Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 67(7): 5744-5757, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38553427

RESUMO

To develop a next-generation metal agent and dual-agent multitargeted combination therapy, we developed a copper (Cu) compound based on the properties of the human serum albumin (HSA)-indomethacin (IND) complex to remodel the tumor microenvironment (TME). We optimized a series of Cu(II) isopropyl 2-pyridyl ketone thiosemicarbazone compounds to obtain a Cu(II) compound (C4) with significant cytotoxicity and then constructed an HSA-IND-C4 complex (HSA-IND-C4) delivery system. IND and C4 bind to the hydrophobic cavities of the IB and IIA domains of HSA, respectively. In vivo, the HSA-IND-C4 not only showed enhanced antitumor efficacy relative to C4 and C4 + IND but also improved their targeting ability and decreased their side effects. The antitumor mechanism of C4 + IND involved acting on the different components of the TME. IND inhibited tumor-related inflammation, while C4 not only induced apoptosis and autophagy of cancer cells but also inhibited tumor angiogenesis.


Assuntos
Antineoplásicos , Neoplasias , Pró-Fármacos , Tiossemicarbazonas , Humanos , Albumina Sérica Humana/química , Cobre/química , Albumina Sérica/química , Tiossemicarbazonas/farmacologia , Tiossemicarbazonas/uso terapêutico , Indometacina/uso terapêutico , Microambiente Tumoral , Pró-Fármacos/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Neoplasias/tratamento farmacológico
2.
J Med Chem ; 67(5): 3843-3859, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38442035

RESUMO

To develop a potential theranostic metal agent to reverse the resistance of cancer cells to cisplatin and effectively inhibit tumor growth and metastasis, we proposed to design a cyclometalated iridium (Ir) complex based on the properties of the tumor environment (TME). To the end, we designed and synthesized a series of Ir(III) 2-hydroxy-1-naphthaldehyde thiosemicarbazone complexes by modifying the hydrogen atom(s) of the N-3 position of 2-hydroxy-1-naphthaldehyde thiosemicarbazone compounds and the structure of cyclometalated Ir(III) dimers and then investigated their structure-activity and structure-fluorescence relationships to obtain an Ir(III) complex (Ir5) with remarkable fluorescence and cytotoxicity to cancer cells. Ir5 not only possesses mitochondria-targeted properties but also overcomes cisplatin resistance and effectively inhibits tumor growth and metastasis in vivo. Besides, we confirmed the anticancer mechanisms of Ir5 acting on different components in the TME: directly killing liver cancer cells by inducing necroptosis and activating the necroptosis-related immune response.


Assuntos
Antineoplásicos , Complexos de Coordenação , Naftalenos , Neoplasias , Tiossemicarbazonas , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Antineoplásicos/química , Irídio/farmacologia , Irídio/química , Medicina de Precisão , Necroptose , Neoplasias/tratamento farmacológico , Mitocôndrias , Complexos de Coordenação/química , Linhagem Celular Tumoral
3.
J Inorg Biochem ; 250: 112403, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37866112

RESUMO

To develop the next-generation metal agents for efficiently inhibiting tumor growth, a series of novel mononuclear, binuclear and trinuclear copper (Cu) thiophene-2-formaldehyde thiosemicarbazone complexes and a tetranuclear Cu 1,2,4-triazole-derived complex have been synthesized and their structure-activity relationships have been studied. The trinucleated Cu complex showed the strongest inhibitory activity against T24 cells among all the Cu complexes. Its antitumor effect in vivo was superior to that of cisplatin, with reduced side effects. Further studies on the antitumor mechanism have showed that Cu complexes not only induced apoptosis of cancer cells but also inhibited tumor angiogenesis by inhibiting the migration and invasion of vascular endothelial cells, blocking the cell cycle in the G1 phase, and inducing autophagy.


Assuntos
Antineoplásicos , Complexos de Coordenação , Neoplasias , Humanos , Antineoplásicos/farmacologia , Cobre/farmacologia , Modelos Moleculares , Células Endoteliais , Neoplasias/tratamento farmacológico , Apoptose , Complexos de Coordenação/farmacologia , Linhagem Celular Tumoral , Proliferação de Células
4.
J Med Chem ; 66(22): 15424-15436, 2023 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-37956097

RESUMO

For the integration of targeted diagnosis and treatment of tumor, we innovatively designed and synthesized a single-molecule hetero-multinuclear Er(III)-Cu(II) complex (ErCu2) and then constructed an ErCu2@apoferritin (AFt) nanoparticle (NP) delivery system. ErCu2 and ErCu2@AFt NPs not only provided an evident photoacoustic imaging (PAI) signal of the tumor but also effectively inhibited tumor growth by integrating photothermal therapy, chemotherapy, and immunotherapy. ErCu2@AFt NPs improved the targeting ability and decreased the systemic toxicity of ErCu2 in vivo. Furthermore, we confirmed that ErCu2 and ErCu2@AFt NPs inhibited tumor growth by inducing apoptosis and autophagy of tumor cells and activating the immune system. The study not only provides a novel strategy to develop therapeutic metal agents but also reveals their potential for targeted accurate diagnosis and multimodality therapy of cancer.


Assuntos
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Humanos , Cobre/farmacologia , Apoferritinas , Érbio/uso terapêutico , Técnicas Fotoacústicas/métodos , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Nanopartículas/uso terapêutico , Linhagem Celular Tumoral
5.
J Med Chem ; 66(18): 13072-13085, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37702429

RESUMO

To develop next-generation metal drugs with high efficiency and low toxicity for targeting inhibition of gastric tumor growth and metastasis, we not only optimized a series of ruthenium (Ru, III) 2-hydroxy-1-naphthaldehyde thiosemicarbazone complexes to obtain a Ru(III) complex (4b) with remarkable cytotoxicity in vitro but also constructed a 4b-decitabine (DCT)/liposome (Lip) delivery system (4b-DCT-Lip). The in vivo results showed that 4b-DCT-Lip not only had a stronger capacity to inhibit gastric tumor growth and metastasis than 4b-DCT but also addressed the co-delivery problems of 4b-DCT and improved their targeting ability. Furthermore, we confirmed the mechanism of 4b-DCT/4b-DCT-Lip inhibiting the growth and metastasis of a gastric tumor. DCT-upregulated gasdermin E (GSDME) was cleaved by 4b-activated caspase-3 to afford GSDME-N terminal and then was aggregated to form nonselective pores on the cell membrane of a gastric tumor, thereby inducing pyroptosis and a pyroptosis-induced immune response.


Assuntos
Rutênio , Neoplasias Gástricas , Humanos , Piroptose , Lipossomos , Decitabina , Gasderminas , Rutênio/farmacologia , Rutênio/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Caspase 3/metabolismo
6.
J Med Chem ; 66(13): 8564-8579, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37321208

RESUMO

To obtain next-generation metal drugs that can overcome the deficiencies of platinum (Pt) drugs and treat cancer more effectively, we proposed to develop a multitargeted palladium (Pd) agent to the tumor microenvironment (TME) based on the specific residue(s) of human serum albumin (HSA). To this end, we optimized a series of Pd(II) 2-benzoylpyridine thiosemicarbazone compounds to obtain a Pd agent (5b) with significant cytotoxicity. The HSA-5b complex structure revealed that 5b bound to the hydrophobic cavity in the HSA IIA subdomain and then His-242 replaced a leaving group (Cl) of 5b, coordinating with the Pd center. The in vivo results showed that the 5b/HSA-5b complex had significant capacity of inhibiting tumor growth, and HSA optimized the therapeutic behavior of 5b. In addition, we confirmed that the 5b/HSA-5b complex inhibited tumor growth through multiple actions on different components of TME: killing cancer cells, inhibiting tumor angiogenesis, and activating T cells.


Assuntos
Antineoplásicos , Neoplasias , Pró-Fármacos , Humanos , Albumina Sérica Humana/química , Paládio , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Pró-Fármacos/farmacologia , Neoplasias/tratamento farmacológico , Ligação Proteica , Microambiente Tumoral
7.
J Med Chem ; 66(11): 7268-7279, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37218052

RESUMO

To integrate targeted diagnosis and treatment of cancer, we proposed to develop a gadolinium (Gd) agent based on the properties of apoferritin (AFt). To this end, we not only optimized a series of Gd(III) 8-hydroxyquinoline-2-carboxaldehyde-thiosemicarbazone compounds to obtain a Gd(III) compound (C4) with remarkable T1-weighted magnetic resonance imaging (MRI) performance and cytotoxicity to cancer cells in vitro but also constructed an AFt-C4 nanoparticle (NP) delivery system. Importantly, AFt-C4 NPs improved the targeting ability of C4 in vivo and showed enhanced MRI performance and tumor growth inhibition ratio relative to C4 alone. Furthermore, we confirmed that C4 and AFt-C4 NPs inhibited tumor growth through apoptosis, ferroptosis, and ferroptosis-induced immune response.


Assuntos
Nanopartículas , Neoplasias , Humanos , Gadolínio , Apoferritinas , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Imageamento por Ressonância Magnética/métodos , Meios de Contraste/farmacologia
8.
J Med Chem ; 66(8): 5669-5684, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37071741

RESUMO

To develop the next-generation Pt drug with remarkable activity and low toxicity to maximally inhibit tumor growth, we optimized a Pt(II) thiosemicarbazone compound (C4) with remarkable cytotoxicity to SK-N-MC cells and then constructed a new human serum albumin-C4 (HSA-C4) complex delivery system. The in vivo results showed that C4 and the HSA-C4 complex have remarkable therapeutic efficiency and almost no toxicity; they induced apoptosis and inhibited tumor angiogenesis. This system showed potential as a practical Pt drug. This study could pave the way for developing next-generation dual-targeted Pt drugs and achieving their targeting therapy for cancer.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Albumina Sérica Humana , Platina , Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Neoplasias/tratamento farmacológico , Linhagem Celular Tumoral
9.
J Med Chem ; 65(7): 5392-5406, 2022 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-35324188

RESUMO

To effectively integrate diagnosis and therapy for tumors, we proposed to develop an indium (In) agent based on the unique property of human serum albumin (HSA) nanoparticles (NPs). A novel In(III) quinoline-2-formaldehyde thiosemicarbazone compound (C5) was optimized with remarkable cytotoxicity and fluorescence to cancer cells in vitro. An HSA-C5 complex NP delivery system was then successfully constructed. Importantly, the HSA-C5 complex NPs have stronger bioimaging and therapeutic efficiency relative to C5 alone in vivo. Besides, the results of gene chip analysis revealed that C5/HSA-C5 complex NPs act on cancer cells through multiple mechanisms: inducing autophagy, apoptosis, and inhibiting the PI3K-Akt signaling pathway.


Assuntos
Índio , Nanopartículas , Linhagem Celular Tumoral , Humanos , Índio/farmacologia , Fosfatidilinositol 3-Quinases , Albumina Sérica Humana/metabolismo
10.
Microsyst Nanoeng ; 8: 13, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35136652

RESUMO

Effective capture and analysis of a single circulating tumor cell (CTC) is instrumental for early diagnosis and personalized therapy of tumors. However, due to their extremely low abundance and susceptibility to interference from other cells, high-throughput isolation, enrichment, and single-cell-level functional protein analysis of CTCs within one integrated system remains a major challenge. Herein, we present an integrated multifunctional microfluidic system for highly efficient and label-free CTC isolation, CTC enrichment, and single-cell immunoblotting (ieSCI). The ieSCI-chip is a multilayer microfluidic system that combines an inertia force-based cell sorter with a membrane filter for label-free CTC separation and enrichment and a thin layer of a photoactive polyacrylamide gel with microwell arrays at the bottom of the chamber for single-cell immunoblotting. The ieSCI-chip successfully identified a subgroup of apoptosis-negative (Bax-negative) cells, which traditional bulk analysis did not detect, from cisplatin-treated cells. Furthermore, we demonstrated the clinical application of the ieSCI-chip with blood samples from breast cancer patients for personalized CTC epithelial-to-mesenchymal transition (EMT) analysis. The expression level of a tumor cell marker (EpCAM) can be directly determined in isolated CTCs at the single-cell level, and the therapeutic response to anticancer drugs can be simultaneously monitored. Therefore, the ieSCI-chip provides a promising clinical translational tool for clinical drug response monitoring and personalized regimen development.

11.
Phenomics ; 2(5): 323-335, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36939755

RESUMO

Although many methods have been developed to explore the function of cells by clustering high-dimensional (HD) single-cell omics data, the inconspicuously differential expressions of biomarkers of proteins or genes across all cells disturb the cell cluster delineation and downstream analysis. Here, we introduce a hashing-based framework to improve the delineation of cell clusters, which is based on the hypothesis that one variable with no significant differences can be decomposed into more diversely latent variables to distinguish cells. By projecting the original data into a sparse HD space, fly and densefly hashing preprocessing retain the local structure of data, and improve the cluster delineation of existing clustering methods, such as PhenoGraph. Moreover, the analyses on mass cytometry dataset show that our hashing-based framework manages to unveil new hidden heterogeneities in cell clusters. The proposed framework promotes the utilization of cell biomarkers and enriches the biological findings by introducing more latent variables. Supplementary Information: The online version contains supplementary material available at 10.1007/s43657-022-00056-z.

12.
J Med Chem ; 64(10): 6777-6791, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-34000198

RESUMO

To effectively treat gastric cancer, we innovatively attempted to develop a metal agent to integrate immunotherapy and chemotherapy by dual targeting the cellular components in the tumor microenvironment (TME) based on the specific residue of human serum albumin (HSA) nanoparticles (NPs). We synthesized a series of Au(III) α-N-heterocyclic thiosemicarbazone compounds and obtained a Au agent (5b) with remarkable cytotoxicity to gastric cancer cells; moreover, we successfully constructed a novel HSA-5b complex NP delivery system. Importantly, the in vivo results showed that 5b/HSA-5b NPs effectively inhibited gastric tumor growth and HSA-5b NPs enhanced the therapeutic efficiency, bioavailability, and targeting ability compared with those of 5b alone. Furthermore, the in vitro/in vivo results revealed that 5b/HSA-5b NPs could integrate chemotherapy and immunotherapy by synergistically attacking two different cellular components in TME at the same time, namely, polarizing the tumor-associated macrophages and inducing apoptosis of gastric cancer cells.


Assuntos
Antineoplásicos/química , Complexos de Coordenação/química , Ouro/química , Nanopartículas Metálicas/química , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cristalografia por Raios X , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imunoterapia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Albumina Sérica Humana/química , Albumina Sérica Humana/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia , Relação Estrutura-Atividade , Tiossemicarbazonas/química
13.
Adv Mater ; 33(22): e2101108, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33899289

RESUMO

Fast and accurate profiling of exogenous gene expression in host cells is crucial for studying gene function in cellular and molecular biology, but still faces the challenge of incomplete co-expression of reporter genes and target genes. Here, a single-cell transfection analysis chip (scTAC) is presented, which is based on the in situ microchip immunoblotting method, for rapid and accurate analysis of exogenous gene expression in thousands of individual host cells. scTAC not only can assign information of exogenous gene activity to specific transfected cells, but enables the acquisition of continuous protein expression even in low co-expression scenarios. It is demonstrated that scTAC can reveal the relationship of expression level between reporter genes and target genes, which is helpful for evaluating transient transfection strategy efficiency. The advantages of this method for the study of fusion protein expression and downstream protein expression in signaling pathway in rare cells are shown. Empirically, an EGFP-TSPAN8 fusion plasmid is transfected into MCF-7 breast cancer cells and the expressions of two cancer stemness biomarkers (ALDHA1 and SOX2) are analyzed. The scTAC method clearly reveals an interesting phenomenon that transfected adherent MCF-7 cells exhibit some stem cell characteristics, but they do not have stem cell appearance.


