Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Toxicon ; 232: 107206, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37356552

RESUMO

In recent years it has become possible to design snakebite antivenoms with diverse pharmacokinetic properties. Owing to the pharmacokinetic variability of venoms, the choice of antivenom scaffold may influence a treatment's neutralisation coverage. Computation offers a useful medium through which to assess the pharmacokinetics and pharmacodynamics of envenomation-treatment systems, as antivenoms with identical neutralising capacities can be simulated. In this study, we simulate envenomation and treatment with a variety of antivenoms, to define the properties of effective antivenoms. Systemic envenomation and treatment were described using a two-compartment pharmacokinetic model. Treatment of Naja sumatrana and Cryptelytrops purpureomaculatus envenomation was simulated with a set of 200,000 theoretical antivenoms across 10 treatment time delays. These two venoms are well-characterised and have differing pharmacokinetic properties. The theoretical antivenom set varied across molecular weight, dose, kon, koff, and valency. The best and worst treatments were identified using an area under the curve metric, and a global sensitivity analysis was performed to quantify the influence of the input parameters on treatment outcome. The simulations show that scaffolds of diverse molecular formats can be effective. Molecular weight and valency have a negligible direct impact on treatment outcome, however low molecular weight scaffolds offer more flexibility across the other design parameters, particularly when treatment is delayed. The simulations show kon to primarily mediate treatment efficacy, with rates above 105 M-1s-1 required for the most effective treatments. koff has the greatest impact on the performance of less effective scaffolds. While the same scaffold preferences for improved treatment are seen for both model snakes, the parameter bounds for C. purpureomaculatus envenomation are more constrained. This paper establishes a computational framework for the optimisation of antivenom design.


Assuntos
Antivenenos , Mordeduras de Serpentes , Animais , Antivenenos/farmacologia , Antivenenos/uso terapêutico , Mordeduras de Serpentes/tratamento farmacológico , Serpentes , Farmacocinética , Venenos de Serpentes/farmacocinética
2.
Sci Rep ; 11(1): 11663, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083615

RESUMO

The interaction of platelet GPIbα with von Willebrand factor (VWF) is essential to initiate platelet adhesion and thrombosis, particularly under high shear stress conditions. However, no drug targeting GPIbα has been developed for clinical practice. Here we characterized anfibatide, a GPIbα antagonist purified from snake (Deinagkistrodon acutus) venom, and evaluated its interaction with GPIbα by surface plasmon resonance and in silico modeling. We demonstrated that anfibatide interferds with both VWF and thrombin binding, inhibited ristocetin/botrocetin- and low-dose thrombin-induced human platelet aggregation, and decreased thrombus volume and stability in blood flowing over collagen. In a single-center, randomized, and open-label phase I clinical trial, anfibatide was administered intravenously to 94 healthy volunteers either as a single dose bolus, or a bolus followed by a constant rate infusion of anfibatide for 24 h. Anfibatide inhibited VWF-mediated platelet aggregation without significantly altering bleeding time or coagulation. The inhibitory effects disappeared within 8 h after drug withdrawal. No thrombocytopenia or anti-anfibatide antibodies were detected, and no serious adverse events or allergic reactions were observed during the studies. Therefore, anfibatide was well-tolerated among healthy subjects. Interestingly, anfibatide exhibited pharmacologic effects in vivo at concentrations thousand-fold lower than in vitro, a phenomenon which deserves further investigation.Trial registration: Clinicaltrials.gov NCT01588132.


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Venenos de Crotalídeos/uso terapêutico , Fibrinolíticos/uso terapêutico , Lectinas Tipo C/uso terapêutico , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Venenos de Serpentes/uso terapêutico , Animais , Coagulação Sanguínea/efeitos dos fármacos , Venenos de Crotalídeos/química , Venenos de Crotalídeos/isolamento & purificação , Venenos de Crotalídeos/farmacocinética , Crotalinae , Fibrinolíticos/química , Fibrinolíticos/isolamento & purificação , Fibrinolíticos/farmacocinética , Voluntários Saudáveis , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/isolamento & purificação , Modelos Moleculares , Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas , Complexo Glicoproteico GPIb-IX de Plaquetas/química , Ligação Proteica , Conformação Proteica , Ristocetina/farmacologia , Venenos de Serpentes/química , Venenos de Serpentes/isolamento & purificação , Venenos de Serpentes/farmacocinética , Relação Estrutura-Atividade , Trombina/farmacologia , Trombose/prevenção & controle , Fator de von Willebrand/química , Fator de von Willebrand/metabolismo
3.
Toxins (Basel) ; 12(3)2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32188075

RESUMO

Snake venom is comprised of a combination of different proteins and peptides with a wide range of molecular weights and different disposition processes inherent to each compound. This causes venom to have a complex exposure profile. Our study investigates 1) how each molecular weight fraction (toxin) of venom contributes to the overall time course of the snake venom, and 2) the ability to determine toxin profiles based on the profile of the overall venom only. We undertook an in silico simulation and modelling study. Sixteen variations of venom, comprising of two to nine toxins with different molecular weights were investigated. The pharmacokinetic parameters (i.e., clearance,, and volume of distribution,) of each toxin were generated based on a log-linear relationship with molecular weight. The concentration-time data of each toxin were simulated for 100 virtual patients using MATLAB and the total concentration-time data of each toxin were modelled using NONMEM. We found that the data of sixteen mixtures were best described by either two- or three-compartment models, despite the venom being made up of more than three different toxins. This suggests that it is generally not possible to determine individual toxin profiles based on measurements of total venom concentrations only.


Assuntos
Modelos Biológicos , Mordeduras de Serpentes/metabolismo , Venenos de Serpentes , Animais , Simulação por Computador , Conjuntos de Dados como Assunto , Relação Dose-Resposta a Droga , Meia-Vida , Humanos , Peso Molecular , Venenos de Serpentes/química , Venenos de Serpentes/farmacocinética , Distribuição Tecidual
4.
J Theor Biol ; 455: 212-221, 2018 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-30036530

RESUMO

Cooperation between VEGFR2 and integrin αVß3 is critical for neovascularization in wound healing, cardiovascular ischemic diseases, ocular diseases, and tumor angiogenesis. In the present study, we developed a rule-based computational model to investigate the potential mechanism by which the Src-induced integrin association with VEGFR2 enhances VEGFR2 activation. Simulations demonstrated that the main function of integrin is to reduce the degradation of VEGFR2 and hence stabilize the activation signal. In addition, receptor synthesis rate and recruitment from internal compartment were found to be sensitive determinants of the activation state of VEGFR2. The model was then applied to simulate the effect of integrin-binding peptides such as tumstatin and cilengitide on VEGFR2 signaling. Further, computational modeling proposed potential molecular mechanisms for the angiogenesis-modulating activity of other integrin-binding peptides. The model highlights the complexity of the crosstalk between αVß3 integrin and VEGFR2 and the necessity of utilizing models to elucidate potential mechanisms in angiogenesis-modulating peptide therapy.


Assuntos
Autoantígenos , Colágeno Tipo IV , Células Endoteliais/metabolismo , Integrina alfaVbeta3/metabolismo , Modelos Biológicos , Neovascularização Patológica , Transdução de Sinais/efeitos dos fármacos , Venenos de Serpentes , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Autoantígenos/farmacologia , Colágeno Tipo IV/farmacocinética , Colágeno Tipo IV/farmacologia , Humanos , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Venenos de Serpentes/farmacocinética , Venenos de Serpentes/farmacologia
5.
Toxins (Basel) ; 10(2)2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29414889

RESUMO

Understanding snake venom pharmacokinetics is essential for developing risk assessment strategies and determining the optimal dose and timing of antivenom required to bind all venom in snakebite patients. This review aims to explore the current knowledge of snake venom pharmacokinetics in animals and humans. Literature searches were conducted using EMBASE (1974-present) and Medline (1946-present). For animals, 12 out of 520 initially identified studies met the inclusion criteria. In general, the disposition of snake venom was described by a two-compartment model consisting of a rapid distribution phase and a slow elimination phase, with half-lives of 5 to 48 min and 0.8 to 28 h, respectively, following rapid intravenous injection of the venoms or toxins. When the venoms or toxins were administered intramuscularly or subcutaneously, an initial absorption phase and slow elimination phase were observed. The bioavailability of venoms or toxins ranged from 4 to 81.5% following intramuscular administration and 60% following subcutaneous administration. The volume of distribution and the clearance varied between snake species. For humans, 24 out of 666 initially identified publications contained sufficient information and timed venom concentrations in the absence of antivenom therapy for data extraction. The data were extracted and modelled in NONMEM. A one-compartment model provided the best fit, with an elimination half-life of 9.71 ± 1.29 h. It is intended that the quantitative information provided in this review will provide a useful basis for future studies that address the pharmacokinetics of snakebite in humans.


Assuntos
Venenos de Serpentes/farmacocinética , Animais , Humanos
6.
Toxins (Basel) ; 9(5)2017 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-28505100

RESUMO

Animal-derived antivenoms constitute the mainstay in the therapy of snakebite envenoming. The efficacy of antivenoms to neutralize toxicity of medically-relevant snake venoms has to be demonstrated through meticulous preclinical testing before their introduction into the clinical setting. The gold standard in the preclinical assessment and quality control of antivenoms is the neutralization of venom-induced lethality. In addition, depending on the pathophysiological profile of snake venoms, the neutralization of other toxic activities has to be evaluated, such as hemorrhagic, myotoxic, edema-forming, dermonecrotic, in vitro coagulant, and defibrinogenating effects. There is a need to develop laboratory assays to evaluate neutralization of other relevant venom activities. The concept of the 3Rs (Replacement, Reduction, and Refinement) in Toxinology is of utmost importance, and some advances have been performed in their implementation. A significant leap forward in the study of the immunological reactivity of antivenoms against venoms has been the development of "antivenomics", which brings the analytical power of mass spectrometry to the evaluation of antivenoms. International partnerships are required to assess the preclinical efficacy of antivenoms against snake venoms in different regions of the world in order to have a detailed knowledge on the neutralizing profile of these immunotherapeutics.


Assuntos
Antivenenos/uso terapêutico , Mordeduras de Serpentes/tratamento farmacológico , Venenos de Serpentes/toxicidade , Animais , Antivenenos/farmacologia , Avaliação Pré-Clínica de Medicamentos , Órgãos Governamentais , Humanos , Testes de Neutralização , Proteômica , Proteínas de Répteis/análise , Mordeduras de Serpentes/metabolismo , Venenos de Serpentes/química , Venenos de Serpentes/farmacocinética , Resultado do Tratamento
7.
Sci Rep ; 6: 26778, 2016 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-27231196

RESUMO

In vitro antivenom efficacy studies were compared to rodent lethality studies to test two Indian snake antivenoms (VINS and BHARAT) against four Sri Lankan snakes. In vitro efficacy was tested at venom concentrations consistent with human envenoming. Efficacy was compared statistically for one batch from each manufacturer where multiple vials were available. In binding studies EC50 for all VINS antivenoms were less than BHARAT for D. russelii [553 µg/mL vs. 1371 µg/mL;p = 0.016), but were greater for VINS antivenoms compared to BHARAT for N. naja [336 µg/mL vs. 70 µg/mL;p < 0.0001]. EC50 of both antivenoms was only slighty different for E. carinatus and B. caeruleus. For procoagulant activity neutralisation, the EC50 was lower for VINS compared to BHARAT - 60 µg/mL vs. 176 µg/mL (p < 0.0001) for Russell's viper and 357 µg/mL vs. 6906µg/mL (p < 0.0001) for Saw-scaled viper. Only VINS antivenom neutralized in vitro neurotoxicity of krait venom. Both antivenoms partially neutralized cobra and didn't neutralize Russell's viper neurotoxicity. Lethality studies found no statistically significant difference in ED50 values between VINS and BHARAT antivenoms. VINS antivenoms appeared superior to BHARAT at concentrations equivalent to administering 10 vials antivenom, based on binding and neutralisation studies. Lethality studies were inconsistent suggesting rodent death may not measure relevant efficacy outcomes in humans.


Assuntos
Antivenenos/farmacologia , Venenos de Serpentes/farmacocinética , Venenos de Serpentes/toxicidade , Animais , Galinhas , Coagulantes/farmacologia , Técnicas In Vitro , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/inervação , Serpentes , Sri Lanka
8.
Clin Cancer Res ; 21(22): 5092-9, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26199386

RESUMO

PURPOSE: Fit-for-purpose pharmacodynamic biomarkers could expedite development of combination antiangiogenic regimens. Plasma sVEGFR2 concentrations ([sVEGFR2]) mark sunitinib effects on the systemic vasculature. We hypothesized that cilengitide would impair microvasculature recovery during sunitinib withdrawal and could be detected through changes in [sVEGFR2]. EXPERIMENTAL DESIGN: Advanced solid tumor patients received 50 mg sunitinib daily for 14 days. For the next 14 days, patients were randomized to arm A (cilengitide 2,000 mg administered intravenously twice weekly) or arm B (no treatment). The primary endpoint was change in [sVEGFR2] between days 14 and 28. A candidate pharmacodynamic biomarker of cilengitide inhibition of integrin αvß3, serum c-telopeptide collagen crosslinks (CTx), was also measured. RESULTS: Of 21 patients, 14 (7 per arm) received all treatments without interruption and had all blood samples available for analysis. The mean change and SD of [sVEGFR2] for all sunitinib-treated patients was consistent with previous data. There was no significant difference in the mean change in [sVEGFR2] from days 14 to 28 between the arms [arm A: 2.8 ng/mL; 95% confidence interval (CI), 2.1-3.6 vs. arm B: 2.0 ng/mL; 95% CI, 0.72-3.4; P = 0.22, 2-sample t test]. Additional analyses suggested (i) prior bevacizumab therapy to be associated with unusually low baseline [sVEGFR2] and (ii) sunitinib causes measurable changes in CTx. CONCLUSIONS: Cilengitide had no measurable effects on any circulating biomarkers. Sunitinib caused measurable declines in serum CTx. The properties of [sVEGFR2] and CTx observed in this study inform the design of future combination antiangiogenic therapy trials.


Assuntos
Biomarcadores Tumorais/sangue , Colágeno Tipo I/sangue , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Peptídeos/sangue , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacocinética , Biomarcadores Tumorais/farmacocinética , Colágeno Tipo I/farmacocinética , Feminino , Humanos , Indóis/administração & dosagem , Indóis/farmacocinética , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias/sangue , Neoplasias/genética , Neoplasias/patologia , Neovascularização Patológica/sangue , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Peptídeos/farmacocinética , Pirróis/administração & dosagem , Pirróis/farmacocinética , Venenos de Serpentes/administração & dosagem , Venenos de Serpentes/farmacocinética , Sunitinibe , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
9.
J Clin Pharmacol ; 55(7): 815-24, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25683324

RESUMO

Cilengitide (EMD 121974, manufactured by Merck KGaA, Darmstadt, Germany) is an αv-integrin receptor antagonist showing high affinity for αvß3 and αvß5.This study determined the mass balance of cilengitide in healthy volunteers receiving a single intravenous infusion of 2.1 MBq (14) C-cilengitide spiked into 250 mL of 2000 mg of cilengitide. Blood, urine, and feces were collected up to day 15 or until excretion of radioactivity was below 1% of the administered dose. Total radioactivity derived from the administration of (14) C-cilengitide and unlabeled cilengitide levels were determined and used for calculation of pharmacokinetic parameters.(14) C-cilengitide-related radioactivity was completely recovered (94.5%; 87.4%-100.6%) and was mainly excreted into urine (mean, 79.0%; range, 70.3%-88.2%) and to a lesser extent into feces (mean, 15.5%; range, 9.3%-20.3%). Of the administered dose, 77.5% was recovered as unchanged cilengitide in urine. The concentration profiles of cilengitide and total radioactivity in plasma were comparable. No circulating metabolites were identified in plasma and urine. Two metabolites,M606-1 and M606-2, were identified in feces considered to be formed by intestinal peptidases or by peptidases from fecal bacteria. In conclusion, the data show that following intravenous administration, (14) C-cilengitide was completely recovered, was excreted mainly via renal elimination, and was not metabolized systemically.


Assuntos
Integrina alfaVbeta3/antagonistas & inibidores , Receptores de Vitronectina/antagonistas & inibidores , Venenos de Serpentes/farmacocinética , Adulto , Humanos , Infusões Intravenosas , Masculino , Venenos de Serpentes/administração & dosagem , Adulto Jovem
10.
J Ethnopharmacol ; 165: 1-8, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25681542

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Snakebite envenomation causes 5000-10,000 mortalities and results in more than 5-15,000 amputations in sub-Saharan Africa alone every year. The inaccessibility of antiserum therapy is a vast problem, and only about 2.5% of the actual need for antiserum in Africa is covered. Numerous plants have shown in vitro inhibitory activity against one or more of the hydrolytic enzymes involved in snakebite-induced necrosis. However, a more thorough examination of the plant species in ex vivo and in vitro cell assay models is needed to test their ability to inhibit necrosis. MATERIALS AND METHODS: Extracts which had previously shown in vitro inhibitory activity against necrosis enzymes, were tested in an ex vivo air-liquid-interface model, and a wound healing scratch assay as well as for their ability to permeate the skin barrier and inhibit venom induced cell death. RESULTS: Of the 14 water extracts and 16 ethanol extracts tested at a concentration of 10 µg/mL, only the ethanol extracts of Tamarindus indica and Paullinia pinnata resulted in a small but significant increase in cell migration of around 10% compared to treatment with buffer after 24h treatment. The remaining extracts showed no effect, or they even delayed the cell migration compared to the treatment with buffer. After 48 h treatment, 10 of the tested extracts showed a decreased cell migration compared to no treatment. At a 100 µg/mL concentration all the extracts inhibited cell migration and five extracts killed some of the cells, while four extracts killed all the cells. Ten of the thirty extracts were tested in a Franz cell set-up but none of the extracts tested did permeate the skin barrier over a 48 h period, and will therefore be of very limited use topically in the initial treatment of snakebites in its present form. None of the extracts were able to directly interact with the enzyme to lower the cell toxicity of the venom. Two extracts, Dichrostachys cinerea and Grewia mollis, were tested in the ex vivo model, but none of them inhibited the tissue destruction caused by venom. CONCLUSION: On the basis of this study, topical treatment with plant extracts for snakebite-induced tissue necrosis cannot be recommended.


