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.
Front Oncol ; 13: 1211759, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37576888

RESUMO

OX40 (CD134), a member of the TNF receptor superfamily, is a widely studied costimulatory immune checkpoint. Several OX40 agonistic antibodies are in the clinical stage for cancer treatment, among which PF-04518600 is the leader and currently in phase II trial. It has been recognized that one potential mode of action for anti-OX40 antibodies is the deletion of intratumoral Tregs. Thus, a novel human anti-OX40 antibody, BAT6026, was generated with enhanced antibody dependent cellular cytotoxicity (ADCC) via fucose deletion to strengthen its Treg depletion activity. This characteristic of BAT6026 differentiates it from other previously reported anti-OX40 antibodies in the field of tumor therapy. The affinity of BT6026 to OX40 was 0.28nM, approximately 8 times stronger than that of PF-04518600. BAT6026 effectively competed for the binding of ligand OX40L to OX40, whereas PF-04518600 only partially competed. Moreover, compared to PF-04518600, BAT6026 activated T cells more effectively when clustered by FcγRs engagement and stimulated SEB-pretreated PBMCs to secrete IL-2 cytokines in vitro. In addition, BAT6026 demonstrated stronger anti-tumor activity than PF-04518600 in an OX40-humanized mouse MC38 tumor model. BAT6026 also showed a significantly synergistic effect on tumor inhibition when combined treatment with PD-1 antibody. Analysis of tumor-infiltrating T cells revealed that BAT6026 treatment significantly reduced Treg cells and increased CD8+ T cells in tumor. Preclinical safety assessment in non-human primates demonstrated a good safety profile for BAT6026. Together these data warrant further development of BAT6026 into clinical trials for patients with cancer.

2.
Drugs R D ; 23(3): 267-288, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37479945

RESUMO

BACKGROUND: BAT1706 is a proposed biosimilar of bevacizumab, a vascular endothelial growth factor A (VEGF-A)-targeting biologic used to treat several different cancers, including metastatic colorectal cancer. A comprehensive physicochemical and functional similarity assessment is a key component of demonstrating biosimilarity between a reference biologic and a proposed biosimilar. Here we report the physicochemical and functional similarity of BAT1706 and reference bevacizumab sourced from both the United States (US-bevacizumab) and the European Union (EU-bevacizumab). METHOD: A large range of product attributes, including primary and higher order structure, post-translational modifications, purity, stability, and potency, were characterized for BAT1706 and EU/US-bevacizumab using sensitive state-of-the-art analytical techniques. Up to 18 lots of US- and 29 lots of EU-bevacizumab, and 10 unique drug substance lots of BAT1706, were assessed. RESULT: BAT1706 was shown to have an identical amino acid sequence and an indistinguishable higher-order structure compared with EU/US-bevacizumab. BAT1706 and EU/US-bevacizumab also exhibited similar post-translational modifications, glycan profiles, and charge variants. Potency, assessed using a wide range of bioassays, was also shown to be comparable between BAT1706 and EU/US-bevacizumab, with statistical equivalence demonstrated for VEGF-A binding and neutralizing activity. CONCLUSION: Overall, this extensive comparability exercise demonstrated BAT1706 to match EU/US-bevacizumab in terms of all physicochemical and functional attributes assessed.


Assuntos
Medicamentos Biossimilares , Fator A de Crescimento do Endotélio Vascular , Humanos , Bevacizumab/farmacologia , Medicamentos Biossimilares/farmacologia , Bioensaio , Fosforilação
3.
Cancer Commun (Lond) ; 41(2): 171-182, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33528890

RESUMO

BACKGROUND: The introductions of anti- human epidermal growth factor receptor-2 (HER2) agents have significantly improved the treatment outcome of patients with HER2-positive breast cancer. BAT8001 is a novel antibody-drug conjugate targeting human epidermal growth factor receptor-2 (HER2)-expressing cells composed of a trastuzumab biosimilar linked to the drug-linker Batansine. This dose-escalation, phase I study was designed to assess the safety, tolerability, pharmacokinetics, and preliminary anti-tumor activity of BAT8001 in patients with HER2-positive locally advanced or metastatic breast cancer. METHODS: This trial was conducted in subjects with histologically confirmed HER2-positive breast cancer (having evaluable lesions and an Eastern Cooperative Oncology Group performance status of 0 or 1) using a 3 + 3 design of escalating BAT8001 doses. Patients received BAT8001 intravenously in a 21-day cycle, with dose escalation in 5 cohorts: 1.2, 2.4, 3.6, 4.8, and 6.0 mg/kg. The primary objective was to evaluate the safety and tolerability of BAT8001. Preliminary activity of BAT8001 was also assessed as a secondary objective. RESULTS: Between March 2017 to May 2018, 29 HER2-positive breast cancer patients were enrolled. The observed dose-limiting toxicities were grade 4 thrombocytopenia and grade 3 elevated transaminase. The maximum tolerated dose was determined to be 3.6 mg/kg. Grade 3 or greater adverse events (AEs) occurred in 14 (48.3%) of 29 patients, including thrombocytopenia in 12 (41.4%) patients, aspartate aminotransferase increased in 4 (13.8%) patients, γ-glutamyl transferase increased in 2 (6.9%) patients, alanine aminotransferase increased in 2 (6.9%) patients, diarrhea in 2 (6.9%) patients. Objective response was observed in 12 (41.4%; 95% confidence interval [CI] = 23.5%-61.1%) and disease control (including patients achieving objective response and stable disease) was observed in 24 (82.8%; 95% CI = 64.2%-94.2%) patients. CONCLUSIONS: BAT8001 demonstrated favorable safety profiles, with promising anti-tumor activity in patients with HER2-positive locally advanced or metastatic breast cancer. BAT8001 has the potential to provide a new therapeutic option in patients with metastatic HER2-positive breast cancer.


Assuntos
Neoplasias da Mama , Imunoconjugados , Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Dose Máxima Tolerável , Trastuzumab/uso terapêutico
4.
Expert Opin Investig Drugs ; 30(1): 77-83, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33241960

RESUMO

Background: To compare the pharmacokinetic (PK) profile, safety, and immunogenicity between golimumab and the biosimilar BAT2506 in healthy Chinese male subjects. Research design and methods: A total of 180 healthy male subjects were recruited for this randomized, double-blinded, single-dose, parallel study. They received 50 mg BAT2506 or golimumab (1:1 ratio) by single subcutaneous injection. The evaluation index included maximum plasma concentration (Cmax), area under the plasma concentration-time curve (AUC0-t, AUC0-∞), safety, and immunogenicity. Results: The results showed that the 90% confidence interval (CI) of the geometric mean ratio (GMR) of BAT2506 to reference drug (golimumab) for Cmax, AUC0-∞ and AUC0-t were 99.26% (90.59-108.76%), 102.06% (93.31%-111.64%), and 102.05% (93.51-111.38%), respectively. All 90% CIs were within the range of 80-125% range, which is the limitation of the equivalence margin. Furthermore, similarity of treatment-emergent adverse events was also found between the two drugs. A total of 14 subjects (7.8%) developed anti-drug antibody after administration. Conclusions: Our study confirmed the PK similarity between BAT2506 and golimumab, and showed good tolerance of BAT2506 in healthy subjects. There were no differences in safety and immunogenicity between the two drugs. Therefore, BAT2506 meets the criteria for biosimilarity to golimumab. Trial registration: The trial is registered at ClinicalTrials.gov (CT.gov identifier: NCT04152759).


Assuntos
Anticorpos Monoclonais/administração & dosagem , Medicamentos Biossimilares/administração & dosagem , Inibidores do Fator de Necrose Tumoral/administração & dosagem , Adulto , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Área Sob a Curva , Povo Asiático , Medicamentos Biossimilares/efeitos adversos , Medicamentos Biossimilares/farmacocinética , Método Duplo-Cego , Humanos , Injeções Subcutâneas , Masculino , Equivalência Terapêutica , Inibidores do Fator de Necrose Tumoral/efeitos adversos , Inibidores do Fator de Necrose Tumoral/farmacocinética
5.
J Oral Maxillofac Surg ; 77(8): 1575.e1-1575.e6, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31102578

RESUMO

PURPOSE: The present study has proposed a novel orthodontic extraction method with a removable appliance to avoid inferior alveolar nerve (IAN) injury during impacted mandibular third molar removal. PATIENTS AND METHODS: In the present study, 16 patients were enrolled and divided into 2 groups per patient choice. In the orthodontic extraction group (n = 8), a removable appliance was first applied to move the root tips away from the IAN, and the tooth was subsequently removed. In the traditional extraction group (n = 8), each patient had the tooth removed immediately by the same surgeon. RESULTS: All teeth were extracted successfully. All 8 patients in the orthodontic extraction group had had their impacted mandibular third molar removed without IAN injury after surgery. In contrast, 4 patients in the traditional extraction group had experienced transient IAN injury, and the symptoms persisted for 2 to 8 weeks. None of the patients experienced permanent IAN damage. CONCLUSIONS: Orthodontic extraction with a removable appliance to separate the IAN and impacted mandibular third molar could be a good alternative treatment option to avoid IAN injury in high-risk cases.


Assuntos
Dente Serotino , Extração Dentária , Dente Impactado , Traumatismos do Nervo Trigêmeo , Humanos , Mandíbula , Nervo Mandibular , Extração Dentária/efeitos adversos , Dente Impactado/cirurgia , Traumatismos do Nervo Trigêmeo/etiologia
6.
Oral Dis ; 25(4): 1076-1083, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30746817

RESUMO

OBJECTIVES: Salivary adenoid cystic carcinoma (SACC) is one of the most common malignant salivary gland tumors. Our study aims to investigate whether hypoxia-induced autophagy was up-regulated in the progression of SACC. MATERIALS AND METHODS: We performed differentially expressed gene analysis and pathway enrichment analysis and then calculated the correlation analysis on GSE59701 and GSE28996 datasets. The expression of HIF-1ɑ and MAP1LC3B was analyzed in the paraffin-embedded specimens by immunohistochemical method and in the hypoxic SACC-LM cells by immunofluorescence. TEM microscopy was also performed to observe the formation of autophagosomes in SACC tissue and the hypoxic SACC-LM cells. RESULTS: The autophagy pathway was up-regulated in SACC datasets, and five genes including MAP1LC3B were positively correlated with HIF-1ɑ. Immunohistochemistry results showed that autophagy was activated and the expression of HIF-1ɑ and MAP1LC3B was positively correlated in SACC specimens. In hypoxic SACC-LM cells, we also identified the up-regulation of autophagy and the close correlation between HIF-1ɑ and MAP1LC3B expression. Autophagosomes were observed both in the tissue and the hypoxic SACC-LM cells by TEM microscopy. CONCLUSIONS: Our study showed that autophagy is up-regulated in dataset, SACC tissue, and hypoxic cell line; hypoxia-induced autophagy in SACC might play a vital role in the development of SACC.


Assuntos
Autofagia/genética , Carcinoma Adenoide Cístico , Regulação Neoplásica da Expressão Gênica , Neoplasias das Glândulas Salivares , Linhagem Celular Tumoral , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Imuno-Histoquímica
7.
Artigo em Inglês | MEDLINE | ID: mdl-21439861

RESUMO

OBJECTIVES: Fibrous dysplasia (FD) is a focal bone lesion composed primarily of immature bone marrow stromal cells along with spicules of immature woven bone. However, cellular differentiation and proliferation in mutant human bone marrow stromal cells (hBMSCs) of FD have not been fully elucidated. Therefore, the aim of this study was to investigate the occurrence of G(s)α mutation at the Arg(201) codon in hBMSCs and human trabecular bone cells (hTBCs, osteoblast-like cells). In addition, we evaluated the gene expression and protein secretion of amphiregulin from hBMSCs and hTBCs and assessed the biologic activity and possible mechanism of amphiregulin in our system. STUDY DESIGN: Mutant hBMSCs from FD patients and hTBCs from disease-free bone specimens of the same patient were successfully cultured. We studied the G(s)α mutations at the Arg(201) codon by means of polymerase chain reaction (PCR)-restriction fragment length polymorphism. Gene expression and protein secretion of amphiregulin in hBMSCs and hTBCs was confirmed by reverse-transcription (RT) PCR and Western blotting analysis. The modulation proliferation and possible mechanism of the exogenous addition of amphiregulin and epidermal growth factor receptor tyrosine kinase inhibitor (AG-1478) were assessed by using Wst-1 assays. RESULTS: The G(s)α mutations in clonal adherent mutant hBMSCs and hTBCs were successfully identified. We identified amphiregulin to be highly expressed in hBMSCs compared with hTBCs. The growth of hBMSCs was stimulated by the exogenous addition of amphiregulin and inhibited by AG-1478. CONCLUSIONS: The G(s)α-mutant hBMSCs were successfully identified, and amphiregulin may play a significant role in the proliferation of hBMSCs.


Assuntos
Células da Medula Óssea/metabolismo , Receptores ErbB/metabolismo , Displasia Fibrosa Óssea/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Doenças Mandibulares/genética , Adolescente , Substituição de Aminoácidos , Anfirregulina , Arginina/genética , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Corantes , Análise Mutacional de DNA , Família de Proteínas EGF , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Feminino , Displasia Fibrosa Óssea/metabolismo , Glicoproteínas/biossíntese , Glicoproteínas/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Mutação de Sentido Incorreto , Osteoblastos/citologia , Osteoblastos/metabolismo , Polimorfismo de Fragmento de Restrição , Quinazolinas , Transdução de Sinais , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Sais de Tetrazólio , Tirfostinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA