Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Expert Opin Biol Ther ; : 1-8, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38752402

RESUMO

BACKGROUND: This study aimed to assess the efficacy and safety of MW031 in Chinese postmenopausal women with osteoporosis. PATIENTS AND METHODS: In this randomized, double-blind, placebo-controlled, multicenter clinical trial, 448 postmenopausal women with osteoporosis were randomized 3:1 to receive MW031 and placebo for 12 months. The primary efficacy endpoint was the percentage change from baseline in BMD at lumbar spine in month 12. The safety and immunogenicity profiles were also included. RESULTS: Of 448 randomized patients, 421 completed the study (MW031, n = 322; placebo, n = 99).After 12 months of MW031 treatment, BMD increased by 5.80% at lumbar spine,3.65% at total hip, and 2.93% at femoral neck. The model-adjusted difference was 3.86% (P<0.0001), 2.34% (P<0.0001), and 1.05% (p = 0.08) compared with placebo group, respectively. For the bone turnover markers, serum CTX level in MW031 group decreased to the maximum difference in month 1 (-71.71%, 95% CI: -77.83%, -65.60%, P<0.0001) compared with the placebo group. The safety analysis showed no significant differences in the proportion of patients reporting any adverse events between the two groups. CONCLUSION: This study demonstrated that MW031 safely and effectively increased BMD and rapidly decreased the level of bone resorption marker in Chinese postmenopausal women with osteoporosis. TRIAL REGISTRATION: NCT05215977 (ClinicalTrials.gov.).

2.
JAMA Oncol ; 10(4): 448-455, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38329745

RESUMO

Importance: The bioequivalence of denosumab biosimilar has yet to be studied in a 53-week, multicenter, large-scale, and head-to-head trial. A clinically effective biosimilar may help increase access to denosumab in patients with solid tumor-related bone metastases. Objectives: To establish the biosimilarity of MW032 to denosumab in patients with solid tumor-related bone metastases based on a large-scale head-to-head study. Design, Setting, and Participants: In this 53-week, randomized, double-blind, phase 3 equivalence trial, patients with solid tumors with bone metastasis were recruited from 46 clinical sites in China. Overall, 856 patients were screened and 708 eligible patients were randomly allocated to receive either MW032 or denosumab. Interventions: Patients were randomly assigned (1:1) to receive MW032 or reference denosumab subcutaneously every 4 weeks until week 49. Main Outcomes and Measures: The primary end point was percentage change from baseline to week 13 of natural logarithmic transformed urinary N-telopeptide/creatinine ratio (uNTx/uCr). Results: Among the 701 evaluable patients (350 in the MW032 group and 351 in the denosumab group), the mean (range) age was 56.1 (22.0-86.0) years and 460 patients were women (65.6%). The mean change of uNTx/uCr from baseline to week 13 was -72.0% (95% CI, -73.5% to -70.4%) in the MW032 group and -72.7% (95% CI, -74.2% to -71.2%) in the denosumab group. These percent changes corresponded to mean logarithmic ratios of -1.27 and -1.30, or a difference of 0.02. The 90% CI for the difference (-0.04 to 0.09) was within the equivalence margin (-0.13 to 0.13); the mean changes of uNTx/uCr and bone-specific alkaline phosphatase (s-BALP) at each time point were also similar during 53 weeks. The differences of uNTx/uCr change were 0.015 (95% CI, -0.06 to 0.09), -0.02 (95% CI, -0.09 to 0.06), -0.05 (95% CI, -0.13 to 0.03) and 0.001 (95% CI, -0.10 to 0.10) at weeks 5, 25, 37, and 53, respectively. The differences of s-BALP change were -0.006 (95% CI, 0.06 to 0.05), 0.00 (95% CI, -0.07 to 0.07), -0.085 (95% CI, -0.18 to 0.01), -0.09 (95% CI, -0.20 to 0.02), and -0.13 (95% CI, -0.27 to 0.004) at weeks 5, 13, 25, 37 and 53, respectively. No significant differences were observed in the incidence of skeletal-related events (-1.4%; 95% CI, -5.8% to 3.0%) or time to first on-study skeletal-related events (unadjusted HR, 0.86; P = .53; multiplicity adjusted HR, 0.87; P = .55) in the 2 groups. Conclusions and Relevance: MW032 and denosumab were biosimilar in efficacy, population pharmacokinetics, and safety profile. Availability of denosumab biosimilars may broaden the access to denosumab and reduce the drug burden for patients with advanced tumors. Trial Registration: ClinicalTrials.gov Identifier: NCT04812509.


Assuntos
Medicamentos Biossimilares , Neoplasias Ósseas , Humanos , Feminino , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Masculino , Denosumab , Anticorpos Monoclonais Humanizados , Neoplasias Ósseas/secundário , Creatinina , Método Duplo-Cego
3.
Expert Opin Biol Ther ; 23(8): 705-715, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36892190

RESUMO

BACKGROUND: MW031 is a biosimilar candidate of denosumab (Prolia®). This study aimed to compare the pharmacokinetics, pharmacodynamics, safety and immunogenicity of MW031 to denosumab in healthy Chinese participants. RESEARCH DESIGN AND METHODS: In this single-center, randomized, double-blind, parallel-controlled, single-dose trial, participants were given 60 mg MW031 (N = 58) or denosumab (N = 61) by subcutaneous injection and observed for 140 days. The primary endpoint was the bioequivalence of PK parameters (Cmax, AUC0-∞), and secondary endpoints including PD parameter, safety, and immunogenicity. RESULTS: A comparison of main PK parameters showed that the geometric mean ratios (GMR) (90% confidence intervals [CIs]) of AUC0-∞ and Cmax for MW031 over denosumab were 105.48% (98.96%, 112.43%) and 98.58% (92.78%, 104.75%), respectively. The inter-CV values of AUC0-∞ and Cmax for MW031 ranged from 19.9% to 23.1%. PD parameter (sCTX) in the MW031 and denosumab groups were similar, and the positivity rates of immunogenicity were 0% in both groups. This study also showed similar safety profiles in both groups, and there were no drug-related, high-incidence and previously unreported adverse reactions. CONCLUSION: This trial confirmed similar pharmacokinetic profiles of MW031 and denosumab in healthy male participants, and pharmacodynamic profile, immunogenicity and safety were comparable for both drugs. TRIAL REGISTRATION: NCT04798313; CTR20201149.


Assuntos
Anticorpos Monoclonais Humanizados , Medicamentos Biossimilares , Denosumab , Humanos , Masculino , Área Sob a Curva , Medicamentos Biossimilares/efeitos adversos , Medicamentos Biossimilares/metabolismo , Medicamentos Biossimilares/farmacocinética , Denosumab/efeitos adversos , Denosumab/imunologia , Denosumab/farmacocinética , Método Duplo-Cego , População do Leste Asiático , Voluntários Saudáveis , Equivalência Terapêutica , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacocinética , Ligante RANK/antagonistas & inibidores , Ligante RANK/imunologia , Injeções Subcutâneas
4.
Int Immunopharmacol ; 107: 108666, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35286913

RESUMO

OBJECTIVE: To compare the pharmacokinetics (PK), pharmacodynamics (PD), safety and immunogenicity between MW032 (denosumab biosimilar) and Xgeva® (denosumab) in healthy Chinese subjects. STUDY DESIGN: In this single-center, randomized, double-blind, single-dose, parallel-controlled design study, 120 healthy male subjects were randomized 1:1 to receive a single dose subcutaneous injection of 120 mg MW032 or Xgeva®, with an observation period of 161 days. The primary endpoint was the bioequivalence of PK parameters (Cmax, AUC0-t), and secondary endpoints including PD parameters, safety, and immunogenicity. RESULTS: One hundred and twelve subjects completed the study, including 56 subjects in each group. The geometric mean ratio and 90% CI for AUC0-t and Cmax were 1.117 (1.034, 1.205) and 1.060 (0.984, 1.142), respectively, which were both within the equivalence interval (0.8, 1.25). The inter-subject variation ranged from 21.37% to 27.37%. The PD parameters between MW032 and Xgeva® were similar. There was no statistically significant difference in the positive incidence of anti-drug antibody test between the two groups. Both MW032 and Xgeva® appeared to be well-tolerated and no Grade 3 or above serious adverse reactions occurred. The adverse reactions observed in the study were reported for denosumab generally. Moreover, there were no high-incidence or previously unreported adverse reactions. CONCLUSION: This study evidenced that the PK profiles of MW032, a denosumab biosimilar, and Xgeva® were bioequivalent. We also found that the PDs, safety, and immunogenicity were similar between the two drugs. Therefore, our results supported the next confirmatory studies for the development of MW032.


Assuntos
Medicamentos Biossimilares , Denosumab , Medicamentos Biossimilares/efeitos adversos , China , Denosumab/efeitos adversos , Denosumab/farmacocinética , Método Duplo-Cego , Voluntários Saudáveis , Humanos , Masculino , Equivalência Terapêutica
5.
Vaccine ; 32(30): 3854-60, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24797700

RESUMO

Dengue is a major infectious disease that affects people living in tropical and subtropical regions around the world. The causative agents are dengue virus serotype 1, 2, 3, and 4 (DENV1, 2, 3, and 4). Developing a vaccine for dengue is a high priority for public health, but traditional methods have faced numerous obstacles due to the unique immunopathogenesis of dengue virus infection. Here, we report a novel dengue vaccine candidate based on dengue pseudoinfectious virus (PIV) produced by the incorporation of a dengue subgenomic replicon into viral particles in highly efficient packaging cells. The subgenomic replicon was constructed by deleting the capsid protein (C) gene from the dengue viral genome and optimizing the signal peptide sequence of pre-membrane protein (prM) to facilitate the formation of viral particles. Packaging cells were developed for inducible expression of a bi-protein Cpr, where the protein pr is the "pr" segment of viral protein prM that holds the protein C on the endoplasmic reticulum (ER). When the replicon was introduced into the packaging cells, protein C was released from the bi-protein Cpr by a replicon-encoded viral protease. Coordinate expression of viral structural proteins by the replicon and packaging cells led to the incorporation of the replicon into viral particle to produce PIVs. Animal tests showed that the dengue PIV vaccine was highly immunogenic and the immune response protected mice challenged with a hundred-fold LD50 inoculation of dengue virus. The method described here has the potential to be applied to vaccine development for other flaviviruses.


Assuntos
Vacinas contra Dengue/biossíntese , Dengue/prevenção & controle , Replicon , Proteínas Estruturais Virais/biossíntese , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Cricetinae , Vacinas contra Dengue/imunologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Deleção de Genes , Camundongos , Dados de Sequência Molecular , Transfecção , Montagem de Vírus
6.
J Oncol ; 2011: 534548, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21209716

RESUMO

Head and neck squamous cell cancer (HNSCC) is one of the top ten cancers in the United States. The survival rate of HNSCC has only marginally improved over the last two decades. In addition, African-American men bear a disproportionate burden of this preventable disease. Therefore, a critical challenge of preventive health approaches is warranted. Salvianolic acid B (Sal-B) isolated from Salvia miltiorrhiza Bge, which is a well-know Chinese medicines has been safely used to treat and prevent aging diseases for thousand of years. Recently, the anticancer properties of Sal-B have received more attention. Sal-B significantly inhibits or delays the growth of HNSCC in both cultured HNSCC cells and HNSCC xenograft animal models. The following anticancer mechanisms have been proposed: the inhibition of COX-2/PGE-2 pathway, the promotion of apoptosis, and the modulation of angiogenesis. In conclusion, Sal-B is a potential HNSCC chemopreventive agent working through antioxidation and anti-inflammation mechanisms.

7.
Cancer Prev Res (Phila) ; 3(6): 787-96, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20501859

RESUMO

Head and neck squamous cell carcinoma (HNSCC) development is closely associated with inflammation. Cyclooxygenase-2 (COX-2) is an important mediator of inflammation. Therefore, celecoxib, a selective inhibitor of COX-2, was hailed as a promising chemopreventive agent for HNSCC. Dose-dependent cardiac toxicity limits long-term use of celecoxib, but it seems likely that this may be diminished by lowering its dose. We found that salvianolic acid B (Sal-B), isolated from Salvia miltiorrhiza Bge, can effectively suppress COX-2 expression and induce apoptosis in a variety of cancer cell lines. In this study, we report that combination of Sal-B with low-dose celecoxib results in a more pronounced anticancer effect in HNSCC than either agent alone. The combination effects were assessed in four HNSCC cell lines (JHU-06, JHU-011, JHU-013, and JHU-022) by evaluating cell viability, proliferation, and tumor xenograft growth. Cell viability and proliferation were significantly inhibited by both the combined and single-agent treatments. However, the combination treatment significantly enhanced anticancer efficacy in JHU-013 and JHU-022 cell lines compared with the single treatment regimens. A half-dose of daily Sal-B (40 mg/kg/d) and celecoxib (2.5 mg/kg/d) significantly inhibited JHU-013 xenograft growth relative to mice treated with a full dose of Sal-B or celecoxib alone. The combination was associated with profound inhibition of COX-2 and enhanced induction of apoptosis. Taken together, these results strongly suggest that combination of Sal-B, a multifunctional anticancer agent, with low-dose celecoxib holds potential as a new preventive strategy in targeting inflammatory-associated tumor development.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Benzofuranos/farmacologia , Carcinoma de Células Escamosas/patologia , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Neoplasias de Cabeça e Pescoço/patologia , Pirazóis/farmacologia , Salvia miltiorrhiza , Sulfonamidas/farmacologia , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzofuranos/administração & dosagem , Celecoxib , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/patologia , Linhagem Celular Tumoral/transplante , Inibidores de Ciclo-Oxigenase 2/administração & dosagem , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/sangue , Sinergismo Farmacológico , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Nus , Pirazóis/administração & dosagem , Sulfonamidas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Ai Zheng ; 28(10): 1114-8, 2009 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-19799824

RESUMO

Vitamin E succinate (RRR-alpha-tocopheryl succinate, VES) is an ester derivative of vitamin E. Roles of vitamin E (alpha-tocopherol) family in cancer prevention and therapy have been investigated since 1960s'. Experimental evidences indicated that VES is one of the most effective anticancer compounds of the vitamin E family. VES can effectively inhibit many kinds of tumors without toxic effects on normal cells and tissues. This article reviewed the anticancer mechanisms of VES in the following four aspects: 1) the molecular structure, chemical property and carrier of VES. 2) the mechanisms of VES in inhibiting cancer cell proliferation. 3) the mechanisms of VES-induced apoptosis of cancer cells. 4) the mechanisms of VES in preventing tumor metastasis. Investigation on the anticancer mechanisms of VES would help find new targets and develop new effective and safe drugs for cancer prevention and treatment.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias/patologia , Tocoferóis/farmacologia , Antineoplásicos/química , Proteínas de Transporte/metabolismo , Ciclo Celular/efeitos dos fármacos , Genes ras , Humanos , Lipoproteínas/metabolismo , Metástase Neoplásica , Neovascularização Patológica , Transdução de Sinais , Tocoferóis/química , Transativadores/metabolismo
9.
Clin Cancer Res ; 14(6): 1840-8, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18347187

RESUMO

PURPOSE: Vitamin E succinate (alpha-TOS) inhibits the growth of cancer cells without unacceptable side effects. Therefore, the mechanisms associated with the anticancer action of alpha-TOS, including ceramide-mediated apoptosis, were investigated using head and neck squamous cell carcinoma (HNSCC) in vitro and in vivo. EXPERIMENTAL DESIGN: Five different human HNSCC cell lines (JHU-011, JHU-013, JHU-019, JHU-022, and JHU-029) were treated with alpha-TOS, and its effects on cell proliferation, cell cycle progression, ceramide-mediated apoptosis, and ceramide metabolism were evaluated. The anticancer effect of alpha-TOS was also examined on JHU-022 solid tumor xenograft growth in immunodeficient mice. RESULTS: Alpha-TOS inhibited the growth of all the HNSCC cell lines in vitro in a dose- and time-dependent manner. Thus, JHU-013 and JHU-022 cell lines were more sensitive to alpha-TOS than the other cell lines. Cellular levels of ceramide, sphingomyelinase activity, caspase-3, and p53 were elevated with increasing time of exposure to alpha-TOS. The degradation of poly(ADP-ribose) polymerase protein in JHU-022 cells treated with alpha-TOS provided evidence for apoptosis. The amounts of nuclear factor kappaB, Bcl-2, and Bcl-X(L) proteins were reduced in the cells treated with alpha-TOS for 6 hours. The levels of caspase-9, murine double minute-2, and IkappaB-alpha proteins were unchanged after alpha-TOS treatment. I.p. administration of alpha-TOS slowed tumor growth in immunodeficient mice. CONCLUSIONS: Alpha-TOS showed promising anticancer effects to inhibit HNSCC growth and viability in vivo and in vitro. The induction of enzymes involved in ceramide metabolism by alpha-TOS suggests that ceramide-mediated apoptosis may expand therapeutic strategies in the treatment of carcinoma.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Ceramidas/farmacologia , Neoplasias de Cabeça e Pescoço/patologia , Vitamina E/análogos & derivados , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ceramidas/metabolismo , Feminino , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Camundongos , Camundongos Nus , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Tocoferóis , Células Tumorais Cultivadas , Vitamina E/farmacologia , Vitamina E/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Sheng Wu Gong Cheng Xue Bao ; 22(3): 492-8, 2006 May.
Artigo em Chinês | MEDLINE | ID: mdl-16755933

RESUMO

The Yellow Fever (YF) vaccine, an attenuated yellow fever 17D (YF-17D) live vaccine, is one of the most effective and safest vaccines in the world and is regarded as one of the best candidates for viral expression vector. We here first reported in China the construction and characterization of the recombinant expression vector of yellow fever 17D which contained the proteinase 2A fragment of foot-and-mouth disease virus (FMDV). Three cDNA fragments representing the full-length YF-17D genome, named 5'-end cDNA (A), 3'-end cDNA (B) and middle cDNA (C), were obtained by reverse transcription polymerase chain reaction (RT-PCR), together with the introduction of SP6 enhancer, necessary restriction sites and overlaps for homologous recombination in yeast. Fragment A and B were then introduced into pRS424 in turn by DNA recombination, followed by transfection of fragment C and the recombinant pRS424 containing A and B (pRS-A-B) into yeast. A recombinant vector containing full length cDNA of YF-17D (pRS-YF) was obtained by screening on medium lack of tryptophan and uracil. A recombinant YF-17D expression vector containing FMDV-2A gene fragment (pRS-YF-2A1) was then constructed by methods of DNA recombination and homologous recombination in yeast described above. In vitro transcription of the recombinant vector pRS-YF-2A1 was then carried out and introduced into BHK-21 cells by electroporation. Results of indirect immunofluorescence assay (IFA) and titer determination showed a stable infectious recombinant virus was gotten, whose features such as growth curve were similar to those of the parental YF-17D. The results suggest that the recombinant vector pRS-YF-2A1, by introduction of heterogenous genes via 2A region, is potential to be an effective live vaccine expression vector.


Assuntos
Vírus da Febre Aftosa/genética , Vetores Genéticos , Vacinas Virais/genética , Animais , Linhagem Celular , Clonagem Molecular , Cricetinae , Epitopos/imunologia , Febre Aftosa/prevenção & controle , Vírus da Febre Aftosa/imunologia , Engenharia Genética , Recombinação Genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Vacinas Atenuadas , Vacinas Virais/imunologia , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...