Assuntos
Fitoterapia/métodos , Extratos Vegetais/uso terapêutico , Pele/patologia , Mordeduras de Serpentes/tratamento farmacológico , Cicatrização/efeitos dos fármacos , Células 3T3/efeitos dos fármacos , Administração Tópica , África Subsaariana , Animais , Morte Celular/efeitos dos fármacos , Humanos , Camundongos , Paullinia/química , Extratos Vegetais/farmacocinética , Pele/efeitos dos fármacos , Venenos de Serpentes/farmacocinética , Venenos de Serpentes/farmacologia , Suínos , Tamarindus/química
11.
J Clin Pharmacol ; 54(12): 1391-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24911832

RESUMO

Cilengitide is an αvß3/αvß5-integrin inhibitor investigated as an anticancer agent. This study aimed to develop a cilengitide population pharmacokinetic model using nonlinear mixed-effects modeling of 136 adult patients with advanced solid tumors and to scale the pharmacokinetic parameters to the pediatric population. A stepwise approach was used, beginning with exploratory analyses checking database/target covariate relationships. A two-compartment structural model was developed to describe cilengitide's concentration-time profile and assess covariates' impact on pharmacokinetic parameters. A bootstrap procedure validated the base/final model stability. A two-compartment model best described concentration-time data. Estimated structural model parameters were: 2.79 L h(-) (1) m(-) (2) central compartment mean systemic clearance, 6.75 L m(-) (2) central compartment volume of distribution, 1.3 L h(-) (1) m(-) (2) intercompartmental clearance, and 3.85 L m(-) (2) peripheral compartment volume of distribution. Mean half-life was 0.9 and 3.8 h (α/ß-phase). Co-medications and study populations had no impact, as the different studies were not significant model covariates. Weight and body surface area correlated with the pharmacokinetic parameters (r = 0.95, P < 0.01). Pharmacokinetic parameters were consistent with individual study-derived parameters; their allometric scaling enabled pediatric pharmacokinetic profile predictions as corroborated by independent data. This model provides the basis for pharmacokinetic profile simulations of different dosages/regimens in different populations.


Assuntos
Modelos Biológicos , Neoplasias/metabolismo , Venenos de Serpentes/farmacocinética , Adulto , Idoso , Idoso de 80 Anos ou mais , Área Sob a Curva , Superfície Corporal , Peso Corporal , Criança , Simulação por Computador , Humanos , Pessoa de Meia-Idade
12.
Neuro Oncol ; 15(10): 1438-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24014381

RESUMO

BACKGROUND: Cilengitide, an αv integrin antagonist, has demonstrated activity in recurrent adult glioblastoma (GBM). The Children's Oncology Group ACNS0621 study thus evaluated whether cilengitide is active as a single agent in the treatment of children with refractory high-grade glioma (HGG). Secondary objectives were to investigate the pharmacokinetics and pharmacogenomics of cilengitide in this population. METHODS: Cilengitide (1800 mg/m(2)/dose intravenous) was administered twice weekly until evidence of disease progression or unacceptable toxicity. Thirty patients (age range, 1.1-20.3 years) were enrolled, of whom 24 were evaluable for the primary response end point. RESULTS: Toxicity was infrequent and mild, with the exception of one episode of grade 2 pain possibly related to cilengitide. Two intratumoral hemorrhages were reported, but only one (grade 2) was deemed to be possibly related to cilengitide and was in the context of disease progression. One patient with GBM received cilengitide for 20 months and remains alive with continuous stable disease. There were no other responders, with median time to tumor progression of 28 days (range, 11-114 days). Twenty-one of the 24 evaluable patients died, with a median time from enrollment to death of 172 days (range, 28-325 days). The 3 patients alive at the time of this report had a follow-up time of 37, 223, and 1068 days, respectively. CONCLUSIONS: We conclude that cilengitide is not effective as a single agent for refractory pediatric HGG. However, further study evaluating combination therapy with cilengitide is warranted before a role for cilengitide in the treatment of pediatric HGG can be excluded.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Venenos de Serpentes/uso terapêutico , Adolescente , Adulto , Neoplasias Encefálicas/patologia , Criança , Pré-Escolar , Feminino , Seguimentos , Glioma/patologia , Humanos , Lactente , Masculino , Gradação de Tumores , Recidiva Local de Neoplasia/patologia , Prognóstico , Terapia de Salvação , Venenos de Serpentes/farmacocinética , Taxa de Sobrevida , Distribuição Tecidual , Adulto Jovem
13.
Cent Nerv Syst Agents Med Chem ; 12(4): 315-28, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23270323

RESUMO

The nervous system is a primary target for animal venoms as the impairment of its function results in the fast and efficient immobilization or death of a prey. There are numerous evidences about effects of crude snake venoms or isolated toxins on peripheral nervous system. However, the data on their interactions with the central nervous system (CNS) are not abundant, as the blood-brain barrier (BBB) impedes penetration of these compounds into brain. This updated review presents the data about interaction of snake venom polypeptides with CNS. Such data will be described according to three main modes of interactions: - Direct in vivo interaction of CNS with venom polypeptides either capable to penetrate BBB or injected into the brain. - In vitro interactions of cell or sub-cellular fractions of CNS with crude venoms or purified toxins. - Indirect effects of snake venoms or their components on functioning of CNS under different conditions. Although the venom components penetrating BBB are not numerous, they seem to be the most suitable candidates for the leads in drug design. The compounds with other modes of action are more abundant and better studied, but the lack of the data about their ability to penetrate BBB may substantially aggravate the potentials for their medical perspectives. Nevertheless, many such compounds are used for research of CNS in vitro. These investigations may give invaluable information for understanding the molecular basis of CNS diseases and thus lay the basis for targeted drug design. This aspect also will be outlined in the review.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Neurotoxinas/farmacologia , Peptídeos/farmacologia , Venenos de Serpentes/farmacologia , Analgésicos/isolamento & purificação , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Barreira Hematoencefálica , Sistema Nervoso Central/citologia , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Hemodinâmica/efeitos dos fármacos , Humanos , Injeções Intraventriculares , Camundongos , Fatores de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Neurotoxinas/química , Neurotoxinas/classificação , Neurotoxinas/isolamento & purificação , Neurotoxinas/farmacocinética , Neurotransmissores/agonistas , Neurotransmissores/antagonistas & inibidores , Manejo da Dor , Percepção da Dor/efeitos dos fármacos , Peptídeos/química , Peptídeos/isolamento & purificação , Peptídeos/farmacocinética , Peptídeos/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Venenos de Serpentes/química , Venenos de Serpentes/farmacocinética , Venenos de Serpentes/uso terapêutico , Serpentes , Frações Subcelulares/efeitos dos fármacos
14.
Melanoma Res ; 22(4): 294-301, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22668797

RESUMO

Cilengitide (EMD 121974) is a selective inhibitor of integrins αvß3 and αvß5. The αvß3 promotes the proliferation of tumor-associated endothelial cells and potentially the survival of melanoma cells. We conducted a randomized phase II trial in patients with metastatic melanoma to evaluate the clinical efficacy of cilengitide. Patients with stage IV or unresectable stage III melanoma who were either chemonaive or who had previously received one systemic therapy were enrolled. Patients were randomly assigned to either 500 or 2000 mg of cilengitide administered intravenously twice weekly. The primary aim of this study was to determine the progression-free survival rate at 8 weeks. Tumor samples and blood samples were collected for pharmacodynamic and pharmacokinetic studies. Twenty-nine patients were enrolled, of whom 26 were treated (14 at 500 mg and 12 at 2000 mg). Among those treated, only three were progression free at 8 weeks: two in the 500 mg arm and one in the 2000 mg arm. One patient in the 2000 mg arm showed a prolonged partial response after an initial 28% enlargement of her target lesions. The treatment was well tolerated without clinically significant adverse events. The sole responder and one of two patients with stable disease had no αvß3 expression at baseline. Overall, αvß3 expression was decreased by day 8 of the treatment (P=0.05). Cilengitide was well tolerated by patients in both the treatment arms but had minimal clinical efficacy as a single-agent therapy for metastatic melanoma, and the efficacy was not related to baseline αvß3 expression.


Assuntos
Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Venenos de Serpentes/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Masculino , Melanoma/metabolismo , Melanoma/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Venenos de Serpentes/efeitos adversos , Venenos de Serpentes/farmacocinética
15.
Invest New Drugs ; 30(2): 604-10, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20839028

RESUMO

BACKGROUND: Cilengitide (EMD121974) is a cyclized pentapeptide that is a potent and selective integrin antagonist which has shown activity in malignant gliomas. In all previous studies, cilengitide has been administered in an intermittent fashion. However, cilengitide has a short half-life of 3-5 h with no evidence of drug accumulation. These data prompted the initiation of this phase I study of continuous infusion cilengitide. METHODS: Cilengitide was administered as a continuous infusion without break in 4-week cycles. Plasma samples for pharmacokinetic studies were obtained weekly in cycle 1 immediately prior to and 2 h after infusion bag change. RESULTS: Thirty-five patients were treated (median age 56; 23 males) at dose levels of 1, 2, 4, 8, 12, 18, 27, and 40 mg/h. Toxicities were limited to grade ≤ 2 and showed no relation to dose. Fatigue was most common (17%), while all other toxicities were reported in <10% of patients. No dose-limiting toxicities were observed, and therefore the maximum tolerated dose was not reached. Pharmacokinetic analysis showed that values for clearance and volume of distribution were comparable across dose levels, and the steady-state concentration increased proportionally with dose. CONCLUSIONS: Cilengitide can be safely administered as a continuous infusion at doses up to at least 40 mg/h, which represents the maximum feasible dose due to drug solubility and delivery limitations. The pharmacokinetics of continuous infusion cilengitide are linear and consistent with the results obtained using a twice weekly infusion.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias/tratamento farmacológico , Venenos de Serpentes/administração & dosagem , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Chicago , Feminino , Humanos , Infusões Intravenosas , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/patologia , Venenos de Serpentes/efeitos adversos , Venenos de Serpentes/sangue , Venenos de Serpentes/farmacocinética , Resultado do Tratamento
16.
J Neurooncol ; 106(1): 147-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21739168

RESUMO

Cilengitide is a cyclic pentapeptide that is a specific inhibitor of the αvß3 and αvß5 integrins. Preclinical studies demonstrate antiangiogenic activity and anti-invasive activity in a number of glioma models. This study was designed to evaluate the efficacy and tumor delivery of cilengitide in patients with recurrent glioblastoma. Patients with recurrent glioblastoma who require a surgical resection for optimal clinical care received 3 intravenous doses of cilengitide at either 500 or 2000 mg (day -8, -4, -1) prior to undergoing tumor resection with corresponding blood samples for plasma to tumor comparisons. After recovery from surgery, patients were treated with cilengitide (2000 mg i.v. twice weekly, maximum of 2 years of treatment). The study accrued 30 patients with recurrent glioblastoma, 26 were evaluable for efficacy. The 6-month progression free survival rate was 12%. Cilengitide was detected in all tumor specimens with higher levels in the group receiving 2000 mg dosing while corresponding plasma concentrations were low, often below the lower limit of detection. These results confirm drug delivery and possibly retention in tumor. This study provides evidence that with established dosing, cilengitide is adequately delivered to the tumor, although as a single agent, efficacy in recurrent glioblastoma is modest. However, these results demonstrating drug delivery to tumor do support continued investigation of this agent as preliminary results from recent studies combining cilengitide with cytotoxic therapies are promising.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Venenos de Serpentes/uso terapêutico , Adulto , Idoso , Neoplasias Encefálicas/cirurgia , Terapia Combinada , Intervalo Livre de Doença , Determinação de Ponto Final , Feminino , Glioblastoma/cirurgia , Humanos , Imunoterapia , Integrinas/fisiologia , Avaliação de Estado de Karnofsky , Masculino , Pessoa de Meia-Idade , Recidiva , Venenos de Serpentes/efeitos adversos , Venenos de Serpentes/farmacocinética
17.
Nat Med ; 17(7): 809-11, 2011 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-21706027

RESUMO

Snake venom toxins first transit the lymphatic system before entering the bloodstream. Ointment containing a nitric oxide donor, which impedes the intrinsic lymphatic pump, prolonged lymph transit time in rats and humans and also increased rat survival time after injection of venom. This pharmacological approach should give snakebite victims more time to obtain medical care and antivenom treatment.


Assuntos
Primeiros Socorros/métodos , Doadores de Óxido Nítrico/uso terapêutico , Nitroglicerina/uso terapêutico , Mordeduras de Serpentes/tratamento farmacológico , Adulto , Idoso , Animais , Feminino , Humanos , Sistema Linfático/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Doadores de Óxido Nítrico/administração & dosagem , Nitroglicerina/administração & dosagem , Pomadas , Ratos , Venenos de Serpentes/administração & dosagem , Venenos de Serpentes/antagonistas & inibidores , Venenos de Serpentes/farmacocinética , Adulto Jovem
18.
Future Oncol ; 7(3): 339-54, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21417900

RESUMO

Cilengitide, a cyclicized arginine-glycine-aspartic acid-containing pentapeptide, potently blocks ανß3 and ανß5 integrin activation. Integrins are upregulated in many malignancies and mediate a wide variety of tumor-stroma interactions. Cilengitide and other integrin-targeting therapeutics have preclinical activity against many cancer subtypes including glioblastoma (GBM), the most common and deadliest CNS tumor. Cilengitide is active against orthotopic GBM xenografts and can augment radiotherapy and chemotherapy in these models. In Phase I and II GBM trials, cilengitide and the combination of cilengitide with standard temozolomide and radiation demonstrate consistent antitumor activity and a favorable safety profile. Cilengitide is currently under evaluation in a pivotal, randomized Phase III study (Cilengitide in Combination With Temozolomide and Radiotherapy in Newly Diagnosed Glioblastoma Phase III Randomized Clinical Trial [CENTRIC]) for newly diagnosed GBM. In addition, randomized controlled Phase II studies with cilengitide are ongoing for non-small-cell lung cancer and squamous cell carcinoma of the head and neck. Cilengitide is the first integrin inhibitor in clinical Phase III development for oncology.


Assuntos
Antineoplásicos/uso terapêutico , Glioblastoma/tratamento farmacológico , Integrinas/antagonistas & inibidores , Oligopeptídeos/uso terapêutico , Venenos de Serpentes/uso terapêutico , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/farmacocinética , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Indústria Farmacêutica/tendências , Humanos , Oligopeptídeos/efeitos adversos , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Venenos de Serpentes/efeitos adversos , Venenos de Serpentes/química , Venenos de Serpentes/farmacocinética , Resultado do Tratamento
19.
Int J Cancer ; 124(11): 2719-27, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19199360

RESUMO

We investigated whether cilengitide could amplify the antitumor effects of radiotherapy in an orthotopic rat glioma xenograft model. Cilengitide is a specific inhibitor of alphav series integrins, and acts as an antiangiogenic. U251 human glioma cells express alphavbeta3 and alphavbeta5 integrins. We used in vitro assays of adhesion and growth of tumor and endothelial cells to evaluate cytotoxicity and the potential for cilengitide to enhance radiation toxicity. Treatment was then evaluated in an orthotopic model to evaluate synergy with therapeutic radiation in vivo. In vitro, cilengitide blocked cell adhesion, but did not influence the effects of radiation on U251 cells; cilengitide strongly amplified radiation effects on endothelial cell survival. In vivo, radiotherapy prolonged the survival of U251 tumor-bearing rats from 50 to over 110 days. Cotreatment with cilengitide and radiation dramatically amplified the effects of radiation, producing survival over 200 days and triggering an enhanced apoptotic response and suppression of tumor growth by histology at necropsy. Signaling pathways activated in the tumor included NFkappab, a documented mediator of cellular response to radiation. Because cilengitide has a short plasma half-life (t((1/2)) approximately 20 min), antiangiogenic scheduling typically uses daily injections. We found that a single dose of cilengitide (4 mg/kg) given between 4 and 12 hr prior to radiation was sufficient to produce the same effect. Our results demonstrate that blockade of alphav integrins mediates an unanticipated rapid potentiation of radiation, and suggests possible clinical translation for glioma therapy.


Assuntos
Glioblastoma/radioterapia , Integrina alfaVbeta3/antagonistas & inibidores , Radiossensibilizantes/farmacologia , Receptores de Vitronectina/antagonistas & inibidores , Venenos de Serpentes/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Adesão Celular/efeitos dos fármacos , Adesão Celular/efeitos da radiação , Linhagem Celular Tumoral , Células Endoteliais/efeitos da radiação , Glioblastoma/patologia , Humanos , Integrina alfaVbeta3/análise , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Wistar , Receptores de Vitronectina/análise , Venenos de Serpentes/farmacocinética , Fator de Transcrição RelA/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Clin Oncol ; 26(6): 919-24, 2008 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-18281665

RESUMO

PURPOSE: A phase I trial of the antiangiogenesis agent cilengitide (EMD 121974), an alpha v beta 3,5 integrin antagonist, was performed to estimate the maximum-tolerated dose (MTD) and describe dose-limiting toxicities (DLTs) and the incidence and severity of other toxicities when administered to children with refractory brain tumors. PATIENTS AND METHODS: Thirty-one assessable patients received intravenous cilengitide over 1 hour twice a week for up to 52 weeks at dosages from 120 to 2,400 mg/m(2). Serial blood and urine samples for clinical pharmacology studies were obtained in a subset of consenting patients. RESULTS: No DLTs were observed, and thus, the MTD was not estimated. Three of 13 patients at the dosage level of 2,400 mg/m(2) experienced grade 3 or 4 intratumoral hemorrhage (ITH) possibly related to the study drug; however, two of the ITH events were asymptomatic and, by the current toxicity criteria, would be classified as grade 1. For patients treated at cilengitide 2,400 mg/m(2), the 6-month cumulative incidence estimate of ITH is 23% (SE = 13%). No ITH was observed at 1,800 mg/m(2). Three patients completed 1 year of protocol therapy; one patient with glioblastoma multiforme demonstrated complete response, and two patients had stable disease (SD). An additional patient had SD for more than 5 months. CONCLUSION: The phase II dosage of intravenous cilengitide in children with refractory brain tumors is 1,800 mg/m(2). A phase II trial to assess the efficacy of cilengitide therapy for children with refractory brain tumors is being developed by the Children's Oncology Group.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Neoplasias Encefálicas/tratamento farmacológico , Venenos de Serpentes/administração & dosagem , Venenos de Serpentes/efeitos adversos , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Adolescente , Adulto , Inibidores da Angiogênese/farmacocinética , Neoplasias Encefálicas/patologia , Criança , Pré-Escolar , Esquema de Medicação , Feminino , Genótipo , Humanos , Lactente , Infusões Intravenosas , Imageamento por Ressonância Magnética , Masculino , Venenos de Serpentes/farmacocinética , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA