Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell Prolif ; 56(2): e13362, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36433732

RESUMO

Acute kidney injury (AKI) is often secondary to sepsis. Increasing evidence suggests that mitochondrial dysfunction contributes to the pathological process of AKI. In this study, we aimed to examine the regulatory roles of Sirt3 in Lipopolysaccharide (LPS)-induced mitochondrial damage in renal tubular epithelial cells (TECs). Sirt3 knockout mice were intraperitoneally injected with LPS, and cultured TECs were stimulated with LPS to evaluate the effects of Sirt3 on mitochondrial structure and function in TECs. Electron microscopy was used to assess mitochondrial morphology. Immunofluorescence staining was performed to detect protein expression and examine mitochondrial morphology. Western blotting was used to quantify protein expression. We observed that LPS increased apoptosis, induced disturbances in mitochondrial function and dynamics, and downregulated Sirt3 expression in a sepsis-induced AKI mouse model and human proximal tubular (HK-2) cells in vitro. Sirt3 deficiency further exacerbated LPS-induced renal pathological damage, apoptosis and disturbances in mitochondrial function and dynamics. On the contrary, Sirt3 overexpression in HK-2 cells alleviated these lesions. Functional studies revealed that Sirt3 overexpression alleviated LPS-induced mitochondrial damage and apoptosis in TECs by promoting OPA1-mediated mitochondrial fusion through the deacetylation of i-AAA protease (YME1L1), an upstream regulatory molecule of OPA1. Our study has identified Sirt3 as a vital factor that protects against LPS-induced mitochondrial damage and apoptosis in TECs via the YME1L1-OPA1 signaling pathway.


Assuntos
Injúria Renal Aguda , Sepse , Sirtuína 3 , Camundongos , Animais , Humanos , Sirtuína 3/metabolismo , Lipopolissacarídeos/farmacologia , Injúria Renal Aguda/metabolismo , Células Epiteliais/metabolismo , Camundongos Knockout , Apoptose , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/metabolismo , Proteínas Mitocondriais/metabolismo
2.
Lancet Neurol ; 20(9): 721-728, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34418399

RESUMO

BACKGROUND: Non-immunogenic staphylokinase is modified recombinant staphylokinase with low immunogenicity, high thrombolytic activity, and selectivity to fibrin. We aimed to assess the safety and efficacy of a single intravenous bolus of non-immunogenic staphylokinase compared with alteplase in patients with acute ischaemic stroke within 4·5 h after symptom onset. METHODS: We did a randomised, open-label, multicentre, parallel-group, non-inferiority trial in 18 clinical sites in Russia. We included patients aged 18 years and older with a diagnosis of acute ischaemic stroke (up to 25 points on the National Institutes of Health Stroke Scale). The study drug had to be administered within 4·5 h after the onset of symptoms. Patients were randomly assigned to receive either non-immunogenic staphylokinase (10 mg) or alteplase (0·9 mg/kg, maximum 90 mg), both administered intravenously. The randomisation sequence was created by an independent biostatistician using computer-generated random numbers. 84 blocks (block size of four) of opaque sealed envelopes were numbered sequentially from 1 to 336 and were opened in numerical order. Patients were unaware of their assigned treatment and were assessed by the study investigators who were also unaware of the treatment assignment on all trial days. Emergency department staff, who administered the assigned drug and opened the envelopes, were not masked to treatment. The primary efficacy endpoint was a favourable outcome, defined as a modified Rankin scale (mRS) score of 0-1 on day 90. The margin of non-inferiority was established as 16% for the difference in mRS score of 0-1 on day 90. Non-inferiority was tested using Welch's t-test for the primary outcome only. Endpoints were analysed in the per-protocol population, which comprised all randomly assigned patients who completed treatment without any protocol violations; this population was identical to the intention-to-treat population. This trial is completed and registered at ClinicalTrials.gov, NCT03151993. FINDINGS: Of 385 patients recruited from March 18, 2017, to March 23, 2019, 336 (87%) were included in the trial. 168 (50%) patients were randomly assigned to receive non-immunogenic staphylokinase and 168 (50%) to receive alteplase. The median duration of follow-up was 89 days (IQR 89-89). 84 (50%) of 168 patients in the non-immunogenic staphylokinase group had a favourable outcome at day 90 compared with 68 (40%) of 168 patients in the alteplase group (odds ratio [OR] 1·47, 95% CI 0·93 to 2·32; p=0·10). The difference in the rate of favourable outcome at day 90 was 9·5% (95% CI -1·7 to 20·7) and the lower limit did not cross the margin of non-inferiority (pnon-inferiority <0·0001). Symptomatic intracranial haemorrhage occurred in five (3%) patients in the non-immunogenic staphylokinase group and in 13 (8%) patients in the alteplase group (p=0·087). On day 90, 17 (10%) patients in the non-immunogenic staphylokinase group and 24 (14%) patients in the alteplase group had died (p=0·32). 22 (13%) patients in the non-immunogenic staphylokinase group had serious adverse events, compared with 37 (22%) patients in the alteplase group (p=0·044). INTERPRETATION: Non-immunogenic staphylokinase was non-inferior to alteplase for patients with acute ischaemic stroke. Mortality, symptomatic intracranial haemorrhage, and serious adverse events did not differ significantly between groups. Future studies are needed to continue to assess the safety and efficacy of non-immunogenic staphylokinase in patients with acute ischaemic stroke within the 4·5 h time window, and to assess the drug in patients with acute ischaemic stroke outside this time window with reperfusion CT or magnetic resonance angiography followed by thrombectomy if necessary. FUNDING: The Russian Academy of Sciences.


Assuntos
Fibrinolíticos/farmacologia , AVC Isquêmico/tratamento farmacológico , Metaloendopeptidases/farmacologia , Tempo para o Tratamento , Ativador de Plasminogênio Tecidual/farmacologia , Idoso , Idoso de 80 Anos ou mais , Método Duplo-Cego , Feminino , Fibrinolíticos/administração & dosagem , Fibrinolíticos/efeitos adversos , Humanos , Masculino , Metaloendopeptidases/administração & dosagem , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/imunologia , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde , Proteínas Recombinantes , Federação Russa , Ativador de Plasminogênio Tecidual/administração & dosagem , Ativador de Plasminogênio Tecidual/efeitos adversos
3.
Ann Hematol ; 90(5): 569-77, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20957364

RESUMO

NN-PF3 is a non-toxic, anticoagulant, high-molecular-mass (67.81 kDa) metalloprotease from Indian cobra (Naja naja) venom. In the present study, NN-PF3 was investigated for the mechanism of inhibition of collagen-induced aggregation of human platelets. The complete inhibition of collagen-induced aggregation and partial inhibition of ADP- and epinephrine-induced aggregation has the respective IC(50) of 75 ± 5, 185 ± 10, and 232 ± 12 nM, whereas no inhibition of thrombin-, arachidonic acid-, and ristocetin-induced aggregation of platelets was observed in platelet-rich plasma. Further, native NN-PF3 and EDTA-inactivated NN-PF3 inhibited collagen-induced aggregation of washed platelets with respective IC(50) of 75 ± 4 and 180 ± 6 nM. The higher inhibitory effect of native NN-PF3 compared with EDTA-inactivated NN-PF3 suggests the enzymatic and non-enzymatic mechanism of inhibition. NN-PF3 pretreatment affected the collagen binding but not the fibrinogen, and fibronectin binding of washed platelets in adhesion assay suggested that the collagen receptors are affected. Western blot study using anti-integrin α2ß1 mAb 6F1 suggested that NN-PF3 binds to integrin α2ß1 in a primary structure-dependent manner only and is not cleaved. There was a drastic reduction in the intensity of several intracellular signaling phosphotyrosine protein bands when monoclonal anti-phosphotyrosine antibody was used, suggesting that the major activation pathway of platelets get affected, which occurs through glycoprotein VI. NN-PF3 did not bind to collagen as revealed by Western blot using anti-collagen mAb. Furthermore, neither the proteolytic cleavage of fibrinogen nor its degradation products by NN-PF3 contributed for the collagen-induced platelet aggregation inhibition.


Assuntos
Plaquetas/efeitos dos fármacos , Venenos Elapídicos/enzimologia , Integrina alfa2beta1/metabolismo , Metaloendopeptidases/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Animais , Anticoagulantes/efeitos adversos , Anticoagulantes/antagonistas & inibidores , Anticoagulantes/metabolismo , Anticoagulantes/farmacologia , Cálcio/metabolismo , Quelantes/farmacologia , Colágeno Tipo I/antagonistas & inibidores , Colágeno Tipo I/metabolismo , Colágeno Tipo I/farmacologia , Ácido Edético/farmacologia , Humanos , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/efeitos adversos , Inibidores da Agregação Plaquetária/metabolismo , Contagem de Plaquetas , Glicoproteínas da Membrana de Plaquetas/metabolismo , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Zinco/metabolismo
5.
J Vasc Surg ; 51(3): 600-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20110153

RESUMO

PURPOSE: To investigate the safety and effectiveness of a novel thrombolytic, alfimeprase, in catheter-directed thrombolysis (CDT) of acute peripheral arterial occlusions (PAO). METHODS: Between April 2005 and March 2007, patients with acute PAO (Rutherford class I or IIa) of a lower extremity and onset of symptoms within 14 days prior to randomization were included. Studies HA004 and HA007 enrolled respectively 300 and 102 patients. Both studies HA004 and HA007 were placebo-controlled. HA004 had two placebo arms, intrathrombus and perithrombus, while HA007 had intrathrombus placebo arm. HA004 was partially double-blind (perithrombus group was not blinded) and HA007 was double-blind. Patients were randomized to intrathrombus alfimeprase (0.3 mg/kg), intrathrombus (IT) placebo, or perithrombus (PT) placebo (HA004 only) in two divided weight-based infusions 2 hours apart. Depending on arteriographic results after treatment, patients received no further intervention or underwent endovascular therapy or open vascular surgery. The primary endpoint of both studies was efficacy of alfimeprase compared with placebo as measured by avoidance of an open vascular surgery procedure at 30 days. RESULTS: The avoidance of open vascular surgery at 30 days was seen in 52 (34.9%), 42 (37.2%), and 7 patients (18.4%) with alfimeprase, IT placebo, and PT placebo in HA004 and 15 (29.4%) and 9 patients (17.6%) with alfimeprase and IT placebo in HA007; differences between alfimeprase and IT placebo were not statistically significant. Results were similar for secondary endpoints, including arterial flow restoration in 4 hours, 30-day ankle-brachial index, index limb pain severity, and hospital stay duration. The overall rate of adverse events was higher with alfimeprase than placebo. Hemorrhagic and peripheral embolic event rates with alfimeprase were 23% (34 patients) and 10.1% (15 patients) in HA004 and 9.4% (5 patients) and 9.8% (5 patients) in HA007; rates with IT placebo were 11% (12 patients, P = .107) and 5% (5 patients, P = .148) in HA004 and 10% (5 patients, P = .982) and 0% in HA007 (P = .07). No deaths were related to study drug administration. CONCLUSIONS: CDT for acute PAO with alfimeprase was as safe as placebo. However, alfimeprase was no more effective than placebo in increasing 30-day surgery-free survival. The surprising effectiveness of placebo alone demonstrates that the inclusion of a placebo arm is essential to the design of future lytic trials.


Assuntos
Arteriopatias Oclusivas/tratamento farmacológico , Fibrinolíticos/administração & dosagem , Oclusão de Enxerto Vascular/tratamento farmacológico , Metaloendopeptidases/administração & dosagem , Terapia Trombolítica , Doença Aguda , Adulto , Idoso , Idoso de 80 Anos ou mais , Tornozelo/irrigação sanguínea , Arteriopatias Oclusivas/complicações , Arteriopatias Oclusivas/diagnóstico por imagem , Arteriopatias Oclusivas/fisiopatologia , Arteriopatias Oclusivas/cirurgia , Pressão Sanguínea , Artéria Braquial/fisiopatologia , Cateterismo Periférico , Método Duplo-Cego , Feminino , Fibrinolíticos/efeitos adversos , Oclusão de Enxerto Vascular/complicações , Oclusão de Enxerto Vascular/diagnóstico por imagem , Oclusão de Enxerto Vascular/fisiopatologia , Oclusão de Enxerto Vascular/cirurgia , Humanos , Infusões Intra-Arteriais , Claudicação Intermitente/etiologia , Tempo de Internação , Masculino , Metaloendopeptidases/efeitos adversos , Pessoa de Meia-Idade , Razão de Chances , Medição da Dor , Efeito Placebo , Pulsoterapia , Radiografia , Fluxo Sanguíneo Regional , Medição de Risco , Terapia Trombolítica/efeitos adversos , Fatores de Tempo , Resultado do Tratamento , Estados Unidos , Procedimentos Cirúrgicos Vasculares , Adulto Jovem
6.
Neurol Res ; 31(2): 209-14, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19298764

RESUMO

OBJECTIVE: Recanalization therapies for ischemic stroke have been slow to change clinical practice because of perceived and published risks of hemorrhage associated with lytic administration. We quantified alfimeprase in an acute ischemia-reperfusion model, as compared with recombinant tissue plasminogen activator, with hemorrhagic transformation as the primary endpoint and infarction volume and blood-brain barrier permeability as secondary endpoints. METHODS: Five groups were studied in a blinded fashion: alfimeprase at doses of 0.03 (n=8), 0.1 (n=11) and 0.3 mg/kg (n=8); recombinant tissue plasminogen activator at 1 mg/kg (n=9); carrier infused controls (n=9). The middle cerebral artery was occluded for 5 hours followed by removal of the suture for reperfusion. Drugs were infused immediately following reperfusion over a 10-minute period. Approximately 24 hours later, the animals were anesthetized and decapitated, and the brains were rapidly harvested and frozen. Serial brain sections were obtained and inspected for hemorrhages. Infarction and blood-brain barrier permeability were also evaluated in additional experiments in control, 0.1 mg/kg alfimeprase and 1 mg/kg recombinant tissue plasminogen activator-treated rats. RESULTS: The hemorrhagic transformation frequency, neurological deficit and the mortality rate of alfimeprase were significantly lower than for recombinant tissue plasminogen activator at the 0.03 mg/kg dose and not statistically different at the higher doses. Infarction and blood-brain barrier permeability were not significantly different among control, 0.1 mg/kg alfimeprase and recombinant tissue plasminogen activator. DISCUSSION: In this model, alfimeprase, a new fibrinolytic agent, exhibits a profile comparable to recombinant tissue plasminogen activator.


Assuntos
Hemorragia Cerebral/induzido quimicamente , Fibrinolíticos/efeitos adversos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Metaloendopeptidases/efeitos adversos , Traumatismo por Reperfusão/induzido quimicamente , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Peso Corporal/efeitos dos fármacos , Edema Encefálico/tratamento farmacológico , Edema Encefálico/patologia , Permeabilidade Capilar/efeitos dos fármacos , Hemorragia Cerebral/mortalidade , Hemorragia Cerebral/patologia , Hemorragia Cerebral/fisiopatologia , Distribuição de Qui-Quadrado , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Método Duplo-Cego , Fibrinolíticos/uso terapêutico , Infarto da Artéria Cerebral Média/mortalidade , Masculino , Metaloendopeptidases/efeitos dos fármacos , Exame Neurológico , Ratos , Traumatismo por Reperfusão/mortalidade , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Fatores de Tempo , Ativador de Plasminogênio Tecidual/administração & dosagem
7.
Drugs R D ; 9(3): 185-90, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18457471

RESUMO

Alfimeprase, a fibrolase derivative with thrombolytic activity produced by recombinant DNA technology, was discovered by Amgen and was in development with Nuvelo for the treatment of stroke and catheter occlusion. However, development has been discontinued. Fibrolase is a zinc-containing metalloendopeptidase that was first isolated from the venom of the Southern copperhead snake, Agkistrodon contortrix contortrix. Alfimeprase directly degrades fibrin to break down clots. Alfimeprase is infused directly into the thrombus (side-hole catheter pushed through the entire clot) via multiple manual pulsed infusions. Alfimeprase degrades fibrin directly and entrapped blood cells are freed. Excess alfimeprase is rapidly inactivated by alpha-2 macroglobulin through an irreversible, covalent interaction. Phase III development of alfimeprase for peripheral arterial occlusion was discontinued based on poor results from the NAPA-2 and SONOMA-2 trials; however, Nuvelo resumed development of alfimeprase in 2007 for stroke and catheter occlusion.Alfimeprase's thrombolytic activity appears to be localized to the site of delivery because it is rapidly inactivated by alpha-2 macroglobulin, a naturally occurring protein in the blood, as it moves away from the site of delivery and into general blood circulation. In January 2002, Amgen and Hyseq Pharmaceuticals (now Nuvelo) entered into a collaboration to develop and commercialize alfimeprase. Nuvelo is to develop the product through clinical trials and Amgen was to be responsible for its manufacture. Both companies were to participate in commercial activities; Amgen was to have the option to lead these. Full financial terms were not disclosed. Further to this agreement, in November 2004 Amgen granted Nuvelo worldwide rights to develop and commercialize alfimeprase in exchange for milestone and royalty payments. In August 2007, Nuvelo decided to focus on core development programmes that it believed would produce the nearest-term proof-of-concept data. As a result of this realignment of organizational expenses, Nuvelo decided to continue to pursue the development of alfimeprase. In February 2003, Hyseq Pharmaceuticals merged with Variagenics Inc. to form Nuvelo Inc. In January 2006, Nuvelo and Bayer HealthCare entered a collaboration to develop and commercialize alfimeprase. Bayer was to commercialize the drug in all territories outside the US, whilst Nuvelo retains full US rights. Nuvelo was to receive other territory royalties and milestone payments to a total of $US385 million. A $US50 million upfront payment will be made to Nuvelo, while Bayer was to be responsible for 40% of the commercialization cost for global development. This partnership was to also develop stroke and deep vein thrombosis therapies. Data from the SONOMA-3 trial fell short of the company's expectations and so Nuvelo has decided to discontinue further development of alfimeprase. Nuvelo previously had decided to resume development of alfimeprase for the treatment of multiple coagulation-related disorders, including acute ischaemic stroke, catheter occlusion (CO) and acute peripheral arterial occlusion.Previously, results from the NAPA-2 (Novel Arterial Perfusion with Alfimeprase-2) trial and SONOMA-2 (Speedy Opening of Non-functional and Occluded catheters with Mini-dose Alfimeprase) phase III trial of alfimeprase in patients with acute peripheral arterial occlusion and catheter occlusion did not meet their primary endpoints. The primary endpoint of the NAPA-2 trial was the avoidance of surgery within 30 days of treatment with alfimeprase, whilst the SONOMA-2 trial's endpoint was the restoration of function at 15 minutes after dosing. In addition, these trials did not meet their secondary endpoints. As a result, Nuvelo and Bayer suspended enrolment in the NAPA-3 and SONOMA-3 phase III trials until additional analysis was complete; the SONOMA-3 trial was re-initiated. Nuvelo concluded that the delivery method for alfimeprase in the treatment of peripheral arterial occlusive disorders was suboptimal. The company closed the suspended NAPA-3 trial and planned to initiate preclinical studies focused on identifying optimized delivery methods in acute PAO in the second half of 2007. Data from the NAPA-2 trial suggested that efficacy could potentially be enhanced by maintaining alfimeprase longer at the site of the thrombus. Nuvelo's multinational phase III programme for peripheral arterial occlusion consisted of the NAPA-2 and NAPA-3 trials. Both trials were randomized, double-blind studies comparing 0.3 mg/kg of alfimeprase with placebo in a total of 600 patients in 100 centres. The primary endpoint of the NAPA-2 trial was the avoidance of surgery within 30 days of treatment; secondary endpoints included safety and pharmacoeconomics such as length of hospital and intensive care unit stay. Results from both trials have been presented. The phase II trial (NAPA-1) was completed in June 2004. This open-label, dose-escalation trial assessed the safety and efficacy of alfimeprase in 115 patients with acute peripheral arterial occlusion and was conducted in centres across the US, Europe and South Africa. Full data from the trial have been presented, which indicated that the 0.3 mg/kg dose appeared to be the optimal dose for investigation in phase III trials. In March 2003, Nuvelo announced the positive results of a phase I trial initiated in the US in July 2002; an IND was transferred from Amgen to Nuvelo in January 2002. In the SONOMA-2 trial, alfimeprase restored catheter function in patients with occluded catheters within 15 minutes with a p-value of 0.022. However, it did not meet the company's target product profile for commercial success with a p-value < 0.00125. Data from a phase II trial were presented at the 46th Annual Meeting of the American Society of Hematology held in December 2004. The study was closed in July 2004 with 55 patients enrolled. Initially the phase II study was to compare three doses of alfimeprase with the approved dose of alteplase in over 90 patients in the US. Nuvelo initiated the phase II CARNEROS-1 (Catheter Directed Alfimeprase for Restoration of Neurologic Function and Rapid Opening of Arteries in Stroke) study of alfimeprase in the treatment of acute ischaemic stroke in June 2007. CARNEROS-1 was a multicentre, open-label, dose-escalation study beginning with doses of 1, 5 and 10 mg of alfimeprase in 100 patients within 3-9 hours of stroke onset. The primary endpoints were recanalization (unblocking) of the occlusive lesion within 120 minutes of treatment, and symptomatic intracerebral haemorrhage. The first patient was dosed in December 2007; enrolment was taking place in the US and Canada. In the US, three patents have been issued relating to alfimeprase; US Patent Nos 6 261 820 (alfimeprase protein sequence), 6 440 414 (formulation of alfimeprase with a zinc stabilizer) and 6 455 269 (methods for localized administration of alfimeprase).


Assuntos
Desenho de Fármacos , Fibrinolíticos/farmacologia , Metaloendopeptidases/farmacologia , Arteriopatias Oclusivas/tratamento farmacológico , Ensaios Clínicos como Assunto , Fibrinolíticos/efeitos adversos , Fibrinolíticos/uso terapêutico , Humanos , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/uso terapêutico
8.
Thromb Haemost ; 97(6): 1037-45, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17549308

RESUMO

The recombinant protein SAK-RGD-K2-Hir is characterized by its fibrin-specific properties of plasminogen activation combined with antithrombin and antiplatelet activities. It was previously shown in our in-vitro studies to be a more potent and faster-acting thrombolytic agent compared with standard r-SAK. In order to document the effects of the thrombolytic potential of SAK-RGD-K2-Hir we examined this protein in an electrically induced carotid artery thrombosis model and stasis-induced venous model in rats. In the arterial thrombosis model, a bolus injection of SAK-RGD-K2-Hir was less effective than rt-PA and r-SAK. However, the most effective in the improvement and maintenance of carotid patency and in arterial thrombus mass reduction was SAK-RGD-K2. In contrast, all r-SAK derivatives reduced venous thrombus weight significantly in comparison to r-SAK and r-Hir. However, the most observable decrease in thrombus weight was obtained after application of recombinant proteins containing the r-Hir. The bleeding time was significantly prolonged in the animals treated with proteins containing r-Hir at a dose of 1.0 mg/kg. There were no observable changes in plasma fibrinogen concentration. In conclusion, our findings show thrombolytic activity in intravenous bolus injection of the novel thrombolytic agent SAK-RGD-K2-Hir in rats. Although this protein compares favourably with r-SAK in rat venous thrombolysis, we were unable to confirm the beneficial effects of SAK-RGD-K2-Hir over r-SAK and rt-PA in the carotid artery thrombolysis model. Furthermore, our results also suggest that SAK-RGD-K2-Hir bears a risk of bleeding, but this may be true for higher doses.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Trombose das Artérias Carótidas/tratamento farmacológico , Fibrina/metabolismo , Fibrinolíticos/farmacologia , Hirudinas/farmacologia , Metaloendopeptidases/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Grau de Desobstrução Vascular/efeitos dos fármacos , Trombose Venosa/tratamento farmacológico , Animais , Tempo de Sangramento , Trombose das Artérias Carótidas/sangue , Trombose das Artérias Carótidas/metabolismo , Trombose das Artérias Carótidas/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estimulação Elétrica , Fibrinolíticos/efeitos adversos , Fibrinolíticos/metabolismo , Hemorragia/induzido quimicamente , Hirudinas/efeitos adversos , Hirudinas/metabolismo , Ligadura , Masculino , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/metabolismo , Tempo de Tromboplastina Parcial , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/metabolismo , Tempo de Trombina , Fatores de Tempo , Ativador de Plasminogênio Tecidual/farmacologia , Veias Cavas/cirurgia , Trombose Venosa/sangue , Trombose Venosa/metabolismo
9.
IDrugs ; 10(5): 329-35, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17487785

RESUMO

Alfimeprase is an analog of a fibrolase that disrupts formed thrombi through the hydrolysis of fibrin, rather than by activation of plasminogen. Nuvelo Inc, under license from Amgen Inc and together with Bayer AG, is developing this thrombolytic for the potential intravenous treatment of peripheral arterial occlusions and for other cardiovascular indications. Pharmacokinetic studies showed that alfimeprase was rapidly absorbed and achieved therapeutic concentrations at relatively low doses. Preclinical studies showed that adjunctive therapy with antiplatelet agents was necessary to maintain luminal patency. In phase I and II clinical trials alfimeprase effectively thombolysed clots with no drug-related adverse events. However, phase III clinical trials of alfimeprase did not meet their primary endpoints and enrollment in ongoing trials has been suspended pending further analyses and discussion with outside experts and regulatory agencies. In spite of this, the authors conclude that alfimeprase seems to be a lytic agent with much potential. Refinement in its use and dosing needs to be addressed, and further investigation into its pharmacokinetic properties may be worthwhile. Alfimeprase is a drug that is a 'work in progress'.


Assuntos
Desenho de Fármacos , Fibrinolíticos/uso terapêutico , Metaloendopeptidases/uso terapêutico , Doenças Vasculares Periféricas/tratamento farmacológico , Terapia Trombolítica , Animais , Avaliação Pré-Clínica de Medicamentos , Fibrinolíticos/efeitos adversos , Fibrinolíticos/síntese química , Fibrinolíticos/farmacocinética , Fibrinolíticos/farmacologia , Humanos , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/síntese química , Metaloendopeptidases/farmacocinética , Metaloendopeptidases/farmacologia , Patentes como Assunto , Plasminogênio , Relação Estrutura-Atividade , Estados Unidos
10.
J Vasc Interv Radiol ; 16(8): 1075-83, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16105919

RESUMO

PURPOSE: To evaluate the safety profile, pharmacokinetics, and thrombolytic activity of alfimeprase, a novel direct-acting thrombolytic agent, in patients with chronic peripheral arterial occlusion (PAO). MATERIALS AND METHODS: In this multicenter, open-label, single-dose, dose-escalation study, 20 patients with worsening symptoms of lower extremity ischemia within 6 months of enrollment were treated with alfimeprase in five escalating dose cohorts (0.025 mg/kg, 0.05 mg/kg, 0.1 mg/kg, 0.3 mg/kg, and 0.5 mg/kg) by means of intraarterial and sometimes intrathrombic pulsed infusion. The primary endpoint was safety assessed by adverse event rates. Additional safety assessments included vital sign monitoring, serum chemistry testing, hematologic testing, and coagulation testing for 28 days after the procedure, as well as alpha2-macroglobulin and antialfimeprase antibody testing for as long as 3 months after treatment. Pharmacokinetic parameters were evaluated with use of an assay that measures free and alpha2-macroglobulin-bound (ie, total) alfimeprase. RESULTS: No patient experienced a hemorrhagic adverse event. Mean plasminogen and fibrinogen concentrations were not substantially altered by treatment. Three transient treatment-related adverse events were reported in the same patient: one incidence each of increased blood fibrinogen level, skin rash, and headache. All three adverse events were graded as mild. The pharmacokinetic profile of alfimeprase suggested that the half-life for total alfimeprase ranges from 11 to 54 minutes (median, 25 min) in patients with PAO. The serum alpha2-macroglobulin concentrations decreased transiently in a dose response-like manner between 12 and 24 hours and returned to within normal limits approximately 14 days after alfimeprase exposure. CONCLUSIONS: Alfimeprase in doses as high as 0.5 mg/kg was generally well-tolerated in patients with chronic PAO. No bleeding complications were noted. The stable fibrinogen concentrations suggest that the activity of alfimeprase may be limited to the target thrombus. Alfimeprase holds the potential to achieve dissolution of thrombus with a diminished risk of hemorrhage.


Assuntos
Fibrinolíticos/administração & dosagem , Perna (Membro)/irrigação sanguínea , Metaloendopeptidases/administração & dosagem , Doenças Vasculares Periféricas/tratamento farmacológico , Terapia Trombolítica , Trombose/tratamento farmacológico , Adulto , Idoso , Angiografia , Fatores de Coagulação Sanguínea/análise , Feminino , Fibrinolíticos/efeitos adversos , Fibrinolíticos/farmacocinética , Hematócrito , Hemoglobinas/análise , Humanos , Masculino , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/farmacocinética , Pessoa de Meia-Idade , Doenças Vasculares Periféricas/sangue , Doenças Vasculares Periféricas/diagnóstico por imagem , Trombose/sangue , alfa-Macroglobulinas/análise
11.
Dermatol Surg ; 31(3): 263-70, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15841624

RESUMO

BACKGROUND: Palmar hyperhidrosis is a problem of unknown etiology that affects patients both socially and professionally. Botulinum toxin type B (Myobloc), approved by the Food and Drug Administration for use in the treatment of cervical dystonia in the United States in December 2000, has subsequently been used effectively in an off-label indication to treat hyperhidrosis. There are sparse data, however, in the literature evaluating the safety and efficacy of BTX-B for the treatment of palmar hyperhidrosis. OBJECTIVE: We evaluated the safety and efficacy of Myobloc in the treatment of bilateral palmar hyperhidrosis. This was a double-blind, randomized, placebo-controlled study to report on the safety and efficacy of Myobloc. METHODS: Twenty participants (10 men, 10 women) diagnosed with palmar hyperhidrosis were injected with either Myobloc (5,000 U per palm) or a 1.0 mL vehicle (100 mM NaCl, 10 mM succinate, and 0.5 mg/mL human albumin) into bilateral palms (15 Myobloc, 5 placebo). The participants were followed until sweating returned to baseline levels. The main outcome measures were safety, efficacy versus placebo, and duration of effect. RESULTS: A significant difference was found in treatment response at day 30, as determined by participant assessments, between 15 participants injected with Myobloc and 3 participants injected with placebo. The duration of action, calculated in the 17 participants who received Myobloc injections and completed the study, ranged from 2.3 to 4.9 months, with a mean duration of 3.8 months. The single most reported adverse event was dry mouth or throat, which was reported by 18 of 20 participants. The adverse event profile also included indigestion or heartburn (60%), excessively dry hands (60%), muscle weakness (60%), and decreased grip strength (50%). CONCLUSION: Myobloc proved to be efficacious for the treatment of palmar hyperhidrosis. Myobloc had a rapid onset, with most participants responding within 1 week. The duration of action ranged from 2.3 to 4.9 months, with a mean of 3.8 months. The adverse event profile included dry mouth, indigestion or heartburn, excessively dry hands, muscle weakness, and decreased grip strength.


Assuntos
Toxinas Botulínicas , Toxinas Botulínicas/uso terapêutico , Hiperidrose/tratamento farmacológico , Metaloendopeptidases/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Toxinas Botulínicas/efeitos adversos , Toxinas Botulínicas Tipo A , Método Duplo-Cego , Feminino , Humanos , Masculino , Metaloendopeptidases/efeitos adversos , Pessoa de Meia-Idade , Projetos Piloto , Qualidade de Vida
12.
J Immunol ; 170(6): 3377-85, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12626598

RESUMO

Rat anti-glomerular basement membrane (GBM) nephritis is a model of crescentic glomerulonephritis induced by injection of anti-GBM antiserum. To elucidate the mechanism of glomerular injury, we analyzed the gene expression patterns in the kidneys of anti-GBM nephritis rats using DNA arrays, and found that macrophage metalloelastase/matrix metalloproteinase (MMP)-12 was one of the highly expressed genes in the kidneys on days 3 and 7 after the injection of anti-GBM antiserum. Enhancement of MMP-12 mRNA expression was confirmed by Northern blot analysis, and in situ hybridization revealed that MMP-12 mRNA was expressed in ED-1-positive macrophages and multinuclear giant cells in the glomeruli with crescent. Moreover, these cells were positive with anti-rat rMMP-12 Ab on the section of the kidneys of anti-GBM nephritis rats on day 7. To clarify the role of MMP-12, we conducted a neutralization experiment using anti-rat rMMP-12 Ab, which had an ability to inhibit rMMP-12 activity of degrading natural substrate such as bovine elastin or human fibronectin in vitro. Anti-rat rMMP-12 Ab or control Ig was injected in each of six rats on days 0, 2, 4, and 6 after the injection of anti-GBM antiserum. Consequently, crescent formation and macrophage infiltration in the glomeruli were significantly reduced in the rats treated with anti-rat rMMP-12 Ab, and the amount of urine protein was also decreased. These results disclosed that MMP-12 played an important role in glomerular injury in a crescentic glomerulonephritis model, and inhibition of MMP-12 may lead to a new therapeutic strategy for this disease.


Assuntos
Doença Antimembrana Basal Glomerular/enzimologia , Doença Antimembrana Basal Glomerular/patologia , Glomérulos Renais/enzimologia , Glomérulos Renais/patologia , Macrófagos/enzimologia , Metaloendopeptidases/efeitos adversos , Animais , Doença Antimembrana Basal Glomerular/imunologia , Doença Antimembrana Basal Glomerular/prevenção & controle , Northern Blotting , Western Blotting , Movimento Celular/imunologia , Regulação da Expressão Gênica/imunologia , Soros Imunes/administração & dosagem , Hibridização In Situ , Injeções Intravenosas , Masculino , Metaloproteinase 12 da Matriz , Metaloendopeptidases/biossíntese , Metaloendopeptidases/genética , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Endogâmicos WKY , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Especificidade por Substrato
13.
Lakartidningen ; 99(42): 4138-9, 4142-8, 2002 Oct 17.
Artigo em Sueco | MEDLINE | ID: mdl-12448292

RESUMO

This review outlines the current status of anti-angiogenic treatment, with emphasis on clinical trials. In pathological growth, vessels become hyperstimulated and dysfunctional, due to overexpression of angiogenic growth factors, such as vascular endothelial growth factor (VEGF). Thus, various anti-angiogenic substances have been developed that neutralize VEGF; others aim to reduce the capacity of the cells to respond to this factor. In addition, substances that are inhibitors of matrix metalloproteinases or agents that induce programmed cell death (apoptosis) of endothelial cells are being tested. Another class of angiogenesis inhibitors includes those that already are in clinical use but on other indications. The National Cancer Institute (NCI) in the USA provides information on on-going clinical trials, which are being conducted on patients suffering from different solid tumor diseases, such as cancer of the colon, lung, prostate and breast. For treatment regimens with anti-angiogenic substances it is important to consider the appropriate dosing and dose interval. The clinical trials have in many instances only recently been initiated and it is premature to predict the outcome, especially as patients in the trials suffer from seriously progressive disease that has previously been treated and found to be therapy-resistant. In many cases combination therapy with an anti-angiogenic substance together with radiation, chemotherapy or other types of conventional tumor treatment, appears promising.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Fatores de Crescimento Endotelial/efeitos adversos , Fatores de Crescimento Endotelial/fisiologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Fatores de Crescimento de Fibroblastos/efeitos adversos , Fatores de Crescimento de Fibroblastos/fisiologia , Humanos , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/fisiologia , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/diagnóstico por imagem , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica/fisiologia , Receptores de Fatores de Crescimento/antagonistas & inibidores , Receptores de Fatores de Crescimento/fisiologia , Tomografia Computadorizada de Emissão
14.
Blood ; 96(4): 1425-32, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10942387

RESUMO

Site directed mutagenesis (350 variants) of recombinant staphylokinase (SakSTAR), a potent fibrin-selective thrombolytic agent, was undertaken in order to reduce its antigenicity while maintaining its potency. Variants with K35A, (ie, Lys[K] in position 35 substituted with Ala[A]), E65D or E65Q, K74R or K74Q, E80A+D82A, K130T, and K135R displayed increased enzymatic activity or reduced binding of human staphylokinase-specific antibodies. Additive mutagenesis identified 8 variants with intact thrombolytic potencies, which absorbed down to less than a third of SakSTAR-specific antibodies. Intra-arterial administration in 61 patients with peripheral arterial occlusion caused no significant allergic reactions. Median neutralizing antibody titers (with 15 to 85 percentiles), expressed as microgram (microg) compound neutralized per milliliter plasma, were 4.4 (0.3 to 49) for the variants, compared with 12 (4 to 100) in 70 patients given wild-type SakSTAR (P =.002 by Mann-Whitney rank sum test). Overt neutralizing antibody induction (more than 5 microg compound neutralized per milliliter plasma) was observed in 57 of 70 patients (81%) given wild-type SakSTAR, but only in 28 of 60 patients (47%) treated with variants (P <.0001 by Fisher exact test). On the basis of this study, the variant SakSTAR (K35A, E65Q, K74R, D82A, S84A, T90A, E99D, T101S, E108A, K109A, K130T, K135R) (code SY155) has been selected for further clinical development. (Blood. 2000;96:1425-1432)


Assuntos
Linfócitos B/imunologia , Epitopos de Linfócito B/imunologia , Fibrinolíticos/imunologia , Metaloendopeptidases/genética , Metaloendopeptidases/imunologia , Apresentação de Antígeno , Fibrinolíticos/efeitos adversos , Humanos , Metaloendopeptidases/efeitos adversos , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Relação Estrutura-Atividade
15.
Thromb Haemost ; 83(5): 666-71, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10823259

RESUMO

Wild-type or equipotent variants of recombinant staphylokinase (rSak) were given intra-arterially (as a 2 mg bolus injection followed by an infusion of 1 mg/h or 0.5 mg/h overnight, with concomitant heparin [1000 IU/h]) to 191 patients of less than 80 years (62 +/- 1 years, mean +/- SEM), with a peripheral arterial occlusion (PAO) of less than 120 days (mean 14 +/- 1 days, median 11 days, 5 to 95 percentiles 3 to 30 days). Ninety nine patients presented with acute or subacute ischemia, 57 with severe claudication, 33 with chronic rest pain and 2 with gangrene. Occlusion occurred in 122 native arteries and in 69 grafts. Revascularization was complete in 83 percent (158/191), partial in 13 percent (24/191) and absent in 4 percent (7/191) after administration of 12 +/- 0.5 mg rSak over 14 +/- 0.7 h. Complete revascularization of acute occlusions of popliteal or more distal arteries was less frequent (60 percent, 15/25) than of acute occlusions of more proximal native arteries (95 percent, 37/39, p <0.001) or grafts (89 percent, 50/56, p = 0.005). Additional endovascular procedures were performed in 47 percent and subsequent elective bypass surgery in 23 percent of patients. Major bleeding occurred in 12 percent (23/191), one month mortality was 3.1 percent (6/191) and one year mortality was 6.9 percent (12/174). However, four patients (2.1 percent) had an intracranial bleeding following therapy: a 85 year old woman with severe diabetic arteriopathy, who was included in violation of the protocol, a 79 and a 74-year-old woman and a 74-year-old man, all with severe hypertension and limb threatening ischemia; these four patients died within two months after treatment. Amputations were performed within the first year in 16 of 162 surviving patients (9.8 percent): in 7 percent (7/96) with an occluded native artery and 14 percent (9/66) with an occluded graft (p = 0.19). No significant difference in lysis rate, one month mortality or one year amputation-free survival was observed in occlusions of recent onset (< or =14 days, n = 126) as compared to occlusions of longer duration (>14 days, n = 65). Treatment was interrupted prematurely in 4 patients because of a suspected allergic reaction. Fibrinogen levels remained unaffected during treatment (3.3 +/- 0.1 g/l before vs. 3.3 +/- 0.1 g/l after infusion, n = 167). In conclusion, rSak appears to be a highly effective thrombolytic agent in patients with PAO, resulting in a low one month mortality (3.1 percent) and a high one year amputation free survival (84 percent), with an acceptable incidence of major bleedings, but with occasional fatal intracranial hemorrhages.


Assuntos
Arteriopatias Oclusivas/tratamento farmacológico , Fibrinolíticos/uso terapêutico , Metaloendopeptidases/uso terapêutico , Doenças Vasculares Periféricas/tratamento farmacológico , Terapia Trombolítica , Idoso , Amputação Cirúrgica/estatística & dados numéricos , Proteínas Sanguíneas/análise , Avaliação de Medicamentos , Embolia/tratamento farmacológico , Feminino , Fibrinolíticos/administração & dosagem , Fibrinolíticos/efeitos adversos , Seguimentos , Oclusão de Enxerto Vascular/tratamento farmacológico , Hemodinâmica , Hemorragia/induzido quimicamente , Hemostasia , Humanos , Injeções Intra-Arteriais , Masculino , Metaloendopeptidases/administração & dosagem , Metaloendopeptidases/efeitos adversos , Pessoa de Meia-Idade , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Fatores de Risco , Taxa de Sobrevida , Terapia Trombolítica/efeitos adversos , Terapia Trombolítica/mortalidade , Trombose/tratamento farmacológico , Resultado do Tratamento , Grau de Desobstrução Vascular
16.
Am Heart J ; 139(5): 820-3, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10783215

RESUMO

BACKGROUND: We undertook an angiographic, dose-finding study of staphylokinase (SAK42D variant) to evaluate its efficacy and safety in patients with acute ST-segment myocardial infarction. METHODS AND RESULTS: Patients were studied within 6 hours of symptom onset and received SAK42D as a 30-minute infusion with 20% of the total dose given as a bolus. Eighty-two patients with a median age of 60 years (interquartile range 52 to 69 years), 84% male and 43% with an anterior myocardial infarction, were studied at a median time from symptom onset of 2.7 hours. There was a high degree of Thrombolysis in Myocardial Infarction (TIMI) 3 flow achieved with 15 mg of SAK42D, that is, 62%. Therefore after 21 patients had been studied at this dose the next dose of 30 mg was used and 65% TIMI 3 patency was achieved. At the peak dose of 45 mg, TIMI 3 90-minute patency was 63%. There were no allergic reactions, and no patient had intracranial hemorrhage. Four patients had major and 9 moderate bleeding during the study; 2 of the major and 5 of the moderate bleeding events occurred within 48 hours of commencement of treatment. The majority (62%) of these were related to vascular instrumentation, and there was no relation between the extent of bleeding and dose of SAK42D used. Forty-five minutes after cessation of SAK42D, there were small percent decrements in plasma fibrinogen and plasminogen levels that did not reach statistical significance. However, there were dose-related changes in alpha(2) anti-plasmin that revealed a borderline significant reduction that was dose related (P =.053). CONCLUSION: These data revealed similar fibrinolytic efficacy across a 3-fold increment in dose, indicating that this study operated on a flat portion of the dose-response curve. The favorable efficacy/safety profile achieved with staphylokinase is encouraging, and further investigation is warranted.


Assuntos
Angiografia Coronária , Fibrinolíticos/administração & dosagem , Metaloendopeptidases/administração & dosagem , Infarto do Miocárdio/tratamento farmacológico , Terapia Trombolítica , Idoso , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Fibrinolíticos/efeitos adversos , Humanos , Infusões Intravenosas , Masculino , Metaloendopeptidases/efeitos adversos , Pessoa de Meia-Idade , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/mortalidade , Taxa de Sobrevida , Resultado do Tratamento
17.
J Clin Oncol ; 18(1): 178-86, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10623708

RESUMO

PURPOSE: To evaluate the feasibility of administering BAY 12-9566, a matrix metalloproteinase (MMP) inhibitor with relative specificity against MMP-2, MMP-3, and MMP-9, on a protracted oral daily dosing schedule in patients with advanced solid malignancies. The study also sought to determine the principal toxicities of BAY 12-9566, whether plasma BAY 12-9566 steady state concentrations (C(ss)) of biologic relevance could be sustained for prolonged periods, and whether BAY 12-9566 affected plasma concentrations of MMP-2, MMP-9, and tissue inhibitor of MMP-2 (TIMP-2). PATIENTS AND METHODS: Patients with solid malignancies were treated with BAY 12-9566 at daily oral doses ranging from 100 to 1,600 mg. BAY 12-9566 dose schedules included 100 mg once daily, 400 mg once daily, 400 mg twice daily, 400 mg three times daily, 400 mg four times daily, and 800 mg twice daily. Plasma was collected to study the range of BAY 12-9566 C(ss) values achieved, and exploratory studies were performed to assess the effects of BAY 12-9566 on plasma concentrations of MMP-2, MMP-9, and TIMP-2. RESULTS: Twenty-one patients were treated with 47 28-day courses of BAY 12-9566. The most common side effects were headache, nausea, vomiting, abnormalities in hepatic functions, and thrombocytopenia, which were rarely clinically significant. BAY 12-9566 was well tolerated on all dose schedules, and there was no consistent dose-limiting toxicity that precluded treatment in the range of dose schedules evaluated. Instead, dose escalation was terminated because BAY 12-9566 plasma C(ss) values increased less than proportionately and plateaued as the daily dose was increased within the dose range of 100 to 1,600 mg/d, suggesting saturable drug absorption. Mean plasma C(ss) values achieved with all dose schedules exceeded BAY 12-9566 concentrations required to inhibit MMPs in vitro and in vascular invasion and tumor proliferation in vivo models. There were no consistent effects of BAY 12-9566 on the plasma concentrations of MMP-2 and MMP-9 over the continuous dosing period at any dose schedule level. However, plasma levels of TIMP-2 seemed to increase in a dose-dependent manner (r(2) =.50, P =.046). CONCLUSIONS: The recommended dose of BAY 12-9566 for subsequent disease directed studies is 800 mg twice daily, which resulted in biologically relevant plasma C(ss) values and an acceptable toxicity profile. Although exploratory studies of MMPs in plasma were not revealing, it is conceivable that some tumor types and disease settings are more likely to produce more readily quantifiable levels of activated MMPs than others. Therefore, attempts to identify and quantify surrogate markers of MMP inhibitory effects should continue to be performed in disease-directed studies in more homogenous patient populations.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Antineoplásicos/administração & dosagem , Metaloendopeptidases/administração & dosagem , Neoplasias/tratamento farmacológico , Compostos Orgânicos , Administração Oral , Adulto , Idoso , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/farmacocinética , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Compostos de Bifenilo , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Metaloproteinase 2 da Matriz/sangue , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Metaloproteinase 9 da Matriz/sangue , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Inibidores de Metaloproteinases de Matriz , Dose Máxima Tolerável , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/farmacocinética , Pessoa de Meia-Idade , Fenilbutiratos , Inibidor Tecidual de Metaloproteinase-2/sangue , Inibidor Tecidual de Metaloproteinase-2/efeitos dos fármacos
18.
J Mal Vasc ; 23(2): 92-8, 1998 Apr.
Artigo em Francês | MEDLINE | ID: mdl-9608920

RESUMO

Staphylokinase which is extracted from Staphylococci has been known for more than 40 years as a profibrinolytic compound. It has been obtained more recently by genetic engineering. Staphylokinase activates plasminogen into plasmin. It is a promising new thrombolytic agent for the treatment of acute myocardial infarction and thromboembolic disease in general. It is a fibrin specific thrombolytic agent which does not decrease plasma fibrinogen level in treated patients. However it is antigenic and the group of Louvain has made much effort to reduce its antigenicity without decreasing its thrombolytic activity. Mutants with a reduced antigenic activity have been recently obtained. These new thrombolytic agents could be superior to currently used drugs. Staphylokinase mutants could have a better benefit/cost ratio than other thrombolytic agents.


Assuntos
Fibrinolíticos/farmacologia , Metaloendopeptidases/farmacologia , Engenharia de Proteínas , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Humanos , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/genética , Dados de Sequência Molecular , Mutação , Embolia Pulmonar/tratamento farmacológico
19.
J Clin Oncol ; 16(6): 2150-6, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9626215

RESUMO

PURPOSE: This phase I study was performed to evaluate the safety and pharmacokinetics of escalating doses of Marimastat (British Biotech, Inc, Oxford, United Kingdom) in patients with advanced malignancies and to determine the phase II recommended dose to be used in subsequent studies. PATIENTS AND METHODS: A standard phase I design was used in this study, in which consecutive groups of three patients were treated with escalating doses of the study drug. Marimastat was administered orally at 25, 50, or 100 mg twice daily to consecutive groups of patients with advanced lung cancer. An additional three patients were added at the highest dose studied (100 mg orally twice daily) to assess whether the inflammatory polyarthitis observed at that dose level can be prevented by a concurrent administration of nonsteroidal antiinflammatory drugs (NSAIDS) and/or low-dose corticosteroids. Blood was drawn for safety monitoring, pharmacokinetic analysis, and plasma levels of metalloproteinase (MMP)-2 and MMP-9 (determined by zymography). A total of 12 patients were studied. RESULTS: The most significant toxicity at the highest dose studied (100 mg orally twice daily) was a symptomatic inflammatory polyarthritis that persisted for up to 8 weeks after discontinuation of the study drug and was dose-limiting. The estimated plasma elimination half-life of Marimastat was 4 to 5 hours. The mean maximum concentration (Cmax) at a reasonably well-tolerated dose (50 mg orally twice daily) was 196 ng/mL and was reached within 1 to 2 hours (Tmax) after administration. Areas under the curve (AUC) tended to correlate with the dose of Marimastat. Zymographic analysis of peripheral-blood ratios of activated proenzymatic forms of MMP-2 and -9 did not show any consistent patterns of change in MMP levels or in a degree of their activation during the course of treatment. CONCLUSION: Marimastat was well absorbed from the gastrointestinal tract, with high levels of the study drug detected in plasma within hours after drug administration. Plasma concentrations of Marimastat achieved at dose levels 2 and 3 (50 mg and 100 mg orally twice daily) were substantially higher than those required for MMP inhibition in vitro. The dose-limiting toxicity (DLT) was severe inflammatory polyarthritis, which seemed to be a cumulative toxicity.


Assuntos
Inibidores Enzimáticos/administração & dosagem , Ácidos Hidroxâmicos/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Metaloendopeptidases/antagonistas & inibidores , Administração Oral , Idoso , Artrite/induzido quimicamente , Colagenases/sangue , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Feminino , Gelatinases/sangue , Humanos , Ácidos Hidroxâmicos/efeitos adversos , Ácidos Hidroxâmicos/farmacocinética , Masculino , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/sangue , Pessoa de Meia-Idade , Resultado do Tratamento
20.
Dev Growth Differ ; 40(2): 221-30, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9572364

RESUMO

Jaspisin, originally isolated from a marine sponge as an inhibitor of the hatching of the sea urchin (Hemicentrotus pulcherrimus) embryo, causes inhibition of sea urchin fertilization. Electron microscopic examination revealed that the acrosome reaction was induced in jaspisin-treated sperm when they were incubated with an intact egg. The acrosome-reacted sperm bound to the vitelline layer by the acrosomal material surrounding the acrosomal process. However, fusion of the acrosomal process and the egg plasma membrane failed to take place. Membrane potential changes were monitored using eggs preloaded with a membrane potential-sensitive fluorochrome, di-8-ANEPPS. Depolarization of the membrane potential, normally observed in the fertilized egg was not observed in the egg inseminated in the presence of jaspisin, indicating the absence of electrical continuity between the jaspisin-treated egg and sperm. Jaspisin inhibited the activities of matrix metallo-endoproteinase members but not of other types of proteinases. These results provide strong, albeit indirect, evidence that a matrix metallo-endoproteinase(s) is involved in the process of gamete fusion during sea urchin fertilization.


Assuntos
Acrossomo/enzimologia , Gelatinases/efeitos adversos , Fusão de Membrana/efeitos dos fármacos , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Ouriços-do-Mar/embriologia , Interações Espermatozoide-Óvulo/efeitos dos fármacos , Estirenos/farmacologia , Acrossomo/efeitos dos fármacos , Animais , Feminino , Masculino , Metaloproteinase 2 da Matriz , Potenciais da Membrana/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA