Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cell Immunol ; 6(1): 22-50, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38883270

RESUMO

Chimeric antigen receptor (CAR)-T cell therapy has shown potential in improving outcomes for individuals with hematological malignancies. However, achieving long-term full remission for blood cancer remains challenging due to severe life-threatening toxicities such as limited anti-tumor efficacy, antigen escape, trafficking restrictions, and limited tumor invasion. Furthermore, the interactions between CAR-T cells and their host tumor microenvironments have a significant impact on CAR-T function. To overcome these considerable hurdles, fresh methodologies and approaches are needed to produce more powerful CAR-T cells with greater anti-tumor activity and less toxicity. Despite advances in CAR-T research, microbial resistance remains a significant obstacle. In this review, we discuss and describe the basics of CAR-T structures, generations, challenges, and potential risks of infections in CAR-T cell therapy.

2.
Cancer Cell ; 42(5): 780-796.e6, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38518774

RESUMO

Emerging as the most potent and durable combinational immunotherapy, dual anti-PD-1 and CTLA-4 immune checkpoint blockade (ICB) therapy notoriously increases grade 3-5 immune-related adverse events (irAEs) in patients. Accordingly, attempts to improve the antitumor potency of anti-PD-1+CTLA-4 ICB by including additional therapeutics have been largely discouraged due to concerns of further increasing fatal toxicity. Here, we screened ∼3,000 Food and Drug Administration (FDA)-approved drugs and identified clofazimine as a potential third agent to optimize anti-PD-1+CTLA-4 ICB. Remarkably, clofazimine outperforms ICB dose reduction or steroid treatment in reversing lethality of irAEs, but unlike the detrimental effect of steroids on antitumor efficacy, clofazimine potentiates curative responses in anti-PD-1+CTLA-4 ICB. Mechanistically, clofazimine promotes E2F1 activation in CD8+ T cells to overcome resistance and counteracts pathogenic Th17 cells to abolish irAEs. Collectively, clofazimine potentiates the antitumor efficacy of anti-PD-1+CTLA-4 ICB, curbs intractable irAEs, and may fill a desperate clinical need to improve patient survival.


Assuntos
Antígeno CTLA-4 , Clofazimina , Inibidores de Checkpoint Imunológico , Receptor de Morte Celular Programada 1 , Animais , Humanos , Camundongos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Clofazimina/farmacologia , Clofazimina/uso terapêutico , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
3.
Chem Biodivers ; 20(12): e202300399, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37910661

RESUMO

Triple-negative breast cancer (TNBC) is a highly heterogeneous and invasive subtype of breast cancer. The prognosis of TNBC is poor because of its high distant metastasis rate. Triptolide is a type of diterpene trioxide natural compound with potential anti-tumor activities. This study explored the metastatic inhibitory effect of triptolide on MDA-MB-231 cells and its underlying mechanism. Triptolide suppressed cell proliferation and induced cell apoptosis in a time- and dose-dependent manner. Low doses of triptolide (0-8 nM) reduced the migration and invasion capabilities of MDA-MB-231 cells. Triptolide decreased ROCK1, p-Akt, N-cadherin, vimentin and MMP-9 expressions, but increased PTEN and E-cadherin expressions on protein and mRNA levels. Furthermore, the down-regulation of ROCK1 expression in MDA-MB-231 cells after being treated by triptolide could be rescued by ROCK1 specific inhibitor Y27632. Molecular docking showed that triptolide and Y27632 shared the same active center of ROCK1 protein. This article's findings taken together showed that ROCK1 is the primary target of triptolide, which can cause cell apoptosis and inhibit the epithelial-mesenchymal transition of MDA-MB-231 cells.


Assuntos
Diterpenos , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Linhagem Celular Tumoral , Proteínas Proto-Oncogênicas c-akt , Células MDA-MB-231 , Simulação de Acoplamento Molecular , Diterpenos/farmacologia , Proliferação de Células , Movimento Celular , Transição Epitelial-Mesenquimal , Quinases Associadas a rho/farmacologia , Quinases Associadas a rho/uso terapêutico , PTEN Fosfo-Hidrolase/farmacologia , PTEN Fosfo-Hidrolase/uso terapêutico
4.
Chem Biol Drug Des ; 95(6): 552-566, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32166894

RESUMO

Antibody-based immunotherapies play a pivotal role in cancer research with efficient achievements in tumor suppression. Tumor survival is assisted by modulation of immune checkpoints to create imbalances between immune cells and cancer cell's environment. The modulation results in T-cell signal inhibition ultimately inert its proliferation and activation against various tumor cells. PD-L1, a 40 kDa transmembrane protein of B7 family, binds with PD-1 on the membrane of T cells which results in inhibition of T-cell proliferation and activation. PD-L1/PD-1 pathway has generated novel target sites for antibodies that can block PD-L1/PD-1 interactions. The blockage results in T-cell proliferation and tumor cell suppression. The PD-L1 immune checkpoint strategies' development, expression and regulations, signal inhibitions, and developmental stages of PD-L1/PD-1 antibodies are briefly discussed here in this review. All this information will provide a base for new therapeutic development against PD-L1 and PD-1 immune checkpoint interactions and will make available promising treatment options.


Assuntos
Anticorpos Monoclonais/química , Antineoplásicos/química , Inibidores de Checkpoint Imunológico/química , Preparações Farmacêuticas/química , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/metabolismo , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia/métodos , Ligantes , Ativação Linfocitária/efeitos dos fármacos , Receptor de Morte Celular Programada 1/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
5.
Genet Mol Biol ; 42(4): e20180391, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31967634

RESUMO

Antibody-drug conjugates (ADC), precisely deliver a cytotoxic agent to antigen-expressing tumor cells by using specific binding strategies of antibodies. The ADC has shown the ability of potent bio-therapeutics development but indefinite stoichiometric linkage and full-length antibody penetration compromised the field of its advancement. Single chain variable fragments convention instead of the full-length antibody may overcome the challenge of rapid penetration and internalization. Programmed cell death ligand-1 interaction with PD-1 has recently revolutionized the field of immunotherapy. We systematically designed scPDL1-DM1 drug conjugate by linking scFv-PD-L1 proteins (scFv) with maytansinoids (DM1) cytotoxic agent through succinimidyl trans-4-maleimidylmethyl cyclohexane-1- carboxylate (SMCC) linker. Binding affinity was confirmed by immunocytochemistry, spectrophotometry and gel electrophoresis analysis. The scPDL1-DM1 showed specific binding with PD-L1 positive tumor cells and retained in vitro anti-cell proliferation activity. The intracellular trafficking of the drug was evaluated in A549 cancer cell lines, and maximum trafficking was observed after two hours of incubation. The generated drug can be utilized as a potent tool for site-specific conjugation, predicting specificity in vitro activities with extended range against PD-L1 positive cancer cells and can be utilized for further in vivo testing and clinical therapeutics development.

6.
Cytotechnology ; 71(3): 705-722, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31098772

RESUMO

PD-L1 is a 40 kDa trans-membrane protein of B7 family and an important T cell regulator. Binding of PD-L1 and PD-1 inhibits proliferation and activation of T cell results cell exhaustion. This phenomenon can be reversed by blocking PD-L1/PD-1 interactions with single chain variables fragment (scFv) fusion proteins and by direct inhibition of tumor cells with drug conjugates. The human phage-displayed scFv library was utilized to generate scFv against the PD-L1 antigen by affinity bio-panning. The positive clones were selected by continuous transfection of bacterial cells and sequence analysis. The binding affinity and specificity of the scFv and antibody fragments were determined by using surface plasmon resonance biosensor, western blot analysis, and immunofluorescence assay. After three rounds of panning selection, about 30% of clones have a binding affinity with targeted PD-L1 antigen. Eight positive clones with accurate sequences were isolated and analyzed for binding affinity with PD-L1 antigen. Three of those with accurate sequences and binding affinity were selected for the recombinant formation and soluble expression by Escherichia coli host machinery. The highly positive recombinant clones with the exact orientation of FR and CDR domains were developed and can be used as a drug carrier tools in ADC formation or direct inhibition of immune checkpoint in cancer immunotherapy. The conjugate achieved its initial potency and need efficient improvement to enhance direct tumor suppression and bio-therapeutics strategies enrichment.

7.
Prep Biochem Biotechnol ; 48(5): 435-445, 2018 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-29561231

RESUMO

Glypican-3 (GPC3) is an integral membrane proteoglycan, which contains a core protein anchored to the cytoplasmic membrane through a glycosylphosphatidylinositol linkage. The glypican-3 can regulate the signaling pathways, thereby enhances cell division, growth, and apoptosis in certain cell types. It is almost nonexistent on the surface of the human normal cell membrane and highly expresses on the membrane of hepatocellular carcinoma (HCC) cells. It has been well established that GPC3 provides a useful diagnostic marker. For generating the polyclonal antibody of GPC3, we expected that GPC3 N-terminal region (amino acid sequence 26-358) could be expressed in Escherichia coli system, however, no active expression was observed after IPTG induction. Interestingly, after deletion of six proline residues from position 26 to 31 in the N-terminus, expression of recombinant GPC3 was clearly detected. We further analyzed the expressed protein deprived of six prolines, to immunize the New Zealand male rabbits for production of active antibodies. The binding affinity of antibody was analyzed by immunofluorescence analysis, immunohistochemical detection, and western blotting. The functional GPC3 N-terminal protein recombinant development, expression, purification, and the polyclonal antibody have been generated provide the basis for the diagnosis of HCC in cancer therapy.


Assuntos
Anticorpos/análise , Carcinoma Hepatocelular/diagnóstico , Glipicanas/análise , Neoplasias Hepáticas/diagnóstico , Animais , Formação de Anticorpos , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , Clonagem Molecular/métodos , Escherichia coli/genética , Imunofluorescência , Glipicanas/genética , Humanos , Masculino , Domínios Proteicos , Coelhos , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética
8.
Drug Des Devel Ther ; 11: 2265-2276, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28814834

RESUMO

Antibody-drug conjugate (ADC) is a milestone in targeted cancer therapy that comprises of monoclonal antibodies chemically linked to cytotoxic drugs. Internalization of ADC takes place via clathrin-mediated endocytosis, caveolae-mediated endocytosis, and pinocytosis. Conjugation strategies, endocytosis and intracellular trafficking optimization, linkers, and drugs chemistry present a great challenge for researchers to eradicate tumor cells successfully. This inventiveness of endocytosis and intracellular trafficking has given considerable momentum recently to develop specific antibodies and ADCs to treat cancer cells. It is significantly advantageous to emphasize the endocytosis and intracellular trafficking pathways efficiently and to design potent engineered conjugates and biological entities to boost efficient therapies enormously for cancer treatment. Current studies illustrate endocytosis and intracellular trafficking of ADC, protein, and linker strategies in unloading and also concisely evaluate practically applicable ADCs.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Imunoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Humanos , Neoplasias/patologia
9.
Immunol Lett ; 190: 34-41, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28684274

RESUMO

Programmed cell death 1 ligand 1 (PD-L1) is a trans-membrane protein highly expressed on the membrane of cancer cell, which binds inhibitory receptor of PD-1 on the T cells and attenuates anti-tumor immune response.The strategy of blocking PD1 and PD-L1 interaction has been widely used for anti-cancer drug development. The DNA encoding extracellular domain of PD-L1 was cloned and expressed with the pET30(+) and Escherichia coli BL21(DE3) system. Cloning of PD-L1 extracellular domain was confirmed by PCR and enzymatic digestion. Sequence analysis of cloned targeted genes showed 100% homology of original sequence. The recombinant protein was expressed using 1mM/mL IPTG and purified by affinity chromatography on a column of Ni-NTA and confirmed by SDS-PAGE and western blot analysis. Results showed that our constructed pET30(+)/PDL1-ECD system efficiently produces desired recombinant protein with molecular weight of 38.1kDa. The prokaryotic expression system provides an easy method to express PD-L1 extracellular domain that further facilitate the role of PD-1/PD-L1 binding inhibition and helps in valuable drug and antibodies production.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno B7-H1/genética , Escherichia coli/genética , Imunoterapia/métodos , Neoplasias/terapia , Domínios Proteicos/genética , Antígeno B7-H1/isolamento & purificação , Western Blotting , Clonagem Molecular , Expressão Gênica , Vetores Genéticos , Humanos , Neoplasias/imunologia , Reação em Cadeia da Polimerase , Proteínas Recombinantes/genética , Análise de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA