectivos , Resultado do Tratamento , Ustekinumab/uso terapêutico
2.
Clin Exp Dermatol ; 47(7): 1324-1336, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35245966

RESUMO

BACKGROUND: Despite the favourable efficacy profile of secukinumab, clinicians encounter varying clinical responses among patients potentially associated with under- and overdosing. As biologics are expensive, their rational use is crucial and evident. Therapeutic drug monitoring could guide clinicians in making decisions about treatment modifications. AIM: In this multicentre, prospective study, we aimed to develop and validate a secukinumab immunoassay and searched for the therapeutic window in patients with psoriasis. METHODS: We determined secukinumab concentrations at trough in sera from 78 patients with psoriasis at multiple timepoints (Weeks 12, 24, 36, 48 and 52; after Week 52, measurements could be taken at an additional three timepoints) during maintenance phase, using an in-house secukinumab immunoassay consisting of a combination of MA-SEC66A2 as capture antibody and MA-SEC67A9, conjugated to horseradish peroxidase, as detecting antibody. At each hospital visit, disease severity was assessed using the Psoriasis Area and Severity Index (PASI). RESULTS: After quantification, 121 serum samples were included for dose-response analysis. Based on a linear mixed-effects model, secukinumab trough concentrations were found to decrease with increasing body mass index (BMI). Based on receiver operating characteristic (ROC) analysis, we concluded that the minimal effective secukinumab threshold was 39.1 mg/L in steady state, and that this was associated with a 92.7% probability of having an optimal clinical response (PASI ≤ 2 or reduction in PASI of ≥ 90%). CONCLUSIONS: Monitoring and targeting a secukinumab trough concentration of 39.1 mg/L may be a viable treatment option in suboptimal responders. In patients with higher BMI, weight-based dosing may be needed in order to prevent underdosing.


Assuntos
Dermatologia , Doença Enxerto-Hospedeiro , Psoríase , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Método Duplo-Cego , Monitoramento de Medicamentos , Doença Enxerto-Hospedeiro/tratamento farmacológico , Humanos , Estudos Prospectivos , Psoríase/tratamento farmacológico , Índice de Gravidade de Doença , Resultado do Tratamento
3.
Mol Ther ; 28(4): 1068-1077, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32101701

RESUMO

Checkpoint-inhibiting antibodies elicit impressive clinical responses, but still face several issues. The current study evaluated whether DNA-based delivery can broaden the application of checkpoint inhibitors, specifically by pursuing cost-efficient in vivo production, facilitating combination therapies, and exploring administration routes that lower immune-related toxicity risks. We therefore optimized plasmid-encoded anti-CTLA-4 and anti-PD-1 antibodies, and studied their pharmacokinetics and pharmacodynamics when delivered alone and in combination via intramuscular or intratumoral electroporation in mice. Intramuscular electrotransfer of these DNA-based antibodies induced complete regressions in a subcutaneous MC38 tumor model, with plasma concentrations up to 4 and 14 µg/mL for anti-CTLA-4 and anti-PD-1 antibodies, respectively, and antibody detection for at least 6 months. Intratumoral antibody gene electrotransfer gave similar anti-tumor responses as the intramuscular approach. Antibody plasma levels, however, were up to 70-fold lower and substantially more transient, potentially improving biosafety of the expressed checkpoint inhibitors. Intratumoral delivery also generated a systemic anti-tumor response, illustrated by moderate abscopal effects and prolonged protection of cured mice against a tumor rechallenge. In conclusion, intramuscular and intratumoral DNA-based delivery of checkpoint inhibitors both enabled long-term anti-tumor responses despite distinct systemic antibody exposure, highlighting the potential of the tumor as delivery site for DNA-based therapeutics.


Assuntos
Anticorpos Monoclonais/genética , Neoplasias do Colo/terapia , Inibidores de Checkpoint Imunológico/administração & dosagem , Plasmídeos/administração & dosagem , Animais , Antígeno CTLA-4/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Sinergismo Farmacológico , Feminino , Inibidores de Checkpoint Imunológico/farmacologia , Injeções Intralesionais , Injeções Intramusculares , Camundongos , Plasmídeos/genética , Receptor de Morte Celular Programada 1/imunologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Int J Mol Sci ; 22(5)2021 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-33800359

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is the main physiological inhibitor of plasminogen activators (PAs) and is therefore an important inhibitor of the plasminogen/plasmin system. Being the fast-acting inhibitor of tissue-type PA (tPA), PAI-1 primarily attenuates fibrinolysis. Through inhibition of urokinase-type PA (uPA) and interaction with biological ligands such as vitronectin and cell-surface receptors, the function of PAI-1 extends to pericellular proteolysis, tissue remodeling and other processes including cell migration. This review aims at providing a general overview of the properties of PAI-1 and the role it plays in many biological processes and touches upon the possible use of PAI-1 inhibitors as therapeutics.


Assuntos
Doenças Cardiovasculares , Movimento Celular/imunologia , Fibrinólise/imunologia , Proteínas de Neoplasias , Neoplasias , Inibidor 1 de Ativador de Plasminogênio , Proteólise , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Fibrose , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/imunologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
5.
Int J Mol Sci ; 22(7)2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916027

RESUMO

Thrombin activatable fibrinolysis inhibitor (TAFI), a proenzyme, is converted to a potent attenuator of the fibrinolytic system upon activation by thrombin, plasmin, or the thrombin/thrombomodulin complex. Since TAFI forms a molecular link between coagulation and fibrinolysis and plays a potential role in venous and arterial thrombotic diseases, much interest has been tied to the development of molecules that antagonize its function. This review aims at providing a general overview on the biochemical properties of TAFI, its (patho)physiologic function, and various strategies to stimulate the fibrinolytic system by interfering with (activated) TAFI functionality.


Assuntos
Carboxipeptidase B2/metabolismo , Animais , Carboxipeptidase B2/antagonistas & inibidores , Ativação Enzimática , Humanos
6.
Int J Mol Sci ; 22(3)2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33540702

RESUMO

Plasminogen activator inhibitor-1 (PAI-1), a key regulator of the fibrinolytic system, is the main physiological inhibitor of plasminogen activators. By interacting with matrix components, including vitronectin (Vn), PAI-1 plays a regulatory role in tissue remodeling, cell migration, and intracellular signaling. Emerging evidence points to a role for PAI-1 in various pathological conditions, including cardiovascular diseases, cancer, and fibrosis. Targeting PAI-1 is therefore a promising therapeutic strategy in PAI-1-related pathologies. A class of small molecule inhibitors including TM5441 and TM5484, designed to bind the cleft in the central ß-sheet A of PAI-1, showed to be potent PAI-1 inhibitors in vivo. However, their binding site has not yet been confirmed. Here, we report two X-ray crystallographic structures of PAI-1 in complex with TM5484. The structures revealed a binding site at the flexible joint region, which is distinct from the presumed binding site. Based on the structural analysis and biochemical data we propose a mechanism for the observed dose-dependent two-step mechanism of PAI-1 inhibition. By binding to the flexible joint region in PAI-1, TM5484 might restrict the structural flexibility of this region, thereby inducing a substrate form of PAI-1 followed by a conversion to an inert form.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/efeitos dos fármacos , Sítios de Ligação , Domínio Catalítico , Cristalização , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/química , Conformação Proteica , Relação Estrutura-Atividade
7.
Blood ; 132(15): 1593-1603, 2018 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-30026184

RESUMO

Joint bleeds are common in congenital hemophilia but rare in acquired hemophilia A (aHA) for reasons unknown. To identify key mechanisms responsible for joint-specific bleeding in congenital hemophilia, bleeding phenotypes after joint injury and tail transection were compared in aHA wild-type (WT) mice (receiving an anti-factor VIII [FVIII] antibody) and congenital HA (FVIII-/-) mice. Both aHA and FVIII-/- mice bled severely after tail transection, but consistent with clinical findings, joint bleeding was notably milder in aHA compared with FVIII-/- mice. Focus was directed to thrombin-activatable fibrinolysis inhibitor (TAFI) to determine its potentially protective effect on joint bleeding in aHA. Joint bleeding in TAFI-/- mice with anti-FVIII antibody was increased, compared with WT aHA mice, and became indistinguishable from joint bleeding in FVIII-/- mice. Measurements of circulating TAFI zymogen consumption after joint injury indicated severely defective TAFI activation in FVIII-/- mice in vivo, consistent with previous in vitro analyses in FVIII-deficient plasma. In contrast, notable TAFI activation was observed in aHA mice, suggesting that TAFI protected aHA joints against bleeding. Pharmacological inhibitors of fibrinolysis revealed that urokinase-type plasminogen activator (uPA)-induced fibrinolysis drove joint bleeding, whereas tissue-type plasminogen activator-mediated fibrinolysis contributed to tail bleeding. These data identify TAFI as an important modifier of hemophilic joint bleeding in aHA by inhibiting uPA-mediated fibrinolysis. Moreover, our data suggest that bleed protection by TAFI was absent in congenital FVIII-/- mice because of severely defective TAFI activation, underscoring the importance of clot protection in addition to clot formation when considering prohemostatic strategies for hemophilic joint bleeding.


Assuntos
Carboxipeptidase B2/metabolismo , Hemartrose/etiologia , Hemartrose/metabolismo , Hemofilia A/complicações , Animais , Carboxipeptidase B2/genética , Modelos Animais de Doenças , Deleção de Genes , Hemartrose/genética , Hemofilia A/genética , Hemofilia A/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
8.
Nanomedicine ; 24: 102103, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31669421

RESUMO

Matrix metalloproteinase-2 (MMP-2) is an endopeptidase involved in cardiovascular disease and cancer. To date, no highly selective MMP-2 inhibitors have been identified for potential use in humans. Aim of our work was to apply the nanobody technology to the generation of highly selective inhibitors of human MMP-2 and to assess their effects on platelet function and their applicability as conjugated nanobodies. We constructed a nanobody library after immunising an alpaca with human active MMP-2 and identified, after phage display and screening, one MMP-2 inhibitory nanobody (VHH-29), able to hinder the effects of MMP-2 on platelet activation, and one nanobody not inhibiting MMP-2 activity (VHH-136) which, chemically conjugated to a fluorescent probe, allowed the detection of human MMP-2 by flow-cytometry and immune-cytochemistry. In conclusion, we have generated and characterized two new nanotechnological molecular tools for human MMP-2 which represent promising agents for the study of MMP-2 in cardiovascular pathophysiology.


Assuntos
Citometria de Fluxo , Metaloproteinase 2 da Matriz/imunologia , Biblioteca de Peptídeos , Anticorpos de Domínio Único , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/imunologia
9.
Int J Mol Sci ; 21(16)2020 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-32824134

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is the main physiological inhibitor of tissue-type (tPA) and urokinase-type (uPA) plasminogen activators (PAs). Apart from being critically involved in fibrinolysis and wound healing, emerging evidence indicates that PAI-1 plays an important role in many diseases, including cardiovascular disease, tissue fibrosis, and cancer. Targeting PAI-1 is therefore a promising therapeutic strategy in PAI-1 related pathologies. Despite ongoing efforts no PAI-1 inhibitors were approved to date for therapeutic use in humans. A better understanding of the molecular mechanisms of PAI-1 inhibition is therefore necessary to guide the rational design of PAI-1 modulators. Here, we present a 1.9 Å crystal structure of PAI-1 in complex with an inhibitory nanobody VHH-s-a93 (Nb93). Structural analysis in combination with biochemical characterization reveals that Nb93 directly interferes with PAI-1/PA complex formation and stabilizes the active conformation of the PAI-1 molecule.


Assuntos
Simulação de Acoplamento Molecular , Inibidor 1 de Ativador de Plasminogênio/química , Anticorpos de Domínio Único/química , Sítios de Ligação , Humanos , Inibidor 1 de Ativador de Plasminogênio/imunologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Estabilidade Proteica , Anticorpos de Domínio Único/imunologia
10.
Br J Cancer ; 121(3): 199-210, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31257362

RESUMO

The monoclonal antibody trastuzumab (Herceptin®), which targets the human epidermal growth factor receptor 2 (HER2), is approved for the treatment of early breast and advanced breast and gastric cancer in which HER2 is overexpressed. Several biosimilar versions of trastuzumab are expected to enter the European market over the course of 2018 and 2019. The biosimilar development pathway consists of a comprehensive comparability exercise between the biosimilar candidate and the reference product, primarily focussing on data from analytical studies. Clinical studies for biosimilar candidates follow a different design to those for a new biological, as the aim is not to independently establish clinical benefit, but to confirm biosimilarity between the two agents. The different trastuzumab biosimilar candidates have followed diverse pathways in their clinical development, with differences in clinical trial design (equivalence or non-inferiority design), patient population (those with metastatic or early breast cancer) and endpoint (overall response rate or pathological complete response). These differences in approach in phase 3 testing must be viewed in the totality of evidence demonstrating biosimilarity. Adequate information on the biosimilar approval pathway, the nature of the biosimilarity exercise and how the clinical development of a biosimilar is tailored to meet the licensing requirements can help informed decision making in clinical practice.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Medicamentos Biossimilares/uso terapêutico , Neoplasias/tratamento farmacológico , Trastuzumab/uso terapêutico , Anticorpos Monoclonais/farmacocinética , Medicamentos Biossimilares/farmacocinética , Ensaios Clínicos como Assunto , Desenvolvimento de Medicamentos , Humanos , Receptor ErbB-2/antagonistas & inibidores
11.
Future Oncol ; 15(28): 3255-3265, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31441323

RESUMO

Biosimilars are biologic products that are highly similar to, and have no clinically meaningful differences from, the approved originator molecule. They are poised to play an increasingly central role in cancer treatment, helping to improve access by driving down costs. Regulatory bodies have set out robust mechanisms for the approval of biosimilars, based on comprehensive and rigorous analytical and nonclinical comparisons with the originator. Product attributes (e.g., post-translational modifications) that are important to the function of the molecule must be similar between biosimilar and originator. This should be followed by a robust clinical development program, assessing pharmacokinetics, pharmacodynamics, efficacy, safety and immunogenicity. Equivalence in one indication might allow extrapolation across all the indications of the originator biologic. The recent approval of several trastuzumab biosimilars provides an example of how this process can work in practice for the benefit of patients, clinicians and payers.


Assuntos
Medicamentos Biossimilares/uso terapêutico , Aprovação de Drogas , Neoplasias/tratamento farmacológico , Farmacovigilância , Medição de Risco/métodos , Humanos , Neoplasias/patologia , Prognóstico
12.
Future Oncol ; 15(7): 777-790, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30500264

RESUMO

Biologics are an integral component in the treatment of various diseases. However, limited patient access to these medicines remains a significant global challenge, prompting development of safe and effective biosimilars. A biosimilar is 'highly similar to a reference (originator) product, for which there are no clinically meaningful differences between the two products in terms of safety, purity and potency'. Biosimilars have the potential to offer possible benefits, including lower treatment costs, thereby increasing patient access and clinical use, which may lead to better overall outcomes. Improved understanding of biosimilars may enhance confidence and trust in these agents. As increasing numbers of biosimilars achieve regulatory approval, this overview aims to address enduring knowledge gaps regarding the development and use of biosimilars.


Assuntos
Medicamentos Biossimilares/normas , Medicamentos Biossimilares/uso terapêutico , Desenvolvimento de Medicamentos/legislação & jurisprudência , Desenvolvimento de Medicamentos/normas , Medicamentos Biossimilares/efeitos adversos , Ensaios Clínicos como Assunto , Aprovação de Drogas/legislação & jurisprudência , Substituição de Medicamentos , Regulamentação Governamental , Guias como Assunto , Humanos , Equivalência Terapêutica
13.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L54-L68, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860148

RESUMO

Elevated active plasminogen activator inhibitor-1 (PAI-1) has an adverse effect on the outcomes of intrapleural fibrinolytic therapy (IPFT) in tetracycline-induced pleural injury in rabbits. To enhance IPFT with prourokinase (scuPA), two mechanistically distinct approaches to targeting PAI-1 were tested: slowing its reaction with urokinase (uPA) and monoclonal antibody (mAb)-mediated PAI-1 inactivation. Removing positively charged residues at the "PAI-1 docking site" (179RHRGGS184→179AAAAAA184) of uPA results in a 60-fold decrease in the rate of inhibition by PAI-1. Mutant prourokinase (0.0625-0.5 mg/kg; n = 12) showed efficacy comparable to wild-type scuPA and did not change IPFT outcomes ( P > 0.05). Notably, the rate of PAI-1-independent intrapleural inactivation of mutant uPA was 2 times higher ( P < 0.05) than that of the wild-type enzyme. Trapping PAI-1 in a "molecular sandwich"-type complex with catalytically inactive two-chain urokinase with Ser195Ala substitution (S195A-tcuPA; 0.1 and 0.5 mg/kg) did not improve the efficacy of IPFT with scuPA (0.0625-0.5 mg/kg; n = 11). IPFT failed in the presence of MA-56A7C10 (0.5 mg/kg; n = 2), which forms a stable intrapleural molecular sandwich complex, allowing active PAI-1 to accumulate by blocking its transition to a latent form. In contrast, inactivation of PAI-1 by accelerating the active-to-latent transition mediated by mAb MA-33B8 (0.5 mg/kg; n = 2) improved the efficacy of IPFT with scuPA (0.25 mg/kg). Thus, under conditions of slow (4-8 h) fibrinolysis in tetracycline-induced pleural injury in rabbits, only the inactivation of PAI-1, but not a decrease in the rate of its reaction with uPA, enhances IPFT. Therefore the rate of fibrinolysis, which varies in different pathologic states, could affect the selection of PAI-1 inhibitors to enhance fibrinolytic therapy.


Assuntos
Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , Inibidor 1 de Ativador de Plasminogênio/química , Doenças Pleurais/tratamento farmacológico , Tetraciclina/toxicidade , Terapia Trombolítica/métodos , Animais , Modelos Animais de Doenças , Feminino , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Doenças Pleurais/induzido quimicamente , Inibidores da Síntese de Proteínas/toxicidade , Coelhos
15.
J Biol Chem ; 291(29): 15156-68, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226628

RESUMO

A peptide segment that binds the active site of a serine protease in a substrate-like manner may behave like an inhibitor or a substrate. However, there is sparse information on which factors determine the behavior a particular peptide segment will exhibit. Here, we describe the first x-ray crystal structure of a nanobody in complex with a serine protease. The nanobody displays a new type of interaction between an antibody and a serine protease as it inserts its complementary determining region-H3 loop into the active site of the protease in a substrate-like manner. The unique binding mechanism causes the nanobody to behave as a strong inhibitor as well as a poor substrate. Intriguingly, its substrate behavior is incomplete, as 30-40% of the nanobody remained intact and inhibitory after prolonged incubation with the protease. Biochemical analysis reveals that an intra-loop interaction network within the complementary determining region-H3 of the nanobody balances its inhibitor versus substrate behavior. Collectively, our results unveil molecular factors, which may be a general mechanism to determine the substrate versus inhibitor behavior of other protease inhibitors.


Assuntos
Camelídeos Americanos/imunologia , Serina Proteases/imunologia , Anticorpos de Domínio Único/química , Sequência de Aminoácidos , Animais , Domínio Catalítico , Regiões Determinantes de Complementaridade , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Conformação Proteica , Serina Proteases/química , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/metabolismo , Anticorpos de Domínio Único/metabolismo , Especificidade por Substrato , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/imunologia
16.
J Transl Med ; 15(1): 131, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28592330

RESUMO

Recombinant monoclonal antibodies (mAbs) are one of today's most successful therapeutic classes in inflammatory diseases and oncology. A wider accessibility and implementation, however, is hampered by the high product cost and prolonged need for frequent administration. The surge in more effective mAb combination therapies further adds to the costs and risk of toxicity. To address these issues, antibody gene transfer seeks to administer to patients the mAb-encoding nucleotide sequence, rather than the mAb protein. This allows the body to produce its own medicine in a cost- and labor-effective manner, for a prolonged period of time. Expressed mAbs can be secreted systemically or locally, depending on the production site. The current review outlines the state of play and clinical prospects of antibody gene transfer, thereby highlighting recent innovations, opportunities and remaining hurdles. Different expression platforms and a multitude of administration sites have been pursued. Viral vector-mediated mAb expression thereby made the most significant strides. Therapeutic proof of concept has been demonstrated in mice and non-human primates, and intramuscular vectored mAb therapy is under clinical evaluation. However, viral vectors face limitations, particularly in terms of immunogenicity. In recent years, naked DNA has gained ground as an alternative. Attained serum mAb titers in mice, however, remain far below those obtained with viral vectors, and robust pharmacokinetic data in larger animals is limited. The broad translatability of DNA-based antibody therapy remains uncertain, despite ongoing evaluation in patients. RNA presents another emerging platform for antibody gene transfer. Early reports in mice show that mRNA may be able to rival with viral vectors in terms of generated serum mAb titers, although expression appears more short-lived. Overall, substantial progress has been made in the clinical translation of antibody gene transfer. While challenges persist, clinical prospects are amplified by ongoing innovations and the versatility of antibody gene transfer. Clinical introduction can be expedited by selecting the platform approach currently best suited for the mAb or disease of interest. Innovations in expression platform, administration and antibody technology are expected to further improve overall safety and efficacy, and unlock the vast clinical potential of antibody gene transfer.


Assuntos
Anticorpos/uso terapêutico , Técnicas de Transferência de Genes , Animais , DNA/metabolismo , Vetores Genéticos , Humanos , RNA/metabolismo , Proteínas Recombinantes/uso terapêutico
17.
Rheumatology (Oxford) ; 56(suppl_4): iv4-iv13, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28903545

RESUMO

Biosimilars are products that contain a similar version of the active substance of an already authorized original biologic medicinal product (reference medicinal product). Their development requires special consideration, as similarity to the reference agent needs to be established through a comprehensive comparability exercise. Given the complex nature of these agents, minor structural differences may emerge, but the process of biosimilarity determination is designed to ascertain that the nature and impact of these differences are not clinically significant. Determination of biosimilarity should follow quality-by-design principles, which provide a deep understanding of the product development process, guided by pre-defined objectives, process control and risk management. Compared with novel biologic development, biosimilar development places greater emphasis on establishing preclinical quality characteristics. Determination of comparability of quality characteristics includes assessment of physicochemical properties, biological activity, immunochemical properties, purity, impurity and quantity, with appropriate in vivo pharmacology studies being conducted thereafter. Head-to-head comparisons are then conducted to determine pharmacokinetic and pharmacodynamic characteristics, and efficacy, safety and tolerability in phase I and phase III clinical studies. Post-approval risk management requirements include implementation of pharmacovigilance systems and risk management through, for example, the conduct of pharmacoepidemiological studies. There are several biosimilars used in the field of rheumatology that are available in the European Union, or in development, that offer the potential to increase affordability/accessibility of biological treatment. The role of these agents in rheumatology will be determined by the confidence placed in them by rheumatologists. These prescribers should expect high-quality data evaluated by an extensive assessment process.


Assuntos
Medicamentos Biossimilares/farmacologia , Aprovação de Drogas , Desenho de Fármacos , Doenças Reumáticas/tratamento farmacológico , Produtos Biológicos/uso terapêutico , Medicamentos Biossimilares/uso terapêutico , Feminino , Previsões , Humanos , Masculino , Farmacovigilância , Doenças Reumáticas/diagnóstico , Reumatologia/métodos
18.
Blood ; 125(8): 1325-32, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25540192

RESUMO

Circulating thrombin-activatable fibrinolysis inhibitor (TAFI) and plasminogen activator inhibitor-1 (PAI-1) are causal factors for thrombolytic failure. Therefore, we evaluated an antibody-engineered bispecific inhibitor against TAFI and PAI-1 (heterodimer diabody, Db-TCK26D6x33H1F7) in several mouse models of thrombosis and stroke. Prophylactic administration of the diabody (0.8 mg/kg) in a thromboplastin-induced model of thromboembolism led to decreased lung fibrin deposition. In a model of cerebral ischemia and reperfusion, diabody administration (0.8 mg/kg, 1 hour postocclusion) led to a mitigated cerebral injury with a 2.3-fold reduced lesion and improved functional outcomes. In a mouse model of thrombin-induced middle cerebral artery occlusion, the efficacy of the diabody was compared to the standard thrombolytic treatment with recombinant tissue-type plasminogen activator (tPA). Early administration of diabody (0.8 mg/kg) caused a twofold decrease in brain lesion size, whereas that of tPA (10 mg/kg) had a much smaller effect. Delayed administration of diabody or tPA had no effect on lesion size, whereas the combined administration of diabody with tPA caused a 1.7-fold decrease in lesion size. In contrast to tPA, the diabody did not increase accumulative bleeding. In conclusion, administration of a bispecific inhibitor against TAFI and PAI-1 results in a prominent profibrinolytic effect in mice without increased bleeding.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Fibrinolíticos/uso terapêutico , Histona Acetiltransferases/imunologia , Serpina E2/imunologia , Acidente Vascular Cerebral/terapia , Fatores Associados à Proteína de Ligação a TATA/imunologia , Terapias em Estudo/métodos , Fator de Transcrição TFIID/imunologia , Tromboembolia Venosa/terapia , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/metabolismo , Modelos Animais de Doenças , Feminino , Imunoterapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Multimerização Proteica , Acidente Vascular Cerebral/patologia , Tromboembolia Venosa/patologia
19.
Ther Drug Monit ; 39(4): 308-315, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28230618

RESUMO

Biopharmaceuticals are primarily therapeutic proteins developed to perform specific functions by acting on the disease pathophysiology. Compared with low-molecular chemically synthesized drugs, production of biopharmaceuticals is much more complex and routes of administration and pharmacokinetics differ. Biopharmaceuticals are blockbusters in the treatment of inflammatory diseases, such as psoriasis, multiple sclerosis, rheumatic diseases, and inflammatory bowel diseases, and the introduction of these drugs has revolutionized treatment. Disadvantages include their high costs and the fact that they can evoke antidrug antibodies leading to decreased efficacy. Treatment can be optimized through the development of dosing algorithms and cost can be reduced by biosimilars, after a comparable biological activity, safety, and efficacy have been demonstrated.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Medicamentos Biossimilares/uso terapêutico , Animais , Anti-Inflamatórios/economia , Anticorpos Monoclonais/economia , Produtos Biológicos/economia , Produtos Biológicos/uso terapêutico , Biofarmácia , Medicamentos Biossimilares/economia , Ensaios Clínicos como Assunto/economia , Ensaios Clínicos como Assunto/métodos , Humanos , Doenças Inflamatórias Intestinais/diagnóstico , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/economia , Psoríase/diagnóstico , Psoríase/tratamento farmacológico , Psoríase/economia , Doenças Reumáticas/diagnóstico , Doenças Reumáticas/tratamento farmacológico , Doenças Reumáticas/economia , Equivalência Terapêutica , Resultado do Tratamento
20.
Stroke ; 47(9): 2419-22, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27470988

RESUMO

BACKGROUND AND PURPOSE: Cerebral ischemia and reperfusion is associated with activation of the coagulation cascade and fibrin deposition in cerebral microvessels. Both thrombin-activatable fibrinolysis inhibitor (TAFI) and plasminogen activator inhibitor-1 (PAI-1) attenuate fibrinolysis and are therefore attractive targets for the treatment of ischemic stroke. METHODS: TAFI and PAI-1 were inhibited by monoclonal antibodies in a mouse model of transient middle cerebral artery occlusion. Twenty-four hours after stroke, mice were neurologically scored, cerebral thrombotic burden was assessed, and brain infarct sizes were calculated. RESULTS: Inhibition of TAFI or PAI-1 significantly decreased cerebral infarct sizes by 50% 24 hours after stroke. This reduction in cerebral damage was associated with a significant decrease in fibrin(ogen) deposition in the ischemic brain. Concurrently, functional recovery of the animals was improved. Interestingly, combined targeting of TAFI and PAI-1 using low, and by themselves inactive, doses of antibodies improved cerebral blood flow and reduced cerebral fibrin(ogen) deposition and infarct sizes by 50%. When dual treatment was delayed to 1 hour after the start of reperfusion, it still reduced brain injury; however, this was not statistically significant. CONCLUSIONS: Targeting of PAI-1 and TAFI is protective in an ischemic stroke model by attenuating fibrin(ogen) deposition, thereby improving reperfusion. Combined inhibition has a co-operative effect that could become useful in ischemic stroke therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Carboxipeptidase B2/imunologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacologia , Modelos Animais de Doenças , Camundongos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa