Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Ther ; 32(2): 490-502, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38098228

RESUMO

Inadequate T cell activation has severely limited the success of T cell engager (TCE) therapy, especially in solid tumors. Enhancing T cell activity while maintaining the tumor specificity of TCEs is the key to improving their clinical efficacy. However, currently, there needs to be more effective strategies in clinical practice. Here, we design novel superantigen-fused TCEs that display robust tumor antigen-mediated T cell activation effects. These innovative drugs are not only armed with the powerful T cell activation ability of superantigens but also retain the dependence of TCEs on tumor antigens, realizing the ingenious combination of the advantages of two existing drugs. Superantigen-fused TCEs have been preliminarily proven to have good (>30-fold more potent) and specific (>25-fold more potent) antitumor activity in vitro and in vivo. Surprisingly, they can also induce the activation of T cell chemotaxis signals, which may promote T cell infiltration and further provide an additional guarantee for improving TCE efficacy in solid tumors. Overall, this proof-of-concept provides a potential strategy for improving the clinical efficacy of TCEs.


Assuntos
Neoplasias , Linfócitos T , Humanos , Superantígenos/uso terapêutico , Antígenos de Neoplasias , Morte Celular
2.
Cancers (Basel) ; 15(24)2023 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-38136320

RESUMO

At present, multiple myeloma (MM) is still an essentially incurable hematologic malignancy. Although BCMA-targeted therapies have achieved remarkable results, BCMA levels were found to be downregulated in patients with MM who relapsed after these treatments. Therefore, the search for other antigens specific to MM has become a priority. Independently of BCMA expression, G-protein-coupled receptor family C group 5 member D (GPRC5D) is mainly expressed in the plasma cells of MM patients, while it is expressed in a limited number of normal tissues. Combining MM-specific antigen GPRC5D and T-cell-mediated therapies would be a promising therapeutic strategy for MM. Recently, we constructed a new anti-GPRC5D × anti-CD3 T-cell-engaging bispecific antibody (TCB), BR109, which was capable of binding to human GPRC5D and human CD3ε. Moreover, BR109 was proven to have relatively good stability and antitumor activity. BR109 could specifically trigger T-cell-mediated cytotoxicity against many GPRC5D-positive MM cells in vitro. Meanwhile, antitumor activity was demonstrated in MM cell line xenograft mouse models with human immune cell reconstitution. These preclinical studies have formed a solid foundation for the evaluation of MM treatment efficacy in clinical trials.

3.
Pharmacol Res ; 192: 106781, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37119880

RESUMO

Targeting single tumor antigens makes it difficult to provide sufficient tumor selectivity for T cell engagers (TCEs), leading to undesirable toxicity and even treatment failure, which is particularly serious in solid tumors. Here, we designed novel trispecific TCEs (TriTCEs) to improve the tumor selectivity of TCEs by logic-gated dual tumor-targeting. TriTCE can effectively redirect and activate T cells to kill tumor cells (∼18 pM EC50) by inducing the aggregation of dual tumor antigens, which was ∼70- or 750- fold more effective than the single tumor-targeted isotype controls, respectively. Further in vivo experiments indicated that TriTCE has the ability to accumulate in tumor tissue and can induce circulating T cells to infiltrate into tumor sites. Hence, TriTCE showed a stronger tumor growth inhibition ability and significantly prolonged the survival time of the mice. Finally, we revealed that this concept of logic-gated dual tumor-targeted TriTCE can be applied to target different tumor antigens. Cumulatively, we reported novel dual tumor-targeted TriTCEs that can mediate a robust T cell response by simultaneous recognition of dual tumor antigens at the same cell surface. TriTCEs allow better selective T cell activity on tumor cells, resulting in safer TCE treatment.


Assuntos
Neoplasias , Linfócitos T , Camundongos , Animais , Neoplasias/metabolismo , Antígenos de Neoplasias
4.
Protein Expr Purif ; 189: 105973, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34560256

RESUMO

The novel anti-PD-L1/TGFBR2-ECD fusion protein (BR102) comprises an anti-PD-L1 antibody (HS636) which is fused at the C terminus of the heavy chain to a TGF-ß1 receptor Ⅱ ectodomain (TGFBR2-ECD), and which can sequester the PD-1/PD-L1 pathway and TGF-ß bioactivity in the immunosuppressive tumor microenvironment. In the expression of TGFBR2-ECD wild-type fused protein (BR102-WT), a 50 kDa clipped species was confirmed to be induced by proteolytic cleavage at a "QKS" site located in the N-terminus of the ectodomain, which resulted in the formation of IgG-like clipping. The matrix metalloproteinase-9 was determined to be associated with BR102-WT digestion. In addition, it was observed that the N-glycosylation modifications of the fusion protein were tightly involved in regulating proteolytic activity and the levels of cleavage could be significantly suppressed by MMP-inhibitors. To avoid proteolytic degradation, eliminating protease-sensitive amino acid motifs and introducing potential glycosylation were performed. Three sensitive motifs were mutated, and the levels of clipping were strongly restrained. The mutant candidates exhibited similar binding affinities to hPD-L1 and hTGF-ß1 as well as highly purified BR102-WT2. Furthermore, the mutants displayed more significant proteolytic resistance than that of BR102-WT2 in the lysate incubation reaction and the plasma stability test. Moreover, the bifunctional candidate Mu3 showed an additive antitumor effect in MC38/hPD-L1 bearing models as compared to that of with anti-PD-L1 antibody alone. In conclusion, in this study, the protease-sensitive features of BR102-WT were well characterized and efficient optimization was performed. The candidate BR102-Mutants exhibited advanced druggability in drug stability and displayed desirable antitumor activity.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Neoplasias do Colo/terapia , Processamento de Proteína Pós-Traducional , Receptor do Fator de Crescimento Transformador beta Tipo II/antagonistas & inibidores , Proteínas Recombinantes de Fusão/genética , Animais , Anticorpos Antineoplásicos/genética , Anticorpos Antineoplásicos/imunologia , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Células CHO , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Cricetulus , Feminino , Glicosilação , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Domínios Proteicos , Proteólise , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Receptor do Fator de Crescimento Transformador beta Tipo II/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/imunologia , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biomedicines ; 9(7)2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34356854

RESUMO

The specific recognition of T cell receptors (TCR) and peptides presented by human leukocyte antigens (pHLAs) is the core step for T cell triggering to execute anti-tumor activity. However, TCR assembly and soluble expression are challenging, which precludes the broad use of TCR in tumor therapy. Herein, we used heterodimeric Fc to assist in the correct assembly of TCRs to achieve the stable and soluble expression of several TCRs in mammalian cells, and the soluble TCRs enable us to yield novel bispecific T cell engagers (TCR/aCD3) through pairing them with an anti-CD3 antibody. The NY-ESO-1/LAGE-1 targeted TCR/aCD3 (NY-TCR/aCD3) that we generated can redirect naïve T cells to specific lysis antigen-positive tumor cells, but the potency of the NY-TCR/aCD3 was disappointing. Furthermore, we found that the activation of T cells by NY-TCR/aCD3 was mild and unabiding, and the activity of NY-TCR/aCD3 could be significantly improved when we replaced naïve T cells with pre-activated T cells. Therefore, we employed the robust T cell activation ability of staphylococcal enterotoxin C2 (SEC2) to optimize the activity of NY-TCR/aCD3. Moreover, we found that the secretions of SEC2-activated T cells can promote HLA-I expression and thus increase target levels, which may further contribute to improving the activity of NY-TCR/aCD3. Our study described novel strategies for soluble TCR expression, and the optimization of the generation and potency of TCR/aCD3 provided a representative for us to fully exploit TCRs for the precision targeting of cancers.

6.
Bioconjug Chem ; 31(12): 2767-2778, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33237767

RESUMO

Despite the significant therapeutic advances in T-cell immunotherapy, many malignancies remain unresponsive, which might be because of the negative regulation of T cells by the tumor microenvironment (TME). T cells discriminate tumor cells and normal cells through T-cell receptors (TCRs); therefore, we generated a novel type of TCR-drug conjugates (TDCs) by referring antibody-drug conjugations (ADCs) to overcome the effects of the TME on T cells while preserving the specificity of TCR for tumor recognition. We selected HLA-A2/NY-ESO-1157-165 (peptide NY-ESO-1157-165 in complex with human leukocyte antigen serotype HLA-A*02:01) as the antigen and the antigen-specific TCR (1G4113) as the carrier. By sortase A-mediated ligation, we obtained three NY-TCR-vcMMAEs and further studied their properties, antitumor activity, and toxicity in vitro and in vivo. We found that all the NY-TCR-vcMMAEs had high endocytosis efficiency and specifically killed HLA-A2/NY-ESO-1157-165 positive tumor cells. In xenograft models, one of the TDCs, NY-TCR-2M, was effectively and specifically distributed into tumor tissues and inhibited tumor growth without inducing obvious toxicity. Our results demonstrated that TCRs can be carriers of toxic payloads and that the TDCs thus formed can specifically inhibit tumor growth, neglecting the immune microenvironment.


Assuntos
Antígenos de Neoplasias/imunologia , Antígenos de Superfície/imunologia , Proliferação de Células/efeitos dos fármacos , Imunoconjugados/farmacologia , Espaço Intracelular/efeitos dos fármacos , Proteínas de Membrana/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Humanos , Imunoconjugados/imunologia , Imunoconjugados/metabolismo , Imunoterapia , Espaço Intracelular/metabolismo , Camundongos
7.
Int J Mol Sci ; 20(16)2019 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-31408937

RESUMO

Wilms tumor 1 (WT1) oncoprotein is an intracellular oncogenic transcription factor which is barely expressed in normal adult tissues but over expressed in a variety of leukemias and solid cancers. WT1-derived HLA-A*02:01 T cell epitope, RMFPNAPYL (RMF), is a validated target for T cell-based immunotherapy. We generated two T cell receptor mimic antibody-drug conjugates (TCRm-ADCs), ESK-MMAE, and Q2L-MMAE, against WT1 RMF/HLA-A*02:01 complex with distinct affinities, which mediate specific antitumor activity. Although ESK-MMAE showed higher tumor growth inhibition ratio in vivo, the efficacy of them was not so promising, which might be due to low expression of peptide/HLA targets. Therefore, we explored a bispecific TCRm-ADC that exerted more potent tumor cytotoxicity compared with TCRm-ADCs. Hence, our findings validate the feasibility of the presenting intracellular peptides as the targets of ADCs, which broadens the antigen selection range of antibody-based drugs and provides new strategies for precision medicine in tumor therapy.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Proteínas WT1/imunologia , Tumor de Wilms/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Antígenos HLA-A/imunologia , Humanos , Neoplasias Renais/imunologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptores de Antígenos de Linfócitos T/imunologia , Tumor de Wilms/imunologia
8.
Medicine (Baltimore) ; 98(14): e15088, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30946364

RESUMO

Polymorphisms of organic anion transporting polypeptides (OATPs) have been reported to affect trough serum digoxin concentration (SDC). However, the association of these polymorphisms with trough SDC in Chinese heart failure patients has not been studied. We aim to explore whether OATP1B1 388A>G, OATP1B1 521T>C, and OATP1B3 699G>A influence trough SDC in Chinese heart failure patients and to make clinical recommendations.Chinese patients (n = 104) diagnosed with heart failure under long-term digoxin therapy (0.125 mg daily) were enrolled in this study. Blood samples were collected for the analysis of trough SDC (immunofluorescence) and the polymorphisms of OATP1B1 388A>G, OATP1B1 521T>C, and OATP1B3 699G>A (PCR-RFLP and Sanger sequencing).Patients with glomerular filtration rate (GFR) under 30 mL/min had significantly higher trough SDC (1.20 ±â€Š0.50 ng/mL) than recommended trough SDC for heart failure patients. Trough SDC was not significantly influenced by mutations of OATP1B1 388A>G (P = .890), 521T>C (P = .054), and OATP1B3 699G>A (P = .854). Patients with OATP1B1 521T>C mutant-type carrier had slightly higher trough SDC (0.98 ±â€Š0.53 ng/mL) than those with wild-type carrier (0.74 ±â€Š0.40 ng/mL) when they have repaired renal function.Heart failure patients with severe renal dysfunction (GFR<60 mL/min) and/or OATP1B1 521T>C mutant-type carriers are recommended a smaller dosage of digoxin and strict therapeutic drug monitoring.


Assuntos
Cardiotônicos/sangue , Digoxina/sangue , Taxa de Filtração Glomerular/efeitos dos fármacos , Transportador 1 de Ânion Orgânico Específico do Fígado/genética , Mutação , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/genética , Idoso , Cardiotônicos/administração & dosagem , Cardiotônicos/efeitos adversos , China , Digoxina/administração & dosagem , Digoxina/efeitos adversos , Feminino , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético , Estudos Prospectivos
9.
Pharmacogenomics J ; 18(6): 721-729, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30250148

RESUMO

Numerous studies have illustrated the relationship between SLCO1B1 T521C polymorphism and statin-induced myopathy risk; however, this association is not consistent. Three electronic databases (PubMed, EMBASE, and the Cochrane Library) were searched from inception to October 2017 to identify potential studies. The summary odds ratios (ORs) with 95% confidence intervals (CIs) were calculated from different genetic models by using a random-effects model. Fourteen studies comprising 3265 myopathy patients and 7743 controls were included. The summary ORs suggested that 521CC (OR: 2.31; 95% CI: 1.15-4.63; P = 0.019), 521TC (OR: 1.34; 95% CI: 1.02-1.76; P = 0.034), and 521CC + TC (OR: 1.82; 95% CI: 1.32-2.51; P < 0.001) were associated with a greater risk of statin-induced myopathy than 521TT. The higher incidence of statin-induced myopathy was found to be significantly correlated with the C allele compared with the T allele (OR: 1.89; 95% CI: 1.36-2.62; P < 0.001). In addition, we observed that 521CC + TC was associated with an increased risk of myopathy in individuals who received simvastatin (OR: 2.35; 95% CI: 1.08-5.12; P = 0.032) or rosuvastatin (OR: 1.69; 95% CI: 1.07-2.67; P = 0.024) when compared with 521TT. The 521C allele was associated with a greater risk of cerivastatin-induced myopathy than the T allele (OR: 1.95; 95% CI: 1.47-2.57; P < 0.001). The findings of this study indicated that SLCO1B1 T521C was associated with a significantly higher risk of statin-induced myopathy, especially for simvastatin, rosuvastatin, and cerivastatin. Future studies should be conducted in subjects receiving specific types of drugs, and any potential adverse events need to be explored.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Transportador 1 de Ânion Orgânico Específico do Fígado/genética , Doenças Musculares/induzido quimicamente , Doenças Musculares/genética , Variantes Farmacogenômicos , Polimorfismo Genético , Idoso , Feminino , Frequência do Gene , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Doenças Musculares/diagnóstico , Farmacogenética , Piridinas/efeitos adversos , Medição de Risco , Fatores de Risco , Rosuvastatina Cálcica/efeitos adversos , Sinvastatina/efeitos adversos
10.
Biomaterials ; 178: 158-169, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29933102

RESUMO

Most tumor-associated proteins are located inside tumor cells and thus are not accessible to current marketed therapeutic monoclonal antibodies or their cytotoxic conjugates. Human leukocyte antigen (HLA) class I can present peptides derived from intracellular tumor-associated proteins and somatically mutated proteins on the cell's surface, forming an HLA/peptide complex as tumor-specific antigens for T cell receptor (TCR) recognition. Therefore, HLA-mediated presentation of intracellular tumor antigen peptides provides a viable way to distinguish tumor cells from normal cells, which is important for broadening antigen selection, especially for antibody-drug conjugates (ADCs) regarding their highly cytotoxic payload. We applied sortase A-mediated conjugation to develop TCR-like ADCs (i.e., EA1 HL-vcMMAE) targeting intracellular MART-1 protein, a melanocyte-differentiating antigen specific for metastatic melanomas, via the cell surface HLA-A2/MART-126-35 peptide complex. Homogenous EA1 HL-vcMMAE (drug to antibody ratio of 4) efficiently eliminated melanoma cells in xenograft mouse models with no obvious toxicity at the therapeutic dosage. Trametinib, an MEK inhibitor serving as an HLA expression enhancing agent, augmented the TL-ADCs' efficacy both in vitro and in vivo by upregulating MART-126-35 peptide presentation, thus providing a strategy for overcoming the limitation of antigen presentation level for TL-ADCs. Hence, our findings validate the strategy of using sortase A-generated TL-ADCs to target tumor-specific intracellular proteins, with or without agents present, to increase presenting TCR epitope peptides.


Assuntos
Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Imunoconjugados/metabolismo , Espaço Intracelular/metabolismo , Antígeno MART-1/metabolismo , Melanoma/patologia , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Especificidade de Anticorpos/efeitos dos fármacos , Apresentação de Antígeno/efeitos dos fármacos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Endocitose/efeitos dos fármacos , Feminino , Antígenos de Histocompatibilidade Classe I/metabolismo , Camundongos SCID , Peptídeos/química , Ligação Proteica/efeitos dos fármacos , Piridonas/farmacologia , Pirimidinonas/farmacologia
11.
Eur J Pharm Sci ; 120: 89-95, 2018 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-29727724

RESUMO

Antibody-drug conjugates (ADCs) consist of cytotoxic agents covalently conjugated to monoclonal antibodies that substantially improve antitumour activity and reduce systemic toxicity. With the growing number of ADCs in clinical applications, more accurate bioanalysis data are urgently needed to facilitate the development and rational use of ADCs. Herein, we used antigen-positive cells as antigen carriers and ofatumumab (OFA-HL) and ofatumumab-based ADC (OFA-HL-MMAE) as examples to establish a new ligand-binding assay (LBA) method based on flow cytometry. We proved that the new method met the required analytical performance criteria and the lower limit of quantitation (LOQ) was 0.2 µg/mL. In addition, the LOQ of the quantitative OFA-HL flow cytometry method was reduced to 0.025 µg/mL by choosing an optimized fluorescent antibody, which indicated that the LOQ of the new method can be improved. What's more, the new method showed good stability and specificity when we used it to determine the concentrations of OFA-HL and OFA-HL-MMAE in mouse serum. During the bioanalysis of ADCs, various factors should be considered. Therefore, choosing optimal methods for ADC bioanalysis is necessary. This new method using in situ antigens not only extends the scope of application of the conventional LBA methods by avoiding the need for soluble antigens, but also improves the authenticity of ADC bioanalysis as a supplementary approach, which is valuable for developing accurate ADC assays.


Assuntos
Anticorpos Monoclonais/sangue , Antineoplásicos Imunológicos/sangue , Citometria de Fluxo , Imunoconjugados/sangue , Oligopeptídeos/sangue , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Antineoplásicos Imunológicos/administração & dosagem , Calibragem , Composição de Medicamentos , Citometria de Fluxo/normas , Imunoconjugados/administração & dosagem , Injeções Intravenosas , Limite de Detecção , Camundongos Endogâmicos BALB C , Oligopeptídeos/administração & dosagem , Padrões de Referência , Reprodutibilidade dos Testes , Tecnologia Farmacêutica/métodos
12.
Molecules ; 23(3)2018 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-29495349

RESUMO

As the population ages, neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD) impose a heavy burden on society and families. The pathogeneses of PD and AD are complex. There are no radical cures for the diseases, and existing therapeutic agents for PD and AD have diverse side effects. Tea contains many bioactive components such as polyphenols, theanine, caffeine, and theaflavins. Some investigations of epidemiology have demonstrated that drinking tea can decrease the risk of PD and AD. Tea polyphenols can lower the morbidity of PD and AD by reducing oxidative stress and regulating signaling pathways and metal chelation. Theanine can inhibit the glutamate receptors and regulate the extracellular concentration of glutamine, presenting neuroprotective effects. Additionally, the neuroprotective mechanisms of caffeine and theaflavins may contribute to the ability to antagonize the adenosine receptor A2AR and the antioxidant properties, respectively. Thus, tea bioactive components might be useful for neuronal degeneration treatment in the future. In the present paper, the neuro protection and the mechanisms of tea and its bioactive components are reviewed. Moreover, the potential challenges and future work are also discussed.


Assuntos
Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacologia , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Chá/química , Animais , Antioxidantes/química , Antioxidantes/farmacologia , Biflavonoides/química , Biflavonoides/farmacologia , Cafeína/química , Cafeína/farmacologia , Catequina/química , Catequina/farmacologia , Glutamatos/química , Glutamatos/farmacologia , Interações Ervas-Drogas , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/epidemiologia , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Compostos Fitoquímicos/uso terapêutico , Extratos Vegetais/uso terapêutico , Polifenóis/química , Polifenóis/farmacologia , Transdução de Sinais/efeitos dos fármacos
13.
Int J Mol Sci ; 17(5)2016 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-27187383

RESUMO

Malignant peritoneal mesothelioma (MPM) is an aggressive rare malignancy associated with asbestos exposure. A better understanding of the molecular pathogenesis of MPM will help develop a targeted therapy strategy. Oncogene targeted depth sequencing was performed on a tumor sample and paired peripheral blood DNA from a patient with malignant mesothelioma of the peritoneum. Four somatic base-substitutions in NOTCH2, NSD1, PDE4DIP, and ATP10B and 1 insert frameshift mutation in BAP1 were validated by the Sanger method at the transcriptional level. A 13-amino acids neo-peptide of the truncated Bap1 protein, which was produced as a result of this novel frameshift mutation, was predicted to be presented by this patient's HLA-B protein. The polyclonal antibody of the synthesized 13-mer neo-peptide was produced in rabbits. Western blotting results showed a good antibody-neoantigen specificity, and Immunohistochemistry (IHC) staining with the antibody of the neo-peptide clearly differentiated neoplastic cells from normal cells. A search of the Catalogue of Somatic Mutations in Cancer (COSMIC) database also revealed that 53.2% of mutations in BAP1 were frameshift indels with neo-peptide formation. An identified tumor-specific neo-antigen could be the potential molecular biomarker for personalized diagnosis to precisely subtype rare malignancies such as MPM.


Assuntos
Antígenos de Neoplasias/genética , Biomarcadores Tumorais/genética , Mutação da Fase de Leitura/genética , Neoplasias Pulmonares/genética , Mesotelioma/genética , Neoplasias Peritoneais/genética , Medicina de Precisão , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética , Idoso , Sequência de Aminoácidos , Apresentação de Antígeno/imunologia , Western Blotting , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Complexo Principal de Histocompatibilidade , Mesotelioma Maligno , Modelos Biológicos , Peso Molecular , Reprodutibilidade dos Testes , Transdução de Sinais , Proteínas Supressoras de Tumor/química , Ubiquitina Tiolesterase/química
14.
Sci Rep ; 5: 14872, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26445897

RESUMO

Poor pharmacokinetics and resistance within some tumor cell lines have been the major obstacles during the preclinical or clinical application of TRAIL (tumor-necrosis-factor (TNF)-related apoptosis-inducing ligand). The half-life of TRAIL114-281 (114 to 281 amino acids) was revealed to be no more than 30 minutes across species. Therefore maleimido activated PEG (polyethylene glycol) and MMAE (Monomethyl Auristatin E) were applied to site-specifically conjugate with the mutated cysteines from different monomers of TRAIL successively, taking advantage of steric effects involved within TRAIL mutant conjugations. As a result, TRAIL trimer was hetero-modified for different purposes. And the resulting PEG-TRAIL-vcMMAE conjugate exhibited dramatically improved half-life (11.54 h), favourable in vivo targeting capability and antitumor activities while no sign of toxicity in xenograft models, suggesting it's a viable therapeutic and drug delivery strategy.


Assuntos
Antineoplásicos/química , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Pulmonares/tratamento farmacológico , Oligopeptídeos/química , Polietilenoglicóis/química , Ligante Indutor de Apoptose Relacionado a TNF/química , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Meia-Vida , Humanos , Células K562 , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Células MCF-7 , Masculino , Camundongos , Camundongos Nus , Ratos , Ratos Sprague-Dawley , Ligante Indutor de Apoptose Relacionado a TNF/farmacocinética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Invest New Drugs ; 32(1): 75-86, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23903896

RESUMO

Utilization of antibodies to deliver highly potent cytotoxic agents to corresponding antigen-overexpressed tumor cells is a clinically validated therapeutic strategy. Ofatumumab (OFA, trade name Arzerra) is a fully human CD20-specific antibody that is active against CD20-positive B-cell lymphoma/chronic lymphocytic leukemia cells. In order to further enhance the anticancer effect of OFA, anti-CD20 OFA has been conjugated with highly cytotoxic monomethyl auristatin E (MMAE) through a cathepsin-B-cleavable valine-citrulline (vc) dipeptide linkage to form OFA-vcMMAE and the anti-tumor activity of OFA-vcMMAE against CD20-positive B lymphoma cells are then evaluated in vitro and in vivo. As a result, conjugation of OFA with MMAE has kept the initial effector functional activities of OFA such as binding affinity, complement-dependent cytotoxicity (CDC) as well as antibody-dependent cell-mediated cytotoxicity (ADCC). In addition, the conjugation of MMAE significantly improved the cytotoxic activity of OFA against CD20-positive cells (i.e., Raji, Daudi and WIL2-S cells) but not against CD20-negative K562 cells. On the other hand, OFA-vcMMAE was modulated from the CD20-positive cell surface and then entered the lysosomes by receptor-mediated endocytosis, underwent proteolytic degradation and released active drug MMAE to induce apoptotic cell death through a caspase-3-like protease-dependent pathway. Surprisingly, OFA-vcMMAE completely inhibited the growth of CD20-positive Daudi and Ramos lymphoma xenografts in vivo, and exhibited greater anti-tumor activity than unconjugated OFA, suggesting that the anti-tumor activity of anti-CD20 antibody can be enhanced by conjugation with MMAE. In the near future, this new approach might be used as a clinical treatment of CD20-positive B lymphoid malignancies.


Assuntos
Aminobenzoatos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antígenos CD20/metabolismo , Linfoma de Células B/tratamento farmacológico , Terapia de Alvo Molecular , Oligopeptídeos/uso terapêutico , Aminobenzoatos/química , Aminobenzoatos/farmacologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Endocitose/efeitos dos fármacos , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos SCID , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Biomaterials ; 34(36): 9115-23, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23981355

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a promising antitumor agent that specifically induces apoptosis in broad-spectrum tumor cell lines, meanwhile leaving normal cells unaffected. Unfortunately, the clinical development of TRAIL was hampered, and could be attributed to its instability, bioavailability or poor delivery. Although N-terminal specific PEGylation provides a means to improve the pharmacokinetic and stability of TRAIL, it took a bit longer time to accomplish the PEGylation process than expected. We therefore designed another PEGylation approach, mutated Cys-SH site-specific PEGylation, to conjugate methoxypoly(ethylene glycol) maleimide (mPEG-MAL) with TRAIL (95-281) mutant N109C. Asn-109 was chosen as the PEGylated site for it is a potential N-linked glycosylation site. It was shown that ~90% TRAIL mutant N109C could be PEGylated by mPEG-MAL within 40 min. And mPEG(MAL)-N109C was revealed to possess superior in vitro stability and antitumor activity than N-terminal specifically PEGylated TRAIL (114-281) (mPEG(ALD)-TRAIL(114-281)). What's more, mPEG(MAL)-N109C exhibited more therapeutic potentials than mPEG(ALD)-TRAIL(114-281) in tumor xenograft model, benefitting from better drug delivery and bioavailability. These results have demonstrated mutated Cys-SH specific PEGylation is an alternative to site-specifically PEGylate TRAIL efficiently and effectively other than N-terminal specific PEGylation.


Assuntos
Cisteína/metabolismo , Mutação/genética , Polietilenoglicóis/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/sangue , Peso Corporal/efeitos dos fármacos , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Rim/efeitos dos fármacos , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Polietilenoglicóis/química , Estabilidade Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Ratos , Ratos Sprague-Dawley , Ligante Indutor de Apoptose Relacionado a TNF/química , Ligante Indutor de Apoptose Relacionado a TNF/farmacocinética , Ligante Indutor de Apoptose Relacionado a TNF/toxicidade
17.
Adv Mater ; 25(34): 4718-22, 2013 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-23847045

RESUMO

Monomethyl auristatin E (MMAE) is conjugated with TNF-related apoptosis-inducing ligand (TRAIL) via a linker that is stable in extracellular fluid, while it is cleaved by cathepsin once the conjugate has entered a tumor cell, thus activating the antimitotic mechanism of MMAE. The TRAIL-MMAE conjugate is a conceptually viable therapeutic strategy with improved in vitro antitumor activity, cell circle arrest and specific accumulation in tumor to treat TRAIL-resistant tumors.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Oligopeptídeos/química , Ligante Indutor de Apoptose Relacionado a TNF/química , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Catepsinas/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Humanos , Células MCF-7 , Microscopia Confocal , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/toxicidade , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/toxicidade
18.
Appl Microbiol Biotechnol ; 97(16): 7253-64, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23208613

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered to be a promising anticancer agent because its active form TRAIL trimer is able to induce apoptosis in different tumor cell lines while sparing normal cells. However, TRAIL trimer possesses a short half-life and low stability, which turns out to be a major obstacle for the development of clinical trials. In our present study, we constructed a recombined TRAIL trimer by genetic fusion of non-collagenous domain (NC1) of human collagen XVIII or its trimerization domain (TD) to C-terminus of TRAIL via a flexible linker, and then refolded the fusion proteins using a two-step refolding approach, namely a combination of dilution and gel filtration chromatography. As a result, both recombinant proteins, TRAIL-NC1 and TRAIL-TD, were expressed in Escherichia coli as inclusion bodies, and they exhibited difficultly to refold efficiently by conventional methods. Thereby, we applied a modified two-step refolding approach to refold fusion proteins. More than 55 % of TRAIL-NC1 and 90 % of TRAIL-TD protein activity was recovered during the two-step refolding approach, and their stability was also increased significantly. Also, size exclusion chromatography showed refolded TRAIL-NC1 was a trimer while TRAIL-TD, hexamer. However, both of them exerted good apoptosis activity on NCI-H460 cells.


Assuntos
Colágeno Tipo XVIII/metabolismo , Dobramento de Proteína , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Antineoplásicos/isolamento & purificação , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Cromatografia em Gel , Colágeno Tipo XVIII/genética , Colágeno Tipo XVIII/isolamento & purificação , Colágeno Tipo XVIII/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Escherichia coli/genética , Expressão Gênica , Humanos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/isolamento & purificação , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
19.
Toxicol Sci ; 128(1): 137-46, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22539622

RESUMO

Arsenic is known to be a human carcinogen as well as one of the most effective drugs for treatment of patients with acute promyelocytic leukemia. The intermediate metabolites of arsenic, monomethylarsonous acid (MMA(III)) and dimethylarsinous acid (DMA(III)), are formed by methylation reactions, and they are more reactive and toxic than the inorganic precursor arsenite (iAs(III)); however, the detailed mechanism of toxicity is poorly understood. Here, we studied the effects of three arsenic compounds (i.e., iAs(III), MMA(III), and DMA(III)) on mitochondrial permeability transition pore (mPTP) and release of apoptotic cytochrome c (Cyt c) after incubating with rat liver mitochondria. Inorganic iAs(III) had no effect on mitochondrial swelling even at higher concentrations ranging up to 100 µM, but swelling was significantly induced in the presence of Ca(2+). Additionally, mitochondrial swelling was strongly induced by exposure to the methylated forms of MMA(III) and DMA(III) in a dose-dependent manner in the absence of Ca(2+), suggesting that the methylated forms may have potent effects on cellular mitochondria. Although mitochondrial swelling was completely inhibited in the presence of cyclosporin-A (an inhibitor of mitochondrial permeability transition) or ruthenium red (an inhibitor of Ca(2+) uniporter) following exposure to methylated arsenicals, the release of apoptotic Cyt c from mitochondria was not inhibited, indicating that release of Cyt c is probably not dependent on mPTP opening. In addition, inhibitors of Bax (e.g., Bax-inhibiting peptide) did not reduce the release of Cyt c from the mitochondria by formation of Bax-voltage-dependent anion channel (VDAC) complex, whereas the recombinant Bcl-x(L )proteins significantly reduced the release of Cyt c after exposure to DMA(III), suggesting that dimethylated DMA(III) directly interacted with the VDAC in mitochondria and caused the release of Cyt c from mitochondria.


Assuntos
Apoptose , Ácido Cacodílico/análogos & derivados , Citocromos c/metabolismo , Canais Iônicos/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Animais , Ácido Cacodílico/farmacologia , Linhagem Celular , Mitocôndrias Hepáticas/enzimologia , Ratos
20.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 38(3): 235-41, 2009 05.
Artigo em Chinês | MEDLINE | ID: mdl-19504630

RESUMO

OBJECTIVE: To establish a GC/MS method for analysis of beta-estradiol (beta-E2), bisphenol A (BPA), diethylstilbestrol (DES) and salbutamol (SAL) in human urine. METHODS: Human urine samples were extracted by cleanert PCX and cleanert PEP cartridges; and derivatized after dried completely. beta-E2, BPA, DES and SAL in the extracts were measured by GC/MS method with DB-5MS capillary column and EI ion-source. RESULT: The calibration curves for beta-E2 in samples were linear over the concentration ranges of 1 approximately 300 ng ml(-1), for BPA were 1 approximately 200 ng ml(-1), for DES were 2 approximately 300 ng ml(-1) and for SAL were 0.01 approximately 1.2 microg ml(-1). The limits of detection were 0.15 ng ml(-1), 0.19 ng ml(-1), 0.23 ng ml(-1) and 1.0 ng ml(-1)for beta-E2, BPA, DES and SAL, respectively. The assay recoveries for beta-E2, BPA, DES and SAL ranged from 93.4 % approximately 110.5 %, 84.7 % approximately 104.9 %, 87.0 % approximately 105.4 % and 81.8 % approximately 96.8 %, respectively. CONCLUSION: The established GC/MS method can detect beta-E2, BPA, DES and SAL in urine samples simultaneously, which can be used in routine assessment and monitoring of beta-E2, BPA, DES and SAL in human body.


Assuntos
Albuterol/urina , Dietilestilbestrol/urina , Estradiol/urina , Cromatografia Gasosa-Espectrometria de Massas , Fenóis/urina , Compostos Benzidrílicos , Poluentes Ambientais/urina , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA