Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 24067, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34911992

RESUMO

Asunercept (company code APG101 [Apogenix AG]; company code CAN008 [CANbridge Pharmaceuticals]) is a novel glycosylated fusion protein that has shown promising effectiveness in glioblastoma. This Phase I study was initiated to evaluate the tolerability and safety of asunercept in combination with standard radiotherapy and temozolomide (RT/TMZ) in Asian patients with newly diagnosed glioblastoma. This was the Phase I portion of a Phase I/II open label, multicenter trial of asunercept plus standard RT/TMZ. Adults with newly-diagnosed glioblastoma received surgical resection followed by standard RT/TMZ plus asunercept 200 mg/week (Cohort 1) or 400 mg/week (Cohort 2) by 30-min IV infusion. The primary endpoint was the safety and tolerability of asunercept during concurrent asunercept and RT/TMZ; dose-limiting toxicities were observed for each dose. Secondary endpoints included pharmacokinetics (PK) and 6-month progression-free survival (PFS6). All patients (Cohort 1, n = 3; Cohort 2, n = 7) completed ≥ 7 weeks of asunercept treatment. No DLTs were experienced. Only one possibly treatment-related treatment emergent adverse event (TEAE), Grade 1 gingival swelling, was observed. No Grade > 3 TEAEs were reported and no TEAE led to treatment discontinuation. Systemic asunercept exposure increased proportionally with dose and showed low inter-patient variability. The PFS6 rate was 33.3% and 57.1% for patients in Cohort 1 and 2, respectively. Patients in Cohort 2 maintained a PFS rate of 57.1% at Month 12. Adding asunercept to standard RT/TMZ was safe and well tolerated in patients with newly-diagnosed glioblastoma and 400 mg/week resulted in encouraging efficacy.Trial registration NCT02853565, August 3, 2016.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Radioterapia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Povo Asiático , Biomarcadores , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/etiologia , Neoplasias Encefálicas/mortalidade , Terapia Combinada , Gerenciamento Clínico , Monitoramento de Medicamentos , Glioblastoma/diagnóstico , Glioblastoma/etiologia , Glioblastoma/mortalidade , Humanos , Imunoglobulina G/administração & dosagem , Estimativa de Kaplan-Meier , Prognóstico , Proteínas Recombinantes de Fusão/administração & dosagem , Temozolomida/administração & dosagem , Resultado do Tratamento , Receptor fas/administração & dosagem
2.
PLoS One ; 13(7): e0201100, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30044833

RESUMO

Many of the cytokine-based cancer immunotherapies are hindered by the devastating side effects of systemic delivery of the cytokines. To address this problem, we previously described a novel approach to locally achieve high doses of interleukin-12 (IL-12) in tumors and demonstrated that bi-functional fusion protein mIL-12/FasTI expressed by stable clones of TC-1 cells efficiently suppressed tumor proliferation by activating natural killer (NK) cells and other cytolytic killer cells and sending apoptotic signals into tumor cells. In the present study, we employed a lentiviral vector-based gene delivery system to deliver this fusion construct directly into tumor cells. We show that lentiviral vector efficiently delivers the fusion constructs into Hela cells in vitro as assayed by RT-PCR and immunohistochemistry (IHC). We also confirm that fusion protein mIL-12/FasTI delivered by the viral vector significantly enhanced killer cell activation, increased caspase-3 activity and decreased tumor growth in vitro. This study offers a further step for fusion protein cancer therapy for cancer patients.


Assuntos
Terapia Genética/métodos , Imunoterapia/métodos , Interleucinas/administração & dosagem , Neoplasias/terapia , Receptor fas/administração & dosagem , Animais , Apoptose , Técnicas de Cocultura , Vetores Genéticos , Células HEK293 , Células HeLa , Humanos , Interleucinas/genética , Lentivirus/genética , Camundongos , Domínios Proteicos , Receptor fas/genética
3.
Leuk Res ; 68: 62-69, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29549809

RESUMO

In low risk MDS, increased apoptosis of erythroid progenitors mediated via CD95 (Fas) activation has been described to result in peripheral cytopenia. Blockade of the CD95 system can improve erythropoiesis in MDS. Asunercept (APG101) is a fusion protein consisting of the extracellular domain of human CD95 and the Fc domain of human IgG1 blocking the interaction between CD95 and its ligand. Here we report on results from a phase I study in 20 transfusion-dependent low and intermediate risk MDS patients treated with intravenous asunercept (EudraCT 2012-003027-37). Primary objectives were safety and tolerability as well as pharmacodynamic effects. Secondary objectives were hematologic improvement, incidence and time to leukemic progression as well as overall survival. Frequency and severity of adverse events were in range of what could be expected in a patient cohort comprising of elderly MDS patients. Two patients experienced a serious adverse event with a suspected relationship to asunercept. The incidence of disease progression was low. In the 20 patients a decrease of the transfusion need from a mean of 10,8 (±5,1) pRBCs during the 12 weeks treatment phase to a mean of 10,0 (±4,2) pRBCs at the end of the study was observed. In conclusion, asunercept was well tolerated and showed efficacy in transfusion-dependent low and intermediate risk MDS patients. Further clinical investigation is warranted, particularly in combination with erythropoiesis stimulating agents (ESAs).


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Transfusão de Sangue , Imunoglobulina G/efeitos adversos , Imunoglobulina G/uso terapêutico , Síndromes Mielodisplásicas/tratamento farmacológico , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/uso terapêutico , Receptor fas/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Progressão da Doença , Relação Dose-Resposta a Droga , Proteína Ligante Fas/antagonistas & inibidores , Feminino , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/farmacologia , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/patologia , Estudos Prospectivos , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacologia , Risco , Receptor fas/administração & dosagem
4.
Clin Cancer Res ; 20(24): 6304-13, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25338498

RESUMO

PURPOSE: Preclinical data indicate anti-invasive activity of APG101, a CD95 ligand (CD95L)-binding fusion protein, in glioblastoma. EXPERIMENTAL DESIGN: Patients (N = 91) with glioblastoma at first or second progression were randomized 1:2 between second radiotherapy (rRT; 36 Gy; five times 2 Gy per week) or rRT+APG101 (400 mg weekly i.v.). Patient characteristics [N = 84 (26 patients rRT, 58 patients rRT+APG101)] were balanced. RESULTS: Progression-free survival at 6 months (PFS-6) rates were 3.8% [95% confidence interval (CI), 0.1-19.6] for rRT and 20.7% (95% CI, 11.2-33.4) for rRT+APG101 (P = 0.048). Median PFS was 2.5 (95% CI, 2.3-3.8) months and 4.5 (95% CI, 3.7-5.4) months with a hazard ratio (HR) of 0.49 (95% CI, 0.27-0.88; P = 0.0162) adjusted for tumor size. Cox regression analysis adjusted for tumor size revealed a HR of 0.60 (95% CI, 0.36-1.01; P = 0.0559) for rRT+APG101 for death of any cause. Lower methylation levels at CpG2 in the CD95L promoter in the tumor conferred a stronger risk reduction (HR, 0.19; 95% CI, 0.06-0.58) for treatment with APG101, suggesting a potential biomarker. CONCLUSIONS: CD95 pathway inhibition in combination with rRT is an innovative concept with clinical efficacy. It warrants further clinical development. CD95L promoter methylation in the tumor may be developed as a biomarker.


Assuntos
Antineoplásicos/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Imunoglobulina G/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Receptor fas/uso terapêutico , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Biomarcadores , Terapia Combinada , Esquema de Medicação , Feminino , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/efeitos adversos , Masculino , Pessoa de Meia-Idade , Prognóstico , Qualidade de Vida , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/efeitos adversos , Resultado do Tratamento , Carga Tumoral , Adulto Jovem , Receptor fas/administração & dosagem , Receptor fas/efeitos adversos
5.
Clin Exp Immunol ; 137(1): 74-80, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15196246

RESUMO

Dendritic cells (DC) genetically engineered to express Fas (CD95) ligand (FasL-DC) have been proposed as immunotherapeutic tools to induce tolerance to allografts. However, we and others recently showed that FasL-DC elicit a vigorous inflammatory response involving granulocytes and can promote Th1-type CD4+ and cytotoxic CD8+ T lymphocytes. This prompted us to evaluate the pathology induced by intravenous injection of FasL-DC in mice. We observed that FasL-DC obtained after retroviral gene transfer of bone marrow precursors derived from Fas-deficient C57Bl/6 mice induce massive pulmonary inflammation and pleuritis one day after a single intravenous injection in C57Bl/6 mice. Two months later, all mice presented granulomatous vasculitis of small to medium sized vessels, alveolar haemorrhage and pleuritis. In these lesions, apoptotic bodies were found in large number. Anti-neutrophilic cytoplasmic and anti-myeloperoxidase autoantibodies were not detected. This study documents that intravenous injection of FasL-DC causes severe lung granulomatous vasculitis. This new animal model for vasculitis is inducible, highly reproducible and shares many features with human Wegener granulomatosis. This model may be an appropriate tool to further investigate the pathogenesis of vasculitis and test new therapeutic strategies. Moreover, our findings highlight the potential severe complications of FasL-DC-based immunotherapy.


Assuntos
Células Dendríticas/imunologia , Pneumopatias/imunologia , Vasculite/imunologia , Receptor fas/imunologia , Animais , Anticorpos Anticitoplasma de Neutrófilos/análise , Complexo Antígeno-Anticorpo/imunologia , Apoptose/imunologia , Autoanticorpos/análise , Linhagem Celular , Granulócitos/imunologia , Granuloma/imunologia , Granuloma/patologia , Injeções Intravenosas , Ligantes , Pulmão/patologia , Pneumopatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Peroxidase/imunologia , Pleurisia/imunologia , Vasculite/patologia , Receptor fas/administração & dosagem
6.
Cell Res ; 13(6): 465-71, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14728803

RESUMO

Hindlimb unloading (HU) in rodent is a well-accepted ground-based model used to simulate some of the conditions of space flight and reproduce its deleterious effects on the musculoskeletal, cardiovascular and immune systems. In this study, the effects of HU on lymphocyte homeostasis in the spleen and thymus of mice were examined. HU was found to drastically deplete various cell populations in the spleen and thymus. These changes are likely to be mediated by apoptosis, since DNA strand breaks indicative of apoptosis were detected by terminal deoxynucleotidyl transferase-mediated nick end-labeling in both splenocytes and thymocytes. Surprisingly, administration of opioid antagonists or interference with the Fas-FasL interaction was able to block HU-induced reductions of splenocytes, but not thymocytes. On the other hand, steroid receptor antagonists blocked the reduction of lymphocyte numbers in both spleen and thymus. Therefore, the effects of HU on the homeostasis of splenocytes and thymocytes must be exerted through distinct mechanisms.


Assuntos
Elevação dos Membros Posteriores/fisiologia , Linfócitos/imunologia , Linfócitos/metabolismo , Baço/imunologia , Timo/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/metabolismo , Apoptose/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/metabolismo , Injeções Intraperitoneais , Linfócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mifepristona/metabolismo , Naltrexona/metabolismo , Antagonistas de Entorpecentes/metabolismo , Receptores de Esteroides/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Baço/citologia , Baço/fisiologia , Timo/citologia , Timo/fisiologia , Receptor fas/administração & dosagem , Receptor fas/metabolismo
7.
J Immunol ; 158(5): 2303-9, 1997 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9036978

RESUMO

Fas ligand (FasL) is a type II membrane protein. Binding of FasL to its receptor, Fas, induces apoptosis. Matrix metalloproteinase cleaves the membrane-bound human FasL to yield the active soluble form. Here, we have produced a large amount of human soluble rFasL using the yeast, Pichia pastoris. The purified rFasL was found to be glycosylated and to exist as a trimer. The rFasL was effective in inducing apoptosis in a Fas-expressing T cell or a fibroblast cell line. The ID50 of rFasL for mouse Fas-expressing T cells was about 0.5 ng/ml. The killing process with rFasL was quick. That is, >80% Fas-expressing mouse cells were killed within 1 h by a saturation concentration of human rFasL. Intravenous administration of 500 microg of human rFasL had a lethal effect in mice. When the mice were pretreated with Propionibacterium acnes, the subsequent injection of 30 microg of human rFasL induced hepatic failure and killed the mice within 24 h. These results indicated that the soluble human FasL is active in inducing apoptosis in vitro and in vivo, and its deleterious effect may be strengthened in patients who are suffering from bacterial infection.


Assuntos
Glicoproteínas de Membrana/toxicidade , Propionibacterium acnes/imunologia , Proteínas Recombinantes/toxicidade , Receptor fas/toxicidade , Animais , Linhagem Celular , Testes Imunológicos de Citotoxicidade , Proteína Ligante Fas , Feminino , Infecções por Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/mortalidade , Humanos , Injeções Intraperitoneais , Ligantes , Falência Hepática/etiologia , Falência Hepática/imunologia , Falência Hepática/mortalidade , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C3H , Proteínas Recombinantes/biossíntese , Receptor fas/administração & dosagem , Receptor fas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...