Assuntos
Ensaios de Triagem em Larga Escala , Expressão Gênica , Hidrogéis
14.
J Med Chem ; 64(9): 5485-5499, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33861929

RESUMO

Anticancer agents that present nonapoptotic cell death pathways are required for treating apoptosis-resistant pancreatic cancer. Here, we synthesized three fluorescent dithiocarbazate-copper complexes, {[CuII(L)(Cl)] 1, [CuII2(L)2(NO3)2] 2, and [CuII2CuI(L)2(Br)3] 3}, to assess their antipancreatic cancer activities. Complexes 1-3 showed significantly greater cytotoxicity toward several pancreatic cancer cell lines with better IC50 than those of the HL ligand and cisplatin. Confocal fluorescence imaging showed that complex 3 was primarily localized in the mitochondria. Primarily, compound 3 also can be applied to in vivo imaging. Further studies revealed that complex 3 kills pancreatic cancer cells by triggering multiple mechanisms, including ferroptosis. Complex 3 is the first copper complex to evoke cellular events consistent with ferroptosis in cancer cells. Finally, it significantly retarded the ASPC-1 cells' growth in a mouse xenograft model.


Assuntos
Antineoplásicos/química , Complexos de Coordenação/química , Cobre/química , Hidrazinas/química , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/metabolismo , Complexos de Coordenação/farmacologia , Complexos de Coordenação/uso terapêutico , Ferroptose/efeitos dos fármacos , Meia-Vida , Humanos , Hidrazinas/uso terapêutico , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Mitocôndrias/química , Mitocôndrias/metabolismo , Conformação Molecular , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/tratamento farmacológico , Transplante Heterólogo
15.
J Inorg Biochem ; 215: 111318, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33301985

RESUMO

On the one hand, to obtain a novel next-generation anticancer metal agent; on the other hand, to improve the targeting ability and decrease side effects of metal agent, we proposed to design active-targeting human serum albumin (HSA) nanoparticles (NPs) to achieve the end. Thus, we not only designed and synthesized two ruthenium (Ru) thiosemicarbazone compounds (C1 and C2) but also succeeded in constructing active Biotin-HSA NPs for Ru(III) compounds. Importantly, Biotin-HSA-C2 NPs not only possessed a stronger capacity for killing MCF-7 cells and inhibiting their migration versusC2 alone but also increased accumulation compared to non-malignant WI-38 cells. Additionally, C2 and Biotin-HSA-C2 NPs act against MCF-7 cells by the following potential mechanism: 1) arresting the cell cycle in the S phase by regulating cyclin and cyclin-dependent kinases; 2) inducing apoptosis by releasing cytochrome c to activate caspase-9/3; 3) inhibiting the expression of p-EGFR and regulating its neighboring cellular pathways, followed by the inactivation of PI3K/Akt and activation of p38 MAPK signaling pathways.


Assuntos
Antineoplásicos/farmacologia , Biotina/química , Nanopartículas/química , Compostos de Rutênio/química , Compostos de Rutênio/farmacologia , Albumina Sérica Humana/química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Cristalografia por Raios X/métodos , Sistemas de Liberação de Medicamentos/métodos , Receptores ErbB/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células MCF-7 , Fosfatidilinositol 3-Quinases/metabolismo , Rutênio/química , Rutênio/farmacologia , Tiossemicarbazonas/química , Tiossemicarbazonas/farmacologia
16.
J Med Chem ; 63(22): 13695-13708, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33185442

RESUMO

Effective delivery of anticancer agents across the blood-brain barrier (BBB) required innovative strategies to achieve glioma regression. To resolve this problem, we proposed to develop a metal agent that target and treat glioma based on the unique property of apoferritin (AFt) nanoparticles (NPs). Thus, we synthesized a series of Au(III) 3-(4-metyl piperidine)thiosemicarbazides compounds and analyzed their structure-activity relationships, obtaining a Au agent (C6) with remarkable cytotoxicity in glioma. Moreover, we confirmed that C6 kills glioma cells by inducing lethal autophagy and apoptosis. Importantly, our results revealed that the successfully constructed apoferritin-C6 NPs (AFt-C6 NPs) can effectively cross the BBB, inhibit glioma growth, and selectively accumulate in tumors.


Assuntos
Apoferritinas/química , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Ouro/química , Nanopartículas/química , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Apoferritinas/administração & dosagem , Apoptose/fisiologia , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Feminino , Glioma/patologia , Ouro/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/administração & dosagem
17.
Anal Chem ; 92(24): 16170-16179, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33232155

RESUMO

Circulating tumor cells (CTCs) are rare cells existing in the bloodstream with a relatively low number, which facilitate as a predictor of cancer progress. However, it is difficult to obtain highly purified intact CTCs with desired viability due to the low percentage of CTCs among blood cells. In this work, we demonstrate a novel self-amplified inertial focused (SAIF) microfluidic chip that enables size-based, high-throughput, label-free separation of CTCs from a patient's blood. The SAIF chip introduced in this study demonstrated the feasibility of an extremely narrow zigzag channel (with 40 µm channel width) connected with two expansion regions to effectively separate different-sized cells with amplified separation distance. The chip performance was optimized with different-sized polystyrene (PS) particles and blood cells spiked with three different types of cancer cells. The separation efficiencies for blood cells and spiked cancer cells are higher than 80%. Recovery rates of cancer cells were tested by spiking 1500 lung cancer cells (A549), breast cancer cells (MCF-7), and cervical cancer cells (HeLa) separately to 3 mL 0.09% saline with 3 × 106 white blood cells (WBCs). The recovery rates for larger cells (MCF-7 and HeLa) were 79.1 and 85.4%, respectively. Viabilities of the cells harvested from outlets were all higher than 97% after culturing for 24, 48, and 72 h. The SAIF chip performance was further confirmed using the real clinical patient blood samples from four lung cancer patients. Theoretical force balance analysis in physics, computational simulations, and experimental observations indicate that the SAIF chip is simple but effective, and high-throughput separation CTCs can be readily achieved without complex structures.


Assuntos
Separação Celular/instrumentação , Dispositivos Lab-On-A-Chip , Células Neoplásicas Circulantes/patologia , Actinas/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Multimerização Proteica , Estrutura Quaternária de Proteína , Fatores de Tempo
18.
Dalton Trans ; 49(47): 17207-17220, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33201167

RESUMO

The anticancer function and anticancer mechanism of indium (In) complexes still remain mysterious to date. Furthermore, it is greatly challenging to design a multi-functional metal agent that not only kills cancer cells but also inhibits their invasion and metastasis. Thus, to develop novel next-generation anticancer metal agents, we designed and synthesized a series of novel In(iii) 2,6-diacetylpyridine bis(thiosemicarbazide) complexes (C1-C4) for the first time and then investigated their structure-activity relationships with human urinary bladder cancer (T-24) cells. In particular, C4 not only showed higher cytotoxicity to cancer cells and less toxicity toward normal cells relative to cisplatin but also inhibited cell invasion and metastasis of T-24 cells. Interestingly, C4 acted against T-24 cells exhibiting multiple mechanisms: (1) arresting the S-phase of cell cycle via regulation of cytokine kinases, (2) activating the mitochondrial-mediated apoptosis, endoplasmic reticulum-stress-mediated cell death, PERK and c-Jun N-terminal kinase 1 (JNK) cell signaling pathways, and (3) inhibiting the expression of telomerase via the regulation of c-myc and h-TERT proteins. Our results suggested that C4 may be developed as a potential multi-functional and multi-targeting anticancer candidate.


Assuntos
Antineoplásicos/farmacologia , Complexos de Coordenação/farmacologia , Índio/farmacologia , Tiossemicarbazonas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/síntese química , Complexos de Coordenação/química , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Índio/química , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade , Tiossemicarbazonas/química
19.
Anal Chem ; 92(9): 6312-6320, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32208602

RESUMO

Imaging mass cytometry (IMC) is an emerging imaging technology that exploits the multiplexed analysis capabilities of the CyTOF mass cytometer to make spatially resolved measurements for tissue sections. In a comprehensive view of tissue composition and marker distribution, recent developments of IMC require highly sensitive, multiplexed assays. Approaching the sensitivity of the IMC technique, we designed a novel type of biocompatible metal-labeled aptamer nanoprobe (MAP), named 167Er-A10-3.2. The small molecular probe was synthesized by conjugating 167Er-polymeric pentetic acid (167Er-DTPA) with an RNA aptamer A10-3.2. For demonstration, 167Er-A10-3.2 was applied for observing protein spatial distribution on prostatic epithelium cell of paraffin embedded Prostatic adenocarcinoma (PaC) tissue sections by IMC technology. The 167Er-A10-3.2 capitalizes on the ability of the aptamer to specifically bind target cancer cells as well as the small size of 167Er-A10-3.2 can accommodate multiple aptamer binding antigen labeled at high density. The detection signal of 167Er-A10-3.2 probe was 3-fold higher than that of PSMA antibody probe for a targeted cell under lower temperature epitope retrieval (37 °C) of PaC tissue. Furthermore, we successfully demonstrated the simultaneously staining ability of aptamer probes in IMC analysis. The successful imaging acquisition using aptamers probes in IMC technology may offer opportunity for the diagnosis of malignancies in the future.


Assuntos
Aptâmeros de Nucleotídeos/química , Érbio/química , Citometria por Imagem/métodos , Anticorpos/química , Anticorpos/imunologia , Antígenos de Superfície/imunologia , Antígenos de Superfície/metabolismo , Aptâmeros de Nucleotídeos/metabolismo , Glutamato Carboxipeptidase II/imunologia , Glutamato Carboxipeptidase II/metabolismo , Humanos , Masculino , Antígeno Prostático Específico/imunologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/patologia
20.
Mol Pharm ; 17(4): 1405-1414, 2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-32096645

RESUMO

To cause tumor regression by acting against cancer cells and inhibiting neovascularization in the tumor microenvironment, we constructed human serum albumin (HSA)-based delivery systems of 2-acetylpyridine-4,4-dimethyl-3-thiosemicarbazone-copper(II) [Cu(Ap44mT)]Cl and paclitaxel to improve both the therapeutic efficacy and the targeting ability in vivo. X-ray crystallography and matrix-assisted laser desorption/ionization time-of-flight mass spectra confirmed that [Cu(Ap44mT)]Cl complexed with HSA, whereas paclitaxel was tethered to the HSA complex by a linker sensitive to the active matrix metalloproteinase 2 (MMP2) protein. Up to 78% of paclitaxel was released from HSA within 2 h owing to MMP2 protein cleavage. In addition, a large amount of Cu(Ap44mT) was released from HSA in a pH 4.7 buffer. In vivo results revealed the following: (1) the tumor inhibitory rates of the HSA conjugate and the two-agent combination were 72.1 and 50.7%, respectively; (2) the inhibition rate of tumor angiogenesis of the HSA conjugate (73.3%) was higher than that of the two-agent combination (52.4%); (3) the increased amount of Cu in the tumor treated with the HSA conjugate was about 2-fold that in the tumor treated with the two-agent combination. Obviously, the HSA conjugate not only possessed a stronger capacity to inhibit neovascularization and the growth of liver tumors but also improved the targeting ability compared to the combination of the two agents alone.


Assuntos
Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Albumina Sérica Humana/química , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada/métodos , Cobre/química , Sistemas de Liberação de Medicamentos/métodos , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/metabolismo , Paclitaxel/química , Paclitaxel/